151
|
Abstract
Carbohydrates or glycans and their conjugates are involved in a wide range of biological processes and play an important role in various diseases, including inflammation, viral and bacterial infections, and tumor progression and metastasis. Studying the biological significances of carbohydrates has been challenging due in part to their structural diversity and the limited access to complex carbohydrate-containing molecules. Conventional methods such as isothermal titration calorimetry and enzyme-linked lectin assay can be laborious and require significant amounts of time and materials. The emerging of glycan microarrays as high-throughput technology for studying carbohydrate interactions has overcome some of these challenges, and has greatly contributed to our understanding of the biological roles of carbohydrates and their glycoconjugates. In addition, glycan microarrays offer new applications in biomedical research, drug discovery and development. This chapter will focus on the biomedical applications of glycan microarrays and their potential role in drug discovery and development.
Collapse
|
152
|
Vinod V, Vijayrajratnam S, Vasudevan AK, Biswas R. The cell surface adhesins of Mycobacterium tuberculosis. Microbiol Res 2019; 232:126392. [PMID: 31841935 DOI: 10.1016/j.micres.2019.126392] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/11/2019] [Accepted: 12/07/2019] [Indexed: 02/07/2023]
Abstract
Bacterial cell surface adhesins play a major role in facilitating host colonization and subsequent establishment of infection. The surface of Mycobacterium tuberculosis, owing to the complex architecture of its cell envelope, expresses numerous adhesins with varied chemical nature, including proteins, lipids, lipoproteins, glycoproteins and glycopolymers. Studies on mycobacterial adhesins show that they bind with multifarious host receptors and extracellular matrix (ECM) components. In this review we have highlighted the adhesins that are abundantly present on the mycobacterial surface and their interactions with host receptors. M. tuberculosis interacts with various host cell surface receptors such as toll like receptors, C-type lectin receptors, scavenger receptors, and Fc and complement receptors. Apart from these, ECM components like fibronectin, collagen, elastin, laminin, fibrillin and vitronectin also provide binding sites for surface adhesins of the tubercle bacilli. M. tuberculosis adhesins include proteins with and without signal peptide sequence and transmembrane proteins. Other surface adhesin macromolecules of M. tuberculosis comprises of lipids, glycolipids and glycopolymers. The interaction between the mycobacterial adhesins and their host receptors result in adhesion of the microbe to the host cells, induction of immune response and aid in the pathogenesis of the disease. A thorough understanding of the different M. tuberculosis surface adhesins and host receptors will provide a better picture of interaction between them at molecular level. The information gained on adhesins and host receptors will prove beneficial in developing novel therapeutic strategies such as the use of anti-adhesin molecules to hinder the adhesion of bacteria to the host cells, thereby preventing establishment of infection. The surface molecules discussed in this review will also benefit in identification of new drug targets, diagnostic markers or vaccine candidates against the deadly pathogen.
Collapse
Affiliation(s)
- Vivek Vinod
- Center for Nanosciences and Molecular Medicine, Amrita School of Medical Sciences and Research Center (AIMS), Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Sukhithasri Vijayrajratnam
- Center for Nanosciences and Molecular Medicine, Amrita School of Medical Sciences and Research Center (AIMS), Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Anil Kumar Vasudevan
- Department of Microbiology, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Raja Biswas
- Center for Nanosciences and Molecular Medicine, Amrita School of Medical Sciences and Research Center (AIMS), Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India.
| |
Collapse
|
153
|
ElTahir Y, Al-Araimi A, Nair RR, Autio KJ, Tu H, Leo JC, Al-Marzooqi W, Johnson EH. Binding of Brucella protein, Bp26, to select extracellular matrix molecules. BMC Mol Cell Biol 2019; 20:55. [PMID: 31783731 PMCID: PMC6884894 DOI: 10.1186/s12860-019-0239-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 11/19/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Brucella is a facultative intracellular pathogen responsible for zoonotic disease brucellosis. Little is known about the molecular basis of Brucella adherence to host cells. In the present study, the possible role of Bp26 protein as an adhesin was explored. The ability of Brucella protein Bp26 to bind to extracellular matrix (ECM) proteins was determined by enzyme-linked immunosorbent assay (ELISA) and biolayer interferometry (BLI). RESULTS ELISA experiments showed that Bp26 bound in a dose-dependent manner to both immobilized type I collagen and vitronectin. Bp26 bound weakly to soluble fibronectin but did not bind to immobilized fibronectin. No binding to laminin was detected. Biolayer interferometry showed high binding affinity of Bp26 to immobilized type I collagen and no binding to fibronectin or laminin. Mapping of Bp26 antigenic epitopes by biotinylated overlapping peptides spanning the entire sequence of Bp26 using anti Bp26 mouse serum led to the identification of five linear epitopes. Collagen and vitronectin bound to peptides from several regions of Bp26, with many of the binding sites for the ligands overlapping. The strongest binding for anti-Bp26 mouse serum, collagen and vitronectin was to the peptides at the C-terminus of Bp26. Fibronectin did not bind to any of the peptides, although it bound to the whole Bp26 protein. CONCLUSIONS Our results highlight the possible role of Bp26 protein in the adhesion process of Brucella to host cells through ECM components. This study revealed that Bp26 binds to both immobilized and soluble type I collagen and vitronectin. It also binds to soluble but not immobilized fibronectin. However, Bp26 does not bind to laminin. These are novel findings that offer insight into understanding the interplay between Brucella and host target cells, which may aid in future identification of a new target for diagnosis and/or vaccine development and prevention of brucellosis.
Collapse
Affiliation(s)
- Yasmin ElTahir
- Department of Animal & Veterinary Sciences, Sultan Qaboos University. College of Agricultural & Marine Sciences, P.O.box 34. 123 Alkhod, Muscat, Sultanate of Oman.
| | - Amna Al-Araimi
- Department of Animal & Veterinary Sciences, Sultan Qaboos University. College of Agricultural & Marine Sciences, P.O.box 34. 123 Alkhod, Muscat, Sultanate of Oman
| | - Remya R Nair
- Department of Animal & Veterinary Sciences, Sultan Qaboos University. College of Agricultural & Marine Sciences, P.O.box 34. 123 Alkhod, Muscat, Sultanate of Oman
| | - Kaija J Autio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014, Oulu, Finland
| | - Hongmin Tu
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014, Oulu, Finland
| | - Jack C Leo
- Section for Genetics and Evolutionary Biology, Department of Biosciences, University of Oslo, 0361, Oslo, Norway.,Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham, NG1 4FQ, UK
| | - Waleed Al-Marzooqi
- Department of Animal & Veterinary Sciences, Sultan Qaboos University. College of Agricultural & Marine Sciences, P.O.box 34. 123 Alkhod, Muscat, Sultanate of Oman
| | - Eugene H Johnson
- Department of Animal & Veterinary Sciences, Sultan Qaboos University. College of Agricultural & Marine Sciences, P.O.box 34. 123 Alkhod, Muscat, Sultanate of Oman
| |
Collapse
|
154
|
Vaca DJ, Thibau A, Schütz M, Kraiczy P, Happonen L, Malmström J, Kempf VAJ. Interaction with the host: the role of fibronectin and extracellular matrix proteins in the adhesion of Gram-negative bacteria. Med Microbiol Immunol 2019; 209:277-299. [PMID: 31784893 PMCID: PMC7248048 DOI: 10.1007/s00430-019-00644-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/14/2019] [Indexed: 01/03/2023]
Abstract
The capacity of pathogenic microorganisms to adhere to host cells and avoid clearance by the host immune system is the initial and most decisive step leading to infections. Bacteria have developed different strategies to attach to diverse host surface structures. One important strategy is the adhesion to extracellular matrix (ECM) proteins (e.g., collagen, fibronectin, laminin) that are highly abundant in connective tissue and basement membranes. Gram-negative bacteria express variable outer membrane proteins (adhesins) to attach to the host and to initiate the process of infection. Understanding the underlying molecular mechanisms of bacterial adhesion is a prerequisite for targeting this interaction by “anti-ligands” to prevent colonization or infection of the host. Future development of such “anti-ligands” (specifically interfering with bacteria-host matrix interactions) might result in the development of a new class of anti-infective drugs for the therapy of infections caused by multidrug-resistant Gram-negative bacteria. This review summarizes our current knowledge about the manifold interactions of adhesins expressed by Gram-negative bacteria with ECM proteins and the use of this information for the generation of novel therapeutic antivirulence strategies.
Collapse
Affiliation(s)
- Diana J Vaca
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Paul-Ehrlich-Str. 40, 60596, Frankfurt, Germany
| | - Arno Thibau
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Paul-Ehrlich-Str. 40, 60596, Frankfurt, Germany
| | - Monika Schütz
- Institute for Medical Microbiology and Infection Control, University Hospital, Eberhard Karls-University, Tübingen, Germany
| | - Peter Kraiczy
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Paul-Ehrlich-Str. 40, 60596, Frankfurt, Germany
| | - Lotta Happonen
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Volkhard A J Kempf
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Paul-Ehrlich-Str. 40, 60596, Frankfurt, Germany.
| |
Collapse
|
155
|
Monoclonal antibody against l-lectin module of SraP blocks adhesion and protects mice against Staphylococcus aureus challenge. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 54:420-428. [PMID: 31706823 DOI: 10.1016/j.jmii.2019.08.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/14/2019] [Accepted: 08/20/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND/PURPOSE SraP is a serine-rich repeat protein (SRRP) from Staphylococcus aureus that binds to sialylated receptors to promote bacterial adhesion to and invasion into host epithelial cells, mediated by the l-lectin module of its ligand-binding region. METHODS The sequence encoding the L-lectin module of SraP was inserted into pET28a plasmid, and the recombinant protein was purified by His label affinity chromatography. A monoclonal antibody (mAb) against the l-lectin module was obtained and confirmed by enzyme-linked immunosorbent assay and western blotting. The effect of the mAb on S. aureus adhesion and invasion was assessed in A549 cells and mice subjected to S. aureus challenge. RESULTS We successfully obtained a mAb against the l-lectin module of SraP. Pre-incubation with the mAb dramatically inhibited the bacteria's ability to adhere to and invade A549 cells. Moreover, mice administered mAb through tail vein injection had significantly fewer bacteria in the blood. CONCLUSION The anti-SraPL-Lectin mAb significantly reduced the adherence and invasion of S. aureus to host cells. This study lays the foundation for the future development of the l-lectin module of SraP as a target for the prevention and treatment of S. aureus infection. Our findings suggest that specific subdomains of SRRPs may represent potential antibacterial drug targets for intervention.
Collapse
|
156
|
Leonard AC, Petrie LE, Cox G. Bacterial Anti-adhesives: Inhibition of Staphylococcus aureus Nasal Colonization. ACS Infect Dis 2019; 5:1668-1681. [PMID: 31374164 DOI: 10.1021/acsinfecdis.9b00193] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Bacterial adhesion to the skin and mucosa is often a fundamental and early step in host colonization, the establishment of bacterial infections, and pathology. This process is facilitated by adhesins on the surface of the bacterial cell that recognize host cell molecules. Interfering with bacterial host cell adhesion, so-called anti-adhesive therapeutics, offers promise for the development of novel approaches to control bacterial infections. In this review, we focus on the discovery of anti-adhesives targeting the high priority pathogen Staphylococcus aureus. This organism remains a major clinical burden, and S. aureus nasal colonization is associated with poor clinical outcomes. We describe the molecular basis of nasal colonization and highlight potentially efficacious targets for the development of novel nasal decolonization strategies.
Collapse
Affiliation(s)
- Allison C. Leonard
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| | - Laurenne E. Petrie
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| | - Georgina Cox
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
157
|
Engin AB, Engin A. Nanoantibiotics: A Novel Rational Approach to Antibiotic Resistant Infections. Curr Drug Metab 2019; 20:720-741. [DOI: 10.2174/1389200220666190806142835] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 01/09/2023]
Abstract
Background:The main drawbacks for using conventional antimicrobial agents are the development of multiple drug resistance due to the use of high concentrations of antibiotics for extended periods. This vicious cycle often generates complications of persistent infections, and intolerable antibiotic toxicity. The problem is that while all new discovered antimicrobials are effective and promising, they remain as only short-term solutions to the overall challenge of drug-resistant bacteria.Objective:Recently, nanoantibiotics (nAbts) have been of tremendous interest in overcoming the drug resistance developed by several pathogenic microorganisms against most of the commonly used antibiotics. Compared with free antibiotic at the same concentration, drug delivered via a nanoparticle carrier has a much more prominent inhibitory effect on bacterial growth, and drug toxicity, along with prolonged drug release. Additionally, multiple drugs or antimicrobials can be packaged within the same smart polymer which can be designed with stimuli-responsive linkers. These stimuli-responsive nAbts open up the possibility of creating multipurpose and targeted antimicrobials. Biofilm formation still remains the leading cause of conventional antibiotic treatment failure. In contrast to conventional antibiotics nAbts easily penetrate into the biofilm, and selectively target biofilm matrix constituents through the introduction of bacteria specific ligands. In this context, various nanoparticles can be stabilized and functionalized with conventional antibiotics. These composites have a largely enhanced bactericidal efficiency compared to the free antibiotic.Conclusion:Nanoparticle-based carriers deliver antibiotics with better biofilm penetration and lower toxicity, thus combating bacterial resistance. However, the successful adaptation of nanoformulations to clinical practice involves a detailed assessment of their safety profiles and potential immunotoxicity.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Ankara, Turkey
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Ankara, Turkey
| |
Collapse
|
158
|
Psonis JJ, Chahales P, Henderson NS, Rigel NW, Hoffman PS, Thanassi DG. The small molecule nitazoxanide selectively disrupts BAM-mediated folding of the outer membrane usher protein. J Biol Chem 2019; 294:14357-14369. [PMID: 31391254 PMCID: PMC6768635 DOI: 10.1074/jbc.ra119.009616] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/30/2019] [Indexed: 12/11/2022] Open
Abstract
Bacterial pathogens assemble adhesive surface structures termed pili or fimbriae to initiate and sustain infection of host tissues. Uropathogenic Escherichia coli, the primary causative agent of urinary tract infections, expresses type 1 and P pili required for colonization of the bladder and kidney, respectively. These pili are assembled by the conserved chaperone-usher (CU) pathway, in which a periplasmic chaperone works together with an outer membrane (OM) usher protein to build and secrete the pilus fiber. Previously, we found that the small molecule and antiparasitic drug nitazoxanide (NTZ) inhibits CU pathway-mediated pilus biogenesis in E. coli by specifically interfering with proper maturation of the usher protein in the OM. The usher is folded and inserted into the OM by the β-barrel assembly machine (BAM) complex, which in E. coli comprises five proteins, BamA-E. Here, we show that sensitivity of the usher to NTZ is modulated by BAM expression levels and requires the BamB and BamE lipoproteins. Furthermore, a genetic screen for NTZ-resistant bacterial mutants isolated a mutation in the essential BamD lipoprotein. These findings suggest that NTZ selectively interferes with an usher-specific arm of the BAM complex, revealing new details of the usher folding pathway and BAM complex function. Evaluation of a set of NTZ derivatives identified compounds with increased potency and disclosed that NTZ's nitrothiazole ring is critical for usher inhibition. In summary, our findings indicate highly specific effects of NTZ on the usher folding pathway and have uncovered NTZ analogs that specifically decrease usher levels in the OM.
Collapse
Affiliation(s)
- John J Psonis
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| | - Peter Chahales
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| | - Nadine S Henderson
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| | - Nathan W Rigel
- Department of Biology, Hofstra University, Hempstead, New York 11549
| | - Paul S Hoffman
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia 22908
| | - David G Thanassi
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York 11794
| |
Collapse
|
159
|
Wang S, Yang D, Wu X, Wang Y, Wang D, Tian M, Li T, Qi J, Wang X, Ding C, Yu S. Autotransporter MisL of Salmonella enterica serotype Typhimurium facilitates bacterial aggregation and biofilm formation. FEMS Microbiol Lett 2019; 365:5036521. [PMID: 29901711 DOI: 10.1093/femsle/fny142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/11/2018] [Indexed: 01/04/2023] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is an important food-borne zoonotic pathogen that causes increased morbidity and mortality worldwide. The autotransporter (AT) proteins are a large and diverse family of extracellular proteins, many of which contribute to the pathogenicity of Gram-negative bacteria. The S. Typhimurium AT protein MisL mediates intestinal colonization in mice. Bioinformatics analyses indicated that MisL clusters with ATs are involved in bacterial biofilm formation, aggregation and adherence. In this study, we found that the misL overexpression increased S. Typhimurium biofilm formation. In addition, the misL deletion reduced bacterial adherence and invasion abilities on HeLa cells, but did not affect the bacterial virulence. Similarly, MisL expression in Escherichia coli strain promoted bacterial biofilm formation as well as adhesion and invasion capacities. However, the misL overexpression had no influence on the bacterial aggregation except for AAEC189Δflu, a strain lacking type I fimbriae. Moreover, we demonstrated that immunization with recombinant MisL protein stimulated the production of high IgG antibody titers, which conferred modest protection against S. Typhimurium infection. This study illustrates the novel biological functions and immunoprotective effects of MisL in S. Typhimurium.
Collapse
Affiliation(s)
- Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Denghui Yang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xiaojun Wu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Dong Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Mingxing Tian
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Tao Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jingjing Qi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xiaolan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Shengqing Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| |
Collapse
|
160
|
Hussain S, Lv F, Qi R, Senthilkumar T, Zhao H, Chen Y, Liu L, Wang S. Förster Resonance Energy Transfer Mediated Rapid and Synergistic Discrimination of Bacteria over Fungi Using a Cationic Conjugated Glycopolymer. ACS APPLIED BIO MATERIALS 2019; 3:20-28. [DOI: 10.1021/acsabm.9b00691] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sameer Hussain
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Ruilian Qi
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Thangaraj Senthilkumar
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Hao Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yanyan Chen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Libing Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
161
|
Zhou L, Chen C, Xie J, Xu C, Zhao Q, Qin JG, Chen L, Li E. Intestinal bacterial signatures of the "cotton shrimp-like" disease explain the change of growth performance and immune responses in Pacific white shrimp (Litopenaeus vannamei). FISH & SHELLFISH IMMUNOLOGY 2019; 92:629-636. [PMID: 31265910 DOI: 10.1016/j.fsi.2019.06.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/24/2019] [Accepted: 06/28/2019] [Indexed: 06/09/2023]
Abstract
Imbalance of intestinal microbiota has been recognized in aquatic animals infected with various diseases. However, the signature of intestinal bacteria of the "cotton shrimp-like" disease in the Pacific white shrimp Litopenaeus vannamei remains unknown. This study investigates the composition, diversity, microbial-mediated function and interspecies interaction of intestinal microbiota on shrimp with different health status using 16S rRNA gene high-throughput sequencing. Meanwhile, the growth performance and the mRNA expression of innate immune gene in hepatopancreas were also investigated. The growth performance and the mRNA expression of innate immune genes (e.g., crustin, toll, and immune deficiency genes) in the hepatopancreas were significantly decreased in diseased shrimp compared with healthy shrimp. Bacteria of the family Rickettsiaceae and genus Tenacibaculum were exclusively enriched and significantly increased in diseased shrimp, respectively, whereas, the Actinobacteria class dramatically deceased. The diseased shrimp exhibited higher ACE and Chao1 indices and lower complexity of intestinal interspecies interaction than healthy shrimp. Microbial-mediated functions predicted by PICRUSt showed that 83% KEGG pathway including nutrient absorption and digestion significantly increased in diseased shrimp. This study provides an overview on the interplay among the "cotton shrimp-like" disease, intestinal microbiota, growth performance and host immune responses from an ecological perspective.
Collapse
Affiliation(s)
- Li Zhou
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, Hainan, 570228, China; Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, 570228, Hainan, China
| | - Chengzhuang Chen
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, 570228, Hainan, China
| | - Jia Xie
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, 570228, Hainan, China
| | - Chang Xu
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, 570228, Hainan, China
| | - Qun Zhao
- Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, 570228, Hainan, China
| | - Jian G Qin
- School of Biological Sciences, Flinders University, Adelaide, SA, 5001, Australia
| | - Liqiao Chen
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Erchao Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, Hainan, 570228, China; Department of Aquaculture, College of Marine Sciences, Hainan University, Haikou, 570228, Hainan, China.
| |
Collapse
|
162
|
Arciola CR, Campoccia D, Montanaro L. Implant infections: adhesion, biofilm formation and immune evasion. Nat Rev Microbiol 2019; 16:397-409. [PMID: 29720707 DOI: 10.1038/s41579-018-0019-y] [Citation(s) in RCA: 1091] [Impact Index Per Article: 218.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Medical device-associated infections account for a large proportion of hospital-acquired infections. A variety of opportunistic pathogens can cause implant infections, depending on the type of the implant and on the anatomical site of implantation. The success of these versatile pathogens depends on rapid adhesion to virtually all biomaterial surfaces and survival in the hostile host environment. Biofilm formation on implant surfaces shelters the bacteria and encourages persistence of infection. Furthermore, implant-infecting bacteria can elude innate and adaptive host defences as well as biocides and antibiotic chemotherapies. In this Review, we explore the fundamental pathogenic mechanisms underlying implant infections, highlighting orthopaedic implants and Staphylococcus aureus as a prime example, and discuss innovative targets for preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy. .,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Davide Campoccia
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Lucio Montanaro
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
163
|
De Lerma Barbaro A, Gariboldi MB, Mastore M, Brivio MF, Giovannardi S. In Vivo Effects of A Pro-PO System Inhibitor on the Phagocytosis of Xenorhabdus Nematophila in Galleria Mellonella Larvae. INSECTS 2019; 10:E263. [PMID: 31443446 PMCID: PMC6780223 DOI: 10.3390/insects10090263] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/03/2019] [Accepted: 08/16/2019] [Indexed: 12/21/2022]
Abstract
Xenorhabdus nematophila is a Gram-negative bacterium symbiont of the entomopathogen nematode Steinernema carpocapsae whose immunosuppressive properties over host's immune response have been thoroughly investigated. In particular, live X. nematophila actively impairs phagocytosis in host's hemocytes through the secretion of inhibitors of eicosanoids synthesis. In this article we have investigated the cell surface structural features of X. nematophila responsible for the elusion from phagocytosis. To this end we have studied the uptake of heat-killed (hk), fluorescein isothiocyanate (FITC)-labeled X. nematophila by phagocytes from both a host insect and a mammalian species. In vitro dead X. nematophila passively resists engulfment by insect hemocytes without impairing the phagocytosis machinery whereas, unexpectedly, in vivo a significant phagocytosis of dead X. nematophila was observed. X. nematophila in vivo phagocytosis was increased by the co-injection of the specific inhibitor of pro-phenoloxidase (PO) system phenylthiourea (PTU), even if these effects were not observed in in vitro tests. Furthermore, biochemical modifications of X. nematophila cell wall implement in vivo phagocytosis, suggesting that this bacterium avoid phagocytosis because the ligand of phagocytic receptors is somehow buried or disguised in the cell wall. Finally, dead X. nematophila escapes engulfment even by human phagocytes suggesting that X. nematophila could be a useful model to investigate escape from phagocytosis by mammalian macrophages.
Collapse
Affiliation(s)
- Andrea De Lerma Barbaro
- Laboratory of Comparative Physiopathology, Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (Varese), Italy
| | - Marzia B Gariboldi
- Laboratory of Anticancer Pharmacology, Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (Varese), Italy
| | - Maristella Mastore
- Laboratory of Comparative Immunology and Parasitology, Department of Theoretical and Applied Sciences, University of Insubria, 21100 Varese, Italy
| | - Maurizio F Brivio
- Laboratory of Comparative Immunology and Parasitology, Department of Theoretical and Applied Sciences, University of Insubria, 21100 Varese, Italy
| | - Stefano Giovannardi
- Laboratory of Comparative Physiopathology, Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (Varese), Italy.
| |
Collapse
|
164
|
Rose R, Häuser S, Stump-Guthier C, Weiss C, Rohde M, Kim KS, Ishikawa H, Schroten H, Schwerk C, Adam R. Virulence factor-dependent basolateral invasion of choroid plexus epithelial cells by pathogenic Escherichia coli in vitro. FEMS Microbiol Lett 2019; 365:5195518. [PMID: 30476042 DOI: 10.1093/femsle/fny274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/20/2018] [Indexed: 12/18/2022] Open
Abstract
Escherichia coli is the most common Gram-negative causative agent of neonatal meningitis and E. coli meningitis is associated with high morbidity and mortality. Previous research has been carried out with regard to the blood-brain barrier and thereby unveiled an assortment of virulence factors involved in E. coli meningitis. Little, however, is known about the role of the blood-cerebrospinal fluid (CSF) barrier (BCSFB), in spite of several studies suggesting that the choroid plexus (CP) is a possible entry point for E. coli into the CSF spaces. Here, we used a human CP papilloma (HIBCPP) cell line that was previously established as valid model for the study of the BCSFB. We show that E. coli invades HIBCPP cells in a polar fashion preferentially from the physiologically relevant basolateral side. Moreover, we demonstrate that deletion of outer membrane protein A, ibeA or neuDB genes results in decreased cell infection, while absence of fimH enhances invasion, although causing reduced adhesion to the apical side of HIBCPP cells. Our findings suggest that the BCSFB might constitute an entry point for E. coli into the central nervous system, and HIBCPP cells are a valuable tool for investigating E. coli entry of the BCSFB.
Collapse
Affiliation(s)
- Rebekah Rose
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| | - Svenja Häuser
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| | - Carolin Stump-Guthier
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, D-38124 Braunschweig, Germany
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, 1-1-1Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| | - Rüdiger Adam
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| |
Collapse
|
165
|
Bravo M, Combes T, Martinez FO, Cerrato R, Rey J, Garcia-Jimenez W, Fernandez-Llario P, Risco D, Gutierrez-Merino J. Lactobacilli Isolated From Wild Boar ( Sus scrofa) Antagonize Mycobacterium bovis Bacille Calmette-Guerin (BCG) in a Species-Dependent Manner. Front Microbiol 2019; 10:1663. [PMID: 31417502 PMCID: PMC6683848 DOI: 10.3389/fmicb.2019.01663] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 07/04/2019] [Indexed: 01/06/2023] Open
Abstract
Background: Wildlife poses a significant burden for the complete eradication of bovine tuberculosis (bTB). In particular, wild boar (Sus scrofa) is one of the most important reservoirs of Mycobacterium bovis, the causal agent of bTB. Wild boar can display from mild TB lesions, usually found in head lymph nodes, to generalized TB lesions distributed in different anatomical regions; but rarely clinical signs, which complicates the diagnosis of Mycobacterium bovis infection and bTB control. Among the possibilities for this variability in lesion distribution is the influence of the host-beneficial commensal-primed immune barrier. In this respect, beneficial microbes may delay bTB dissemination as a consequence of an antagonistic competition for nutrients and phagocytes. In order to explore this possibility, we have tested whether typical commensals such as lactobacilli have the capacity to reduce the survival rate of the surrogate M. bovis strain Bacillus Calmette-Guerin (BCG); and to modulate its phagocyte intake. Results: Three Lactobacillus species, L. casei, L. plantarum, and L. salivarius, isolated from wild boar feces displayed a pH-dependent inhibitory activity against BCG and influenced its intake by porcine blood phagocytes in a species-dependent manner. All lactobacilli showed a very significant bactericidal effect against BCG at low pH, but only isolates of L. plantarum and L. casei displayed such antimycobacterial activity at neutral pH. The genomes of these isolates revealed the presence of two-peptide bacteriocins whose precursor genes up-regulate in the presence of BCG cells. Furthermore, L. plantarum reduced significantly the BCG phagocytic intake, whereas L. casei had the opposite effect. L. salivarius had no significant influence on the phagocytic response to BCG. Conclusions: Our in vitro results show that lactobacilli isolated from wild boar antagonize BCG as a consequence of their antimycobacterial activity and a competitive phagocytic response. These findings suggest that commensal bacteria could play a beneficial role in influencing the outcome of bTB dissemination. Further work with lactobacilli as a potential competitive pressure to control bTB will need to take into account the complex nature of the commensal microbiome, the specific immunity of the wild boar and the in vivo infection context with pathogenic strains of M. bovis.
Collapse
Affiliation(s)
- Maria Bravo
- Innovación en Gestión y Conservación de Ungulados SL, Cáceres, Spain.,Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de Extremadura, Cáceres, Spain
| | - Theo Combes
- Department of Biochemical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Fernando O Martinez
- Department of Biochemical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Rosario Cerrato
- Innovación en Gestión y Conservación de Ungulados SL, Cáceres, Spain
| | - Joaquín Rey
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de Extremadura, Cáceres, Spain
| | | | | | - David Risco
- Innovación en Gestión y Conservación de Ungulados SL, Cáceres, Spain
| | - Jorge Gutierrez-Merino
- Department of Nutritional Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
166
|
Liao C, Fang K, Xiao J, Zhang W, Zhang B, Yuan W, Lu W, Xu D. Critical determinants of human neutrophil peptide 1 for enhancing host epithelial adhesion of Shigella flexneri. Cell Microbiol 2019; 21:e13069. [PMID: 31218775 DOI: 10.1111/cmi.13069] [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: 01/08/2019] [Revised: 05/17/2019] [Accepted: 06/04/2019] [Indexed: 11/27/2022]
Abstract
Human neutrophil peptides (HNPs), also known as human myeloid α-defensins degranulated by infiltrating neutrophils at bacterial infection loci, exhibit broad antomicrobial activities against bacteria, fungi, and viruses. We have made a surprising recent finding that Shigella, a highly contagious, yet poorly adhesive enteric pathogen, exploits human α-defensins including HNP1 to enhance its adhesion to and invasion of host epithelial cells. However, the critical molecular determinants responsible for HNP1-enhanced Shigella adhesion and invasion have yet to be investigated. Using cultured epithelial cells and polarised Caco2 cells as an in vitro infection model, we demonstrated that HNP1 promoted Shigella infection in a structure- and sequence-dependent manner, with two bulky hydrophobic residues, Trp26 and Phe28 important for HNP1 self-assembly, being most critical. The functional importance of hydrophobicity for HNP1-enhanced Shigella infection was further verified by substitutions for Trp26 of a series of unnatural amino acids with straight aliphatic side chains of different lengths. Dissection of the Shigella infection process revealed that bacteria-rather than host cells-bound HNP1 contributed most to the enhancement. Further, mutagenesis analysis of bacterial surface components, while precluding the involvement of lipopolysaccharides (LPS) in the interaction with HNP1, identified outer membrane proteins and the Type 3 secretion apparatus as putative binding targets of HNP1 involved in enhanced Shigella adhesion and invasion. Our findings provide molecular and mechanistic insights into the mode of action of HNP1 in promoting Shigella infection, thus showcasing another example of how innate immune factors may serve as a double-edged sword in health and disease.
Collapse
Affiliation(s)
- Chongbing Liao
- Center for Translational Medicine Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kun Fang
- Department of Internal Medicine, Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiu Xiao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Wei Zhang
- Center for Translational Medicine Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Bing Zhang
- Center for Translational Medicine Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Weirong Yuan
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dan Xu
- Center for Translational Medicine Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
167
|
Deng Z, Liu F, Li C. Therapeutic effect of ethylenediaminetetraacetic acid irrigation solution against wound infection with drug-resistant bacteria in a rat model: an animal study. Bone Joint Res 2019; 8:189-198. [PMID: 31214331 PMCID: PMC6548975 DOI: 10.1302/2046-3758.85.bjr-2018-0280.r3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Objectives Irrigation is the cornerstone of treating skeletal infection by eliminating pathogens in wounds. A previous study shows that irrigation with normal saline (0.9%) and ethylenediaminetetraacetic acid (EDTA) could improve the removal of Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) compared with normal saline (NS) alone. However, it is still unclear whether EDTA solution is effective against infection with drug-resistant bacteria. Methods We established three wound infection models (skin defect, bone-exposed, implant-exposed) by inoculating the wounds with a variety of representative drug-resistant bacteria including methicillin-resistant S. aureus (MRSA), extended spectrum beta-lactamase-producing E. coli (ESBL-EC), multidrug-resistant Pseudomonas aeruginosa (MRPA), vancomycin-resistant Enterococcus (VRE), multidrug-resistant Acinetobacter baumannii (MRAB), multidrug-resistant Enterobacter (MRE), and multidrug-resistant Proteus mirabilis (MRPM). Irrigation and debridement were repeated until the wound culture became negative. The operating times required to eliminate pathogens in wounds were compared through survival analysis. Results Compared with other groups (NS, castile soap, benzalkonium chloride, and bacitracin), the EDTA group required fewer debridement and irrigation operations to achieve pathogen eradication in all three models of wound infection. Conclusion Irrigation with EDTA solution was more effective than the other irrigation fluids used in the treatment of wound infections caused by drug-resistant pathogens. Cite this article: Z. Deng, F. Liu, C. Li. Therapeutic effect of ethylenediaminetetraacetic acid irrigation solution against wound infection with drug-resistant bacteria in a rat model: an animal study. Bone Joint Res 2019;8:189–198. DOI: 10.1302/2046-3758.85.BJR-2018-0280.R3.
Collapse
Affiliation(s)
- Z Deng
- Department of Orthopaedics, Beijing Changping Hospital, Beijing, China
| | - F Liu
- Department of Nursing, Weihai Municipal Hospital, Weihai, China
| | - C Li
- Department of Orthopaedics, Beijing Changping Hospital, Beijing, China
| |
Collapse
|
168
|
Dong H, Terrell JL, Jahnke JP, Zu TNK, Hurley MM, Stratis-Cullum DN. Biofunctionalized Cellulose Nanofibrils Capable of Capture and Antiadhesion of Fimbriated Escherichia coli. ACS APPLIED BIO MATERIALS 2019; 2:2937-2945. [DOI: 10.1021/acsabm.9b00295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hong Dong
- Biotechnology Branch, CCDC Army Research Laboratory, 2800 Powder Mill Road, Adelphi, Maryland 20783, United States
| | - Jessica L. Terrell
- Biotechnology Branch, CCDC Army Research Laboratory, 2800 Powder Mill Road, Adelphi, Maryland 20783, United States
| | - Justin P. Jahnke
- Biotechnology Branch, CCDC Army Research Laboratory, 2800 Powder Mill Road, Adelphi, Maryland 20783, United States
| | - Theresah N. K. Zu
- Biotechnology Branch, CCDC Army Research Laboratory, 2800 Powder Mill Road, Adelphi, Maryland 20783, United States
| | - Margaret M. Hurley
- Biotechnology Branch, CCDC Army Research Laboratory, 2800 Powder Mill Road, Adelphi, Maryland 20783, United States
| | - Dimitra N. Stratis-Cullum
- Biotechnology Branch, CCDC Army Research Laboratory, 2800 Powder Mill Road, Adelphi, Maryland 20783, United States
| |
Collapse
|
169
|
Catron TR, Swank A, Wehmas LC, Phelps D, Keely SP, Brinkman NE, McCord J, Singh R, Sobus J, Wood CE, Strynar M, Wheaton E, Tal T. Microbiota alter metabolism and mediate neurodevelopmental toxicity of 17β-estradiol. Sci Rep 2019; 9:7064. [PMID: 31068624 PMCID: PMC6506524 DOI: 10.1038/s41598-019-43346-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/18/2019] [Indexed: 02/07/2023] Open
Abstract
Estrogenic chemicals are widespread environmental contaminants associated with diverse health and ecological effects. During early vertebrate development, estrogen receptor signaling is critical for many different physiologic responses, including nervous system function. Recently, host-associated microbiota have been shown to influence neurodevelopment. Here, we hypothesized that microbiota may biotransform exogenous 17-βestradiol (E2) and modify E2 effects on swimming behavior. Colonized zebrafish were continuously exposed to non-teratogenic E2 concentrations from 1 to 10 days post-fertilization (dpf). Changes in microbial composition and predicted metagenomic function were evaluated. Locomotor activity was assessed in colonized and axenic (microbe-free) zebrafish exposed to E2 using a standard light/dark behavioral assay. Zebrafish tissue was collected for chemistry analyses. While E2 exposure did not alter microbial composition or putative function, colonized E2-exposed larvae showed reduced locomotor activity in the light, in contrast to axenic E2-exposed larvae, which exhibited normal behavior. Measured E2 concentrations were significantly higher in axenic relative to colonized zebrafish. Integrated peak area for putative sulfonated and glucuronidated E2 metabolites showed a similar trend. These data demonstrate that E2 locomotor effects in the light phase are dependent on the presence of microbiota and suggest that microbiota influence chemical E2 toxicokinetics. More broadly, this work supports the concept that microbial colonization status may influence chemical toxicity.
Collapse
Affiliation(s)
- Tara R Catron
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | | | | | - Drake Phelps
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | | | | | - James McCord
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Randolph Singh
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Jon Sobus
- U.S. EPA/ORD/NERL/EMMD, RTP, NC, USA
| | - Charles E Wood
- U.S. EPA/ORD/NHEERL/ISTD, RTP, NC, USA
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | | | | | | |
Collapse
|
170
|
Guo S, Vance TD, Stevens CA, Voets I, Davies PL. RTX Adhesins are Key Bacterial Surface Megaproteins in the Formation of Biofilms. Trends Microbiol 2019; 27:453-467. [DOI: 10.1016/j.tim.2018.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/13/2018] [Accepted: 12/12/2018] [Indexed: 12/20/2022]
|
171
|
Paxman JJ, Lo AW, Sullivan MJ, Panjikar S, Kuiper M, Whitten AE, Wang G, Luan CH, Moriel DG, Tan L, Peters KM, Phan MD, Gee CL, Ulett GC, Schembri MA, Heras B. Unique structural features of a bacterial autotransporter adhesin suggest mechanisms for interaction with host macromolecules. Nat Commun 2019; 10:1967. [PMID: 31036849 PMCID: PMC6488583 DOI: 10.1038/s41467-019-09814-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/28/2019] [Indexed: 12/31/2022] Open
Abstract
Autotransporters are the largest family of outer membrane and secreted proteins in Gram-negative bacteria. Most autotransporters are localised to the bacterial surface where they promote colonisation of host epithelial surfaces. Here we present the crystal structure of UpaB, an autotransporter that is known to contribute to uropathogenic E. coli (UPEC) colonisation of the urinary tract. We provide evidence that UpaB can interact with glycosaminoglycans and host fibronectin. Unique modifications to its core β-helical structure create a groove on one side of the protein for interaction with glycosaminoglycans, while the opposite face can bind fibronectin. Our findings reveal far greater diversity in the autotransporter β-helix than previously thought, and suggest that this domain can interact with host macromolecules. The relevance of these interactions during infection remains unclear.
Collapse
Affiliation(s)
- Jason J Paxman
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, VIC, Australia
| | - Alvin W Lo
- School of Chemistry and Molecular Biosciences, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Matthew J Sullivan
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Gold Coast, 4222, QLD, Australia
| | - Santosh Panjikar
- Macromolecular Crystallography, Australian Synchrotron, Clayton, 3168, VIC, Australia
- Department of Molecular Biology and Biochemistry, Monash University, Melbourne, 3800, VIC, Australia
| | - Michael Kuiper
- Molecular & Materials Modelling group Data61, CSIRO, Docklands, Melbourne, 8012, VIC, Australia
| | - Andrew E Whitten
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation, Lucas Heights, 2234, NSW, Australia
| | - Geqing Wang
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, VIC, Australia
| | - Chi-Hao Luan
- High Throughput Analysis Laboratory and Department of Molecular Biosciences, Northwestern University, Chicago, 60208, IL, USA
| | - Danilo G Moriel
- School of Chemistry and Molecular Biosciences, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Lendl Tan
- School of Chemistry and Molecular Biosciences, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Kate M Peters
- School of Chemistry and Molecular Biosciences, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Minh-Duy Phan
- School of Chemistry and Molecular Biosciences, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Christine L Gee
- Macromolecular Crystallography, Australian Synchrotron, Clayton, 3168, VIC, Australia
| | - Glen C Ulett
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Gold Coast, 4222, QLD, Australia
| | - Mark A Schembri
- School of Chemistry and Molecular Biosciences, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, 4072, QLD, Australia.
| | - Begoña Heras
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, VIC, Australia.
| |
Collapse
|
172
|
Mathipa MG, Thantsha MS, Bhunia AK. Lactobacillus casei expressing Internalins A and B reduces Listeria monocytogenes interaction with Caco-2 cells in vitro. Microb Biotechnol 2019; 12:715-729. [PMID: 30989823 PMCID: PMC6559204 DOI: 10.1111/1751-7915.13407] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/28/2022] Open
Abstract
Listeria monocytogenes has been implicated in a number of outbreaks including the recent largest outbreak in South Africa. Current methods for prevention of foodborne L. monocytogenes infection are inadequate, thus raising a need for an alternative strategy. Probiotic bioengineering is considered a prevailing approach to enhance the efficacy of probiotics for targeted control of pathogens. Here, the ability of Lactobacillus casei expressing the L. monocytogenes invasion proteins Internalins A and B (inlAB) to prevent infection was investigated. The inlAB operon was cloned and surface‐expressed on L. casei resulting in a recombinant strain, LbcInlAB, and subsequently, its ability to inhibit adhesion, invasion and translocation of L. monocytogenes through enterocyte‐like Caco‐2 cells was examined. Cell surface expression of InlAB on the LbcInlAB was confirmed by Western blotting and immunofluorescence staining. The LbcInlAB strain showed significantly higher (P < 0.0001) adherence, invasion and translocation of Caco‐2 cells than the wild‐type L. casei strain (LbcWT), as well as reduced L. monocytogenes adhesion, invasion and transcellular passage through the cell monolayer than LbcWT. Furthermore, pre‐exposure of Caco‐2 cells to LbcInlAB significantly reduced L. monocytogenes‐induced cell cytotoxicity and epithelial barrier dysfunction. These results suggest that InlAB‐expressing L. casei could be a potential practical approach for prevention of listeriosis.
Collapse
Affiliation(s)
- Moloko G Mathipa
- Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria, South Africa.,Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Mapitsi S Thantsha
- Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.,Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
173
|
Fernandez-Moure JS, Mydlowska A, Shin C, Vella M, Kaplan LJ. Nanometric Considerations in Biofilm Formation. Surg Infect (Larchmt) 2019; 20:167-173. [DOI: 10.1089/sur.2018.237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
| | - Anna Mydlowska
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Michael Vella
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lewis J. Kaplan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Corporal Michael J Crescenz VA Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|
174
|
Zhu Z, Dong C, Weng S, He J. Identification of outer membrane protein TolC as the major adhesin and potential vaccine candidate for Vibrio harveyi in hybrid grouper, Epinephelus fuscoguttatus (♀) × E. lanceolatus (♂). FISH & SHELLFISH IMMUNOLOGY 2019; 86:143-151. [PMID: 30453046 DOI: 10.1016/j.fsi.2018.11.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/13/2018] [Accepted: 11/15/2018] [Indexed: 05/21/2023]
Abstract
Vibrio harveyi is a serious pathogen of scale drop and muscle necrosis disease in marine commercial fishes. Adhesion to and colonization of the host cells surfaces is the first and crucial step for pathogenic bacterial infection, which is usually mediated by outer membrane proteins (Omps). The objectives of this study were to identify the major adhesin in Omps that plays the essential role in adhesion of V. harveyi to the host cells, and to assess the potential of this adhesin as a vaccine candidate for V. harveyi infection. We observed that pathogenic V. harveyi adhered to the surface of grouper embryonic cells (GEM cells) and induced apoptosis of them. Native Omps were extracted from nine different V. harveyi strains, and five common Omp bands were isolated by SDS-PAGE analysis. Western blot analysis and an anti-native Omp antibodies blocking assay indicated that one strong and several weak immunoreactivity Omps bands presence. Next, a total of five Omps, including TolC, Agg (Agglutination protein), Omp47, Fla (Flagellin), and OmpW, were identified and their encoding genes were cloned, characterized, and expressed in E. coli. The purified recombinant TolC could competitively inhibit the invasion of V. harveyi to GEM cells in vitro, and anti-TolC antibody also could significantly block the adhesion of V. harveyi to GEM cells. When used to immunize hybrid groupers, the recombinant TolC could confer significant protection to fish against experimental V. harveyi challenge. These data suggested that outer membrane protein TolC functions as a major adhesin in V. harveyi and could be a potential vaccine candidate for V. harveyi infection.
Collapse
Affiliation(s)
- Zhiming Zhu
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China; State Key Laboratory of Biocontrol / MOE Key Laboratory of Aquatic Product Safety, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China; Institute of Aquatic Economic Animals, and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Chuanfu Dong
- State Key Laboratory of Biocontrol / MOE Key Laboratory of Aquatic Product Safety, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China; Institute of Aquatic Economic Animals, and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China.
| | - Shaoping Weng
- State Key Laboratory of Biocontrol / MOE Key Laboratory of Aquatic Product Safety, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China; Institute of Aquatic Economic Animals, and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Jianguo He
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China; State Key Laboratory of Biocontrol / MOE Key Laboratory of Aquatic Product Safety, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, PR China; Institute of Aquatic Economic Animals, and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China.
| |
Collapse
|
175
|
Inhibitory effects of polysorbate 80 on MRSA biofilm formed on different substrates including dermal tissue. Sci Rep 2019; 9:3128. [PMID: 30816342 PMCID: PMC6395670 DOI: 10.1038/s41598-019-39997-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/04/2019] [Indexed: 12/04/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) forms biofilms on necrotic tissues and medical devices, and causes persistent infections. Surfactants act on biofilms, but their mode of action is still unknown. If used in the clinic, cytotoxicity in tissues should be minimized. In this study, we investigated the inhibitory effect of four different surfactants on MRSA biofilm formation, and found that a nonionic surfactant, polysorbate 80 (PS80), was the most suitable. The biofilm inhibitory effects resulted from the inhibition of bacterial adhesion to substrates rather than biofilm disruption, and the effective dose was less cytotoxic for 3T3 fibroblasts. However, the effects were substrate-dependent: positive for plastic, silicon, and dermal tissues, but negative for stainless-steel. These results indicate that PS80 is effective for prevention of biofilms formed by MRSA on tissues and foreign bodies. Therefore, PS80 could be used in medical practice as a washing solution for wounds and/or pretreatment of indwelling catheters.
Collapse
|
176
|
Asadi Karam MR, Habibi M, Bouzari S. Urinary tract infection: Pathogenicity, antibiotic resistance and development of effective vaccines against Uropathogenic Escherichia coli. Mol Immunol 2019; 108:56-67. [PMID: 30784763 DOI: 10.1016/j.molimm.2019.02.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 02/02/2019] [Accepted: 02/12/2019] [Indexed: 12/13/2022]
Abstract
Urinary tract infections (UTIs) are recognized as one of the most common infectious diseases in the world that can be divided to different types. Uropathogenic Escherichia coli (UPEC) strains are the most prevalent causative agent of UTIs that applied different virulence factors such as fimbriae, capsule, iron scavenger receptors, flagella, toxins, and lipopolysaccharide for their pathogenicity in the urinary tract. Despite the high pathogenicity of UPEC strains, host utilizes different immune systems such as innate and adaptive immunity for eradication of them from the urinary tract. The routine therapy of UTIs is based on the use of antibiotics such as β-lactams, trimethoprim, nitrofurantoin and quinolones in many countries. Unfortunately, the widespread and misuse of these antibiotics resulted in the increasing rate of resistance to them in the societies. Increasing antibiotic resistance and their side effects on human body show the need to develop alternative strategies such as vaccine against UTIs. Developing a vaccine against UTI pathogens will have an important role in reduction the mortality rate as well as reducing economic costs. Different vaccines based on the whole cells (killed or live-attenuated vaccines) and antigens (subunits, toxins and conjugatedvaccines) have been evaluated against UTIs pathogens. Furthermore, other therapeutic strategies such as the use of probiotics and antimicrobial peptides are considered against UTIs. Despite the extensive efforts, limited success has been achieved and more studies are needed to reach an alternative of antibiotics for treatment of UTIs.
Collapse
Affiliation(s)
| | - Mehri Habibi
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave., Tehran, 13164, Iran.
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave., Tehran, 13164, Iran.
| |
Collapse
|
177
|
Martín C, Escobedo S, Pérez-Martínez G, Coll-Marqués JM, Martín R, Suárez JE, Quirós LM. Two alkaline motifs in the Lactobacillus salivarius Lv72 OppA surface are important to its adhesin function. Benef Microbes 2019; 10:101-109. [PMID: 30406694 DOI: 10.3920/bm2018.0052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Glycosaminoglycans are involved in the attachment of Lactobacillus salivarius Lv72, a strain of vaginal origin, to HeLa cell cultures, indicating that they play a fundamental role in the attachment of mutualistic bacteria to the epithelium lining cavities where the normal microbiota thrives. The bacterial OppA protein has been proposed as an adhesin involved in this adherence since, once purified, it significantly interferes with attachment of the lactobacilli to HeLa cell cultures. In this article, the role of OppA is confirmed through the determination of its location at the cell surface and its ability to promote Lactobacillus casei and Lactococcus lactis adherence to eukaryotic cell cultures upon cloning and expression of oppA in these bacteria. The OppA sequence showed five potential domains for glycosaminoglycan-binding, and structural modelling of the protein showed that two of them were located in the vicinity of an OppA superficial groove whose width approached the diameter of the helical form of heparin in solution. Their involvement in the binding was demonstrated through substitution of critical basic amino acids by acidic ones, which resulted in loss of affinity for heparan sulphate and chondroitin sulphate depending on the domain mutated, suggesting that there might be a certain degree of specialisation. In addition, circular dichroism analysis showed that the spectrum changes induced by OppA-heparan sulphate binding were attenuated by the variant proteins, indicating that these motifs are the OppA recognition domains for the eukaryotic cell surface.
Collapse
Affiliation(s)
- C Martín
- 1 Área de Microbiología, Universidad de Oviedo, Julián Clavería 6, 33006 Oviedo, Spain.,2 Instituto Universitario Fernández- Vega, Universidad de Oviedo, Av. Doctores Fernández Vega, 34, 33012 Oviedo, Spain
| | - S Escobedo
- 1 Área de Microbiología, Universidad de Oviedo, Julián Clavería 6, 33006 Oviedo, Spain.,3 Instituto Universitario de Biotecnología, Universidad de Oviedo, Doctor Fernando Bongera s/n, 33006 Oviedo, Spain
| | - G Pérez-Martínez
- 4 Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Catedràtic Agustín Escardino Benlloch, 7, 46980 Valencia, Spain
| | - J M Coll-Marqués
- 4 Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Catedràtic Agustín Escardino Benlloch, 7, 46980 Valencia, Spain
| | - R Martín
- 5 National Institute of Agricultural Research, Commensals and Probiotics- Host Interactions Laboratory, Micalis Institute, AgroParisTech, Paris-Sud University, Allée de Vilvert, 78352 Jouy-en- Josas, France
| | - J E Suárez
- 1 Área de Microbiología, Universidad de Oviedo, Julián Clavería 6, 33006 Oviedo, Spain.,3 Instituto Universitario de Biotecnología, Universidad de Oviedo, Doctor Fernando Bongera s/n, 33006 Oviedo, Spain.,6 Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Río Linares, s/n, 33300 Villaviciosa, Spain
| | - L M Quirós
- 1 Área de Microbiología, Universidad de Oviedo, Julián Clavería 6, 33006 Oviedo, Spain.,2 Instituto Universitario Fernández- Vega, Universidad de Oviedo, Av. Doctores Fernández Vega, 34, 33012 Oviedo, Spain
| |
Collapse
|
178
|
Taylor VL, Fitzpatrick AD, Islam Z, Maxwell KL. The Diverse Impacts of Phage Morons on Bacterial Fitness and Virulence. Adv Virus Res 2019; 103:1-31. [PMID: 30635074 DOI: 10.1016/bs.aivir.2018.08.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The viruses that infect bacteria, known as phages, are the most abundant biological entity on earth. They play critical roles in controlling bacterial populations through phage-mediated killing, as well as through formation of bacterial lysogens. In this form, the survival of the phage depends on the survival of the bacterial host in which it resides. Thus, it is advantageous for phages to encode genes that contribute to bacterial fitness and expand the environmental niche. In many cases, these fitness factors also make the bacteria better able to survive in human infections and are thereby considered pathogenesis or virulence factors. The genes that encode these fitness factors, known as "morons," have been shown to increase bacterial fitness through a wide range of mechanisms and play important roles in bacterial diseases. This review outlines the benefits provided by phage morons in various aspects of bacterial life, including phage and antibiotic resistance, motility, adhesion and quorum sensing.
Collapse
Affiliation(s)
| | | | - Zafrin Islam
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Karen L Maxwell
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
179
|
Crystal structure of GAPDH of Streptococcus agalactiae and characterization of its interaction with extracellular matrix molecules. Microb Pathog 2018; 127:359-367. [PMID: 30553015 DOI: 10.1016/j.micpath.2018.12.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/10/2018] [Indexed: 11/21/2022]
Abstract
GAPDH being a key enzyme in the glycolytic pathway is one of the surface adhesins of many Gram-positive bacteria including Streptococcus agalactiae. This anchorless adhesin is known to bind to host plasminogen (PLG) and fibrinogen (Fg), which enhances the virulence and modulates the host immune system. The crystal structure of the recombinant GAPDH from S. agalactiae (SagGAPDH) was determined at 2.6 Å resolution by molecular replacement. The structure was found to be highly conserved with a typical NAD binding domain and a catalytic domain. In this paper, using biolayer interferometry studies, we report that the multifunctional SagGAPDH enzyme binds to a variety of host molecules such as PLG, Fg, laminin, transferrin and mucin with a KD value of 4.4 × 10-7 M, 9.8 × 10-7 M, 1 × 10-5 M, 9.7 × 10-12 M and 1.4 × 10-7 M respectively. The ligand affinity blots reveal that SagGAPDH binds specifically to α and β subunits of Fg and the competitive binding ELISA assay reveals that the Fg and PLG binding sites on GAPDH does not overlap each other. The PLG binding motif of GAPDH varies with organisms, however positively charged residues in the hydrophobic surroundings is essential for PLG binding. The lysine analogue competitive binding assay and lysine succinylation experiments deciphered the role of SagGAPDH lysines in PLG binding. On structural comparison with S. pneumoniae GAPDH, K171 of SagGAPDH is being predicted to be involved in PLG binding. Further SagGAPDH exhibited enzymatic activity in the presence of Fg, PLG and transferrin. This suggests that these host molecules does not mask the active site and bind at some other region of GAPDH.
Collapse
|
180
|
Planktonic Interference and Biofilm Alliance between Aggregation Substance and Endocarditis- and Biofilm-Associated Pili in Enterococcus faecalis. J Bacteriol 2018; 200:JB.00361-18. [PMID: 30249706 PMCID: PMC6256026 DOI: 10.1128/jb.00361-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022] Open
Abstract
Most bacteria express multiple adhesins that contribute to surface attachment and colonization. However, the network and relationships between the various adhesins of a single bacterial species are less well understood. Here, we examined two well-characterized adhesins in Enterococcus faecalis, aggregation substance and endocarditis- and biofilm-associated pili, and found that they exhibit distinct functional contributions depending on the growth stage of the bacterial community. Pili interfere with aggregation substance-mediated clumping and plasmid transfer under planktonic conditions, whereas the two adhesins structurally complement one another during biofilm development. This study advances our understanding of how E. faecalis, a ubiquitous member of the human gut microbiome and an opportunistic pathogen, uses multiple surface structures to evolve and thrive. Like many bacteria, Enterococcus faecalis encodes a number of adhesins involved in colonization or infection of different niches. Two well-studied E. faecalis adhesins, aggregation substance (AS) and endocarditis- and biofilm-associated pili (Ebp), both contribute to biofilm formation on abiotic surfaces and in endocarditis, suggesting that they may be expressed at the same time. Because different regulatory pathways have been reported for AS and Ebp, here, we examined if they are coexpressed on the same cells and what is the functional impact of coexpression on individual cells and within a population. We found that while Ebp are only expressed on a subset of cells, when Ebp and AS are expressed on the same cells, pili interfere with AS-mediated clumping and impede AS-mediated conjugative plasmid transfer during planktonic growth. However, when the population density increases, horizontal gene transfer rates normalize and are no longer affected by pilus expression. Instead, at higher cell densities during biofilm formation, Ebp and AS differentially contribute to biofilm development and structure, synergizing to promote maximal biofilm formation. IMPORTANCE Most bacteria express multiple adhesins that contribute to surface attachment and colonization. However, the network and relationships between the various adhesins of a single bacterial species are less well understood. Here, we examined two well-characterized adhesins in Enterococcus faecalis, aggregation substance and endocarditis- and biofilm-associated pili, and found that they exhibit distinct functional contributions depending on the growth stage of the bacterial community. Pili interfere with aggregation substance-mediated clumping and plasmid transfer under planktonic conditions, whereas the two adhesins structurally complement one another during biofilm development. This study advances our understanding of how E. faecalis, a ubiquitous member of the human gut microbiome and an opportunistic pathogen, uses multiple surface structures to evolve and thrive.
Collapse
|
181
|
Kumar SS, Tandberg JI, Penesyan A, Elbourne LDH, Suarez-Bosche N, Don E, Skadberg E, Fenaroli F, Cole N, Winther-Larsen HC, Paulsen IT. Dual Transcriptomics of Host-Pathogen Interaction of Cystic Fibrosis Isolate Pseudomonas aeruginosa PASS1 With Zebrafish. Front Cell Infect Microbiol 2018; 8:406. [PMID: 30524971 PMCID: PMC6262203 DOI: 10.3389/fcimb.2018.00406] [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] [Received: 08/12/2018] [Accepted: 10/29/2018] [Indexed: 01/09/2023] Open
Abstract
Pseudomonas aeruginosa is a significant cause of mortality in patients with cystic fibrosis (CF). To explore the interaction of the CF isolate P. aeruginosa PASS1 with the innate immune response, we have used Danio rerio (zebrafish) as an infection model. Confocal laser scanning microscopy (CLSM) enabled visualization of direct interactions between zebrafish macrophages and P. aeruginosa PASS1. Dual RNA-sequencing of host-pathogen was undertaken to profile RNA expression simultaneously in the pathogen and the host during P. aeruginosa infection. Following establishment of infection in zebrafish embryos with PASS1, 3 days post infection (dpi), there were 6739 genes found to be significantly differentially expressed in zebrafish and 176 genes in PASS1. A range of virulence genes were upregulated in PASS1, including genes encoding pyoverdine biosynthesis, flagellin, non-hemolytic phospholipase C, proteases, superoxide dismutase and fimbrial subunits. Additionally, iron and phosphate acquisition genes were upregulated in PASS1 cells in the zebrafish. Transcriptional changes in the host immune response genes highlighted phagocytosis as a key response mechanism to PASS1 infection. Transcriptional regulators of neutrophil and macrophage phagocytosis were upregulated alongside transcriptional regulators governing response to tissue injury, infection, and inflammation. The zebrafish host showed significant downregulation of the ribosomal RNAs and other genes involved in translation, suggesting that protein translation in the host is affected by PASS1 infection.
Collapse
Affiliation(s)
- Sheemal S Kumar
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Julia I Tandberg
- Department of Pharmaceutical Biosciences, Centre of Integrative Microbial Evolution, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Anahit Penesyan
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Liam D H Elbourne
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Nadia Suarez-Bosche
- Microscopy Unit, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Emily Don
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Eline Skadberg
- Department of Pharmaceutical Biosciences, Centre of Integrative Microbial Evolution, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Federico Fenaroli
- Department of Biosciences, The Faculty of Mathematic and Natural Sciences, University of Oslo, Oslo, Norway
| | - Nicholas Cole
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Hanne Cecilie Winther-Larsen
- Department of Pharmaceutical Biosciences, Centre of Integrative Microbial Evolution, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Ian T Paulsen
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
182
|
Mittal A, Singh V, Chowdhary S, Moideen A, Kumar D, Maniar K, Bhattacharyya R, Banerjee D. The Effect of Recombinant Human Erythropoietin on Bacterial Growth: A Dual-Edged Sword. KIDNEY DISEASES 2018; 5:81-90. [PMID: 31001539 DOI: 10.1159/000493684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/12/2018] [Indexed: 01/17/2023]
Abstract
Background Hypererythropoietinemia is associated with common diseases like non-uremic anaemia where infection burden is high. Erythropoietin (EPO) is also given as therapy for anaemia associated with chronic kidney disease and cancer and in those who are at a higher risk of infections. EPO is known to have an effect on macrophages by which it helps in the growth of some intracellular pathogens. However, its direct role on bacterial growth is currently unknown. Summary Here, we investigated the direct effect of recombinant human erythropoietin (rhuEPO) on the growth of pathogenic Escherichia coli, Staphylococcus aureus and Pseudomonas aeruginosa. In silico experiments were designed to gain insight into the mechanisms. We found that 30 IU/L rhuEPO promoted the growth of E. coli and S. aureus and inhibited the growth of P. aeruginosa. In silico observations suggest that bacterial cell surface proteins may interact with the EPO and may cause the observed effects. Key Message It appears that some pathogens can explore EPO to proliferate and growth of others are inhibited by the same. The consequence of such observation is a matter of widespread concern for future research.
Collapse
Affiliation(s)
- Ankur Mittal
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vandana Singh
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sheemona Chowdhary
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amal Moideen
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepak Kumar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kunal Maniar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajasri Bhattacharyya
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Dibyajyoti Banerjee
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
183
|
Trifonova A, Strateva T. Stenotrophomonas maltophilia – a low-grade pathogen with numerous virulence factors. Infect Dis (Lond) 2018; 51:168-178. [DOI: 10.1080/23744235.2018.1531145] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Angelina Trifonova
- Laboratory of Microbiology, Department of Military Epidemiology and Hygiene, Military Medical Academy, Sofia, Bulgaria
| | - Tanya Strateva
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
184
|
Pakharukova N, McKenna S, Tuittila M, Paavilainen S, Malmi H, Xu Y, Parilova O, Matthews S, Zavialov AV. Archaic and alternative chaperones preserve pilin folding energy by providing incomplete structural information. J Biol Chem 2018; 293:17070-17080. [PMID: 30228191 DOI: 10.1074/jbc.ra118.004170] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/14/2018] [Indexed: 11/06/2022] Open
Abstract
Adhesive pili are external component of fibrous adhesive organelles and help bacteria attach to biotic or abiotic surfaces. The biogenesis of adhesive pili via the chaperone-usher pathway (CUP) is independent of external energy sources. In the classical CUP, chaperones transport assembly-competent pilins in a folded but expanded conformation. During donor-strand exchange, pilins subsequently collapse, producing a tightly packed hydrophobic core and releasing the necessary free energy to drive fiber formation. Here, we show that pilus biogenesis in non-classical, archaic, and alternative CUPs uses a different source of conformational energy. High-resolution structures of the archaic Csu-pili system from Acinetobacter baumannii revealed that non-classical chaperones employ a short donor strand motif that is insufficient to fully complement the pilin fold. This results in chaperone-bound pilins being trapped in a substantially unfolded intermediate. The exchange of this short motif with the longer donor strand from adjacent pilin provides the full steric information essential for folding, and thereby induces a large unfolded-to-folded conformational transition to drive assembly. Our findings may inform the development of anti-adhesion drugs (pilicides) to combat bacterial infections.
Collapse
Affiliation(s)
- Natalia Pakharukova
- From the Department of Chemistry, University of Turku, Joint Biotechnology Laboratory (JBL), Arcanum, Vatselankatu 2, Turku FIN-20500, Finland and
| | - Sophie McKenna
- the Department of Life Sciences, Imperial College London, South Kensington Campus, London SW72AZ, United Kingdom
| | - Minna Tuittila
- From the Department of Chemistry, University of Turku, Joint Biotechnology Laboratory (JBL), Arcanum, Vatselankatu 2, Turku FIN-20500, Finland and
| | - Sari Paavilainen
- From the Department of Chemistry, University of Turku, Joint Biotechnology Laboratory (JBL), Arcanum, Vatselankatu 2, Turku FIN-20500, Finland and
| | - Henri Malmi
- From the Department of Chemistry, University of Turku, Joint Biotechnology Laboratory (JBL), Arcanum, Vatselankatu 2, Turku FIN-20500, Finland and
| | - Yingqi Xu
- the Department of Life Sciences, Imperial College London, South Kensington Campus, London SW72AZ, United Kingdom
| | - Olena Parilova
- From the Department of Chemistry, University of Turku, Joint Biotechnology Laboratory (JBL), Arcanum, Vatselankatu 2, Turku FIN-20500, Finland and
| | - Steve Matthews
- the Department of Life Sciences, Imperial College London, South Kensington Campus, London SW72AZ, United Kingdom
| | - Anton V Zavialov
- From the Department of Chemistry, University of Turku, Joint Biotechnology Laboratory (JBL), Arcanum, Vatselankatu 2, Turku FIN-20500, Finland and
| |
Collapse
|
185
|
Wang J, Chen HJ, Hang T, Yu Y, Liu G, He G, Xiao S, Yang BR, Yang C, Liu F, Tao J, Wu MX, Xie X. Physical activation of innate immunity by spiky particles. NATURE NANOTECHNOLOGY 2018; 13:1078-1086. [PMID: 30374159 PMCID: PMC7432992 DOI: 10.1038/s41565-018-0274-0] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/04/2018] [Indexed: 05/18/2023]
Abstract
Microbial biochemicals have been indicated as the primary stimulators of innate immunity, the first line of the body's defence against infections. However, the influence of topological features on a microbe's surface on immune responses remains largely unknown. Here we demonstrate the ability of TiO2 microparticles decorated with nanospikes (spiky particles) to activate and amplify the immune response in vitro and in vivo. The nanospikes exert mechanical stress on the cells, which results in potassium efflux and inflammasome activation in macrophages and dendritic cells during phagocytosis. The spiky particles augment antigen-specific humoral and cellular immune responses in the presence of monophosphoryl lipid A and elicit protective immunity against tumour growth and influenza viral infection. The study offers insights into how surface physical cues can tune the activation of innate immunity and provides a basis for engineering particles with increased immunogenicity and adjuvanticity.
Collapse
Affiliation(s)
- Ji Wang
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Tian Hang
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Yang Yu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Guishi Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Shuai Xiao
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Bo-Ru Yang
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Chengduan Yang
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Fanmao Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Jun Tao
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, USA.
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology, Guangdong Province Key Laboratory of Display Material and Technology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
186
|
Aparicio D, Torres-Puig S, Ratera M, Querol E, Piñol J, Pich OQ, Fita I. Mycoplasma genitalium adhesin P110 binds sialic-acid human receptors. Nat Commun 2018; 9:4471. [PMID: 30367053 PMCID: PMC6203739 DOI: 10.1038/s41467-018-06963-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 09/27/2018] [Indexed: 01/30/2023] Open
Abstract
Adhesion of pathogenic bacteria to target cells is a prerequisite for colonization and further infection. The main adhesins of the emerging sexually transmitted pathogen Mycoplasma genitalium, P140 and P110, interact to form a Nap complex anchored to the cell membrane. Herein, we present the crystal structures of the extracellular region of the virulence factor P110 (916 residues) unliganded and in complex with sialic acid oligosaccharides. P110 interacts only with the neuraminic acid moiety of the oligosaccharides and experiments with human cells demonstrate that these interactions are essential for mycoplasma cytadherence. Additionally, structural information provides a deep insight of the P110 antigenic regions undergoing programmed variation to evade the host immune response. These results enlighten the interplay of M. genitalium with human target cells, offering new strategies to control mycoplasma infections. How the Mycoplasma genitalium cytadhesins P140 and P110 promote host cell invasion remains poorly understood. Here, combining structural analysis with functional assays, Aparicio et al. identify the P110 domain that binds to sialylated receptors essential for mycoplasma cytadherence.
Collapse
Affiliation(s)
- David Aparicio
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC) and Maria de Maeztu Unit of Excellence, Parc Científic de Barcelona, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Sergi Torres-Puig
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Mercè Ratera
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC) and Maria de Maeztu Unit of Excellence, Parc Científic de Barcelona, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Enrique Querol
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Jaume Piñol
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Oscar Q Pich
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | - Ignacio Fita
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC) and Maria de Maeztu Unit of Excellence, Parc Científic de Barcelona, Baldiri Reixac 10, 08028, Barcelona, Spain.
| |
Collapse
|
187
|
Inhibition of adherence of the yeast Candida albicans to buccal epithelial cells by synthetic aromatic glycoconjugates. Eur J Med Chem 2018; 160:82-93. [PMID: 30321803 DOI: 10.1016/j.ejmech.2018.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/19/2018] [Accepted: 10/05/2018] [Indexed: 01/06/2023]
Abstract
The yeast Candida albicans is an opportunistic fungal pathogen which induces superficial and systemic infections in immunocompromised patients. Adherence to host tissue is critical to its ability to colonise and infect the host. The work presented here describes the synthesis of a small library of aromatic glycoconjugates (AGCs) and their evaluation as inhibitors of C. albicans adherence to exfoliated buccal epithelial cells (BECs). We identified a divalent galactoside, ligand 2a, capable of displacing over 50% of yeast cells already attached to the BECs. Fluorescence imaging indicates that 2a may bind to structural components of the fungal cell wall.
Collapse
|
188
|
Meyburgh CM, Bragg RR, Boucher CE. Detection of virulence factors of South African Lactococcus garvieae isolated from rainbow trout, Oncorhynchus mykiss (Walbaum). Onderstepoort J Vet Res 2018; 85:e1-e9. [PMID: 30326716 PMCID: PMC6324078 DOI: 10.4102/ojvr.v85i1.1568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 11/25/2022] Open
Abstract
Lactococcus garvieae is a Gram-positive bacterium that causes mortalities in freshwater and marine fish worldwide and therefore results in severe economic losses in the aquaculture industry. Apart from the apparent integral role of the exopolysaccharide (EPS) capsule in pathogenesis, factors associated with virulence of this bacterium are poorly understood. However, recent studies have indicated that the ability of L. garvieae to cause disease does not depend on the presence of the EPS capsule. Lack of knowledge of virulence factors, pathogenesis and serology of L. garvieae is an impediment to the development of effective typing methods and control measures. This study, therefore, aimed to detect the presence of EPS capsules and other putative virulence factors in South African L. garvieae fish pathogenic isolates and a non-virulent isolate, and to identify possible candidates for subunit vaccine development. No indication of the presence of the EPS capsule was detected by negative staining or amplification of the EPS biosynthesis gene cluster in the virulent isolates or the avirulent strain, discrediting the notion that the EPS capsule is the sole determinant of virulence. However, a set of putative virulence factor genes was detected in all isolates, and candidates for subunit vaccine development (enolase, lactate dehydrogenase phosphoenolpyruvate-protein phosphotransferase) were identified by identification of extracellular proteins of virulent strains.
Collapse
Affiliation(s)
- Cornelia M Meyburgh
- Department of Microbial, Biochemical and Food Biotechnology, University of the Free State.
| | | | | |
Collapse
|
189
|
Tomlin H, Piccinini AM. A complex interplay between the extracellular matrix and the innate immune response to microbial pathogens. Immunology 2018; 155:186-201. [PMID: 29908065 PMCID: PMC6142291 DOI: 10.1111/imm.12972] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/26/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022] Open
Abstract
The role of the host extracellular matrix (ECM) in infection tends to be neglected. However, the complex interactions between invading pathogens, host tissues and immune cells occur in the context of the ECM. On the pathogen side, a variety of surface and secreted molecules, including microbial surface components recognizing adhesive matrix molecules and tissue-degrading enzymes, are employed that interact with different ECM proteins to effectively establish an infection at specific sites. Microbial pathogens can also hijack or misuse host proteolytic systems to modify the ECM, evade immune responses or process biologically active molecules such as cell surface receptors and cytokines that direct cell behaviour and immune defence. On the host side, the ECM composition and three-dimensional ultrastructure undergo significant modifications, which have a profound impact on the specific signals that the ECM conveys to immune cells at the forefront of infection. Unexpectedly, activated immune cells participate in the remodelling of the local ECM by synthesizing ECM glycoproteins, proteoglycans and collagen molecules. The close interplay between the ECM and the innate immune response to microbial pathogens ultimately affects the outcome of infection. This review explores and discusses recent data that implicate an active role for the ECM in the immune response to infection, encompassing antimicrobial activities, microbial recognition, macrophage activation, phagocytosis, leucocyte population balance, and transcriptional and post-transcriptional regulation of inflammatory networks, and may foster novel antimicrobial approaches.
Collapse
Affiliation(s)
- Hannah Tomlin
- School of PharmacyUniversity of NottinghamNottinghamUK
| | | |
Collapse
|
190
|
|
191
|
SssP1, a Streptococcus suis Fimbria-Like Protein Transported by the SecY2/A2 System, Contributes to Bacterial Virulence. Appl Environ Microbiol 2018; 84:AEM.01385-18. [PMID: 30030221 DOI: 10.1128/aem.01385-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022] Open
Abstract
Streptococcus suis is an important Gram-positive pathogen in the swine industry and is an emerging zoonotic pathogen for humans. In our previous work, we found a virulent S. suis strain, CZ130302, belonging to a novel serotype, Chz, to be associated with acute meningitis in piglets. However, its underlying mechanisms of pathogenesis remain poorly understood. In this study, we sequenced and analyzed the complete genomes of three Chz serotype strains, including strain CZ130302 and two avirulent strains, HN136 and AH681. By genome comparison, we found two putative genomic islands (GIs) uniquely encoded in strain CZ130302 and designated them 50K GI and 58K GI. In mouse infection model, the deletion of 50K and 58K GIs caused 270-fold and 3-fold attenuation of virulence, respectively. Notably, we identified a complete SecY2/A2 system, coupled with its secretory protein SssP1 encoded in the 50K GI, which contributed to the pathogenicity of strain CZ130302. Immunogold electron microscopy and immunofluorescence analyses indicated that SssP1 could form fimbria-like structures that extend outward from the bacterial cell surface. The sssP1 mutation also attenuated bacterial adherence in human laryngeal epithelial (HEp-2) cells and human brain microvessel endothelial cells (HBMECs) compared with the wild type. Furthermore, we showed that two analogous Ig-like subdomains of SssP1 have sialic acid binding capacities. In conclusion, our results revealed that the 50K GI and the inside SecY2/A2 system gene cluster are related to the virulence of strain CZ130302, and we clarified a new S. suis pathogenesis mechanism mediated by the secretion protein SssP1.IMPORTANCE Streptococcus suis is an important zoonotic pathogen. Here, we managed to identify key factors to clarify the virulence of S. suis strain CZ130302 from a novel serotype, Chz. Notably, it was shown that a fimbria-like structure was significantly connected to the pathogenicity of the CZ130302 strain by comparative genomics analysis and animal infection assays. The mechanisms of how the CZ130302 strain constructs these fimbria-like structures in the cell surface by genes encoding and production transport were subsequently elucidated. Biosynthesis of the fimbria-like structure was achieved by the production of SssP1 glycoproteins, and its construction was dependent on the SecA2/Y2 secretion system. This study identified a visible fimbria-like protein, SssP1, participating in adhesion to host cells and contributing to the virulence in S. suis These findings will promote a better understanding of the pathogenesis of S. suis.
Collapse
|
192
|
Lessen HJ, Fleming PJ, Fleming KG, Sodt AJ. Building Blocks of the Outer Membrane: Calculating a General Elastic Energy Model for β-Barrel Membrane Proteins. J Chem Theory Comput 2018; 14:4487-4497. [PMID: 29979594 PMCID: PMC6191857 DOI: 10.1021/acs.jctc.8b00377] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The outer membranes of Gram negative bacteria are the first points of contact these organisms make with their environment. Understanding how composition determines the mechanical properties of this essential barrier is of paramount importance. Therefore, we developed a new computational method to measure the elasticity of transmembrane proteins found in the outer membrane. Using all-atom molecular dynamics simulations of these proteins, we apply a set of external forces to mechanically stress the transmembrane β-barrels. Our results from four representative β-barrels show that outer membrane proteins display elastic properties that are approximately 70 to 190 times stiffer than neat lipid membranes. These findings suggest that outer membrane β-barrels are a significant source of mechanical stability in bacteria. Our all-atom approach further reveals that resistance to radial stress is encoded by a general mechanism that includes stretching of backbone hydrogen bonds and tilting of β-strands with respect to the bilayer normal. This computational framework facilitates an increased theoretical understanding of how varying lipid and protein amounts affect the mechanical properties of the bacterial outer membrane.
Collapse
Affiliation(s)
- Henry J. Lessen
- T. C. Jenkins Department of Biophysics, Johns Hopkins University, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | - Patrick J. Fleming
- T. C. Jenkins Department of Biophysics, Johns Hopkins University, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | - Karen G. Fleming
- T. C. Jenkins Department of Biophysics, Johns Hopkins University, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | - Alexander J. Sodt
- T. C. Jenkins Department of Biophysics, Johns Hopkins University, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| |
Collapse
|
193
|
Dingjan T, Gillon É, Imberty A, Pérez S, Titz A, Ramsland PA, Yuriev E. Virtual Screening Against Carbohydrate-Binding Proteins: Evaluation and Application to Bacterial Burkholderia ambifaria Lectin. J Chem Inf Model 2018; 58:1976-1989. [DOI: 10.1021/acs.jcim.8b00185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Tamir Dingjan
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Émilie Gillon
- University Grenoble Alpes, CNRS, CERMAV, 38000 Grenoble, France
| | - Anne Imberty
- University Grenoble Alpes, CNRS, CERMAV, 38000 Grenoble, France
| | - Serge Pérez
- University Grenoble Alpes, CNRS, DPM, 38000 Grenoble, France
| | - Alexander Titz
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
- Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| | - Paul A. Ramsland
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia
- Department of Surgery Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
- Burnet Institute, Melbourne, Victoria 3004, Australia
| | - Elizabeth Yuriev
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
194
|
Guo Y, Matsuoka Y, Miura T, Nishizawa T, Ohta H, Narisawa K. Complete genome sequence of Agrobacterium pusense VsBac-Y9, a bacterial symbiont of the dark septate endophytic fungus Veronaeopsis simplex Y34 with potential for improving fungal colonization in roots. J Biotechnol 2018; 284:31-36. [PMID: 30077583 DOI: 10.1016/j.jbiotec.2018.07.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/27/2018] [Accepted: 07/31/2018] [Indexed: 12/24/2022]
Abstract
A Rhizobium-related bacterium (Rhizobium sp. VsBac-Y9) is a symbiont living with the dark septate endophytic (DSE) fungus Veronaeopsis simplex Y34. Co-inoculation of Rhizobium sp. VsBac-Y9 with V. simplex Y34 improves the fungal colonization of tomato roots, resulting in a significant increase in aboveground biomass. This study sequenced the complete genome of this V. simplex-helper bacterium using the PacBio and Illumina MiSeq platforms. Hybrid assembly using SPAdes outputted a circular chromosome, a linear chromid, and a circular plasmid for a total genome 5,321,211 bp in size with a G + C content of 59.2%. Analysis of concatenated housekeeping genes (atpD-dnaK-groEL-lepA-recA-rpoB-thrE) and calculation of average nucleotide identity, showed that VsBac-Y9 was affiliated with the species Agrobacterium pusense (syn. Rhizobium pusense). Genome analysis revealed that A. pusense VsBac-Y9 contains a series of genes responsible for the host interactions with both fungus and plant. Such genomic information will provide new insights into developing co-inoculants of endophytic fungus and its symbiotic bacterium in future agricultural innovation.
Collapse
Affiliation(s)
- Yong Guo
- College of Agriculture, Ibaraki University, Ibaraki, Japan.
| | - Yuuto Matsuoka
- Graduate School of Agriculture, Ibaraki University, Ibaraki, Japan
| | - Takamasa Miura
- Biological Resource Center, National Institute of Technology and Evaluation, Chiba, Japan
| | - Tomoyasu Nishizawa
- College of Agriculture, Ibaraki University, Ibaraki, Japan; Graduate School of Agriculture, Ibaraki University, Ibaraki, Japan
| | - Hiroyuki Ohta
- College of Agriculture, Ibaraki University, Ibaraki, Japan; Graduate School of Agriculture, Ibaraki University, Ibaraki, Japan
| | - Kazuhiko Narisawa
- College of Agriculture, Ibaraki University, Ibaraki, Japan; Graduate School of Agriculture, Ibaraki University, Ibaraki, Japan.
| |
Collapse
|
195
|
Díez-Vives C, Esteves AIS, Costa R, Nielsen S, Thomas T. Detecting signatures of a sponge-associated lifestyle in bacterial genomes. ENVIRONMENTAL MICROBIOLOGY REPORTS 2018; 10:433-443. [PMID: 29707906 DOI: 10.1111/1758-2229.12655] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 04/23/2018] [Indexed: 06/08/2023]
Abstract
Sponges interact with diverse and rich communities of bacteria that are phylogenetically often distinct from their free-living counterparts. Recent genomics and metagenomic studies have indicated that bacterial sponge symbionts also have distinct functional features from free-living bacteria; however, it is unclear, if such genome-derived functional signatures are common and present in different symbiont taxa. We therefore compared here a large set of genomes from cultured (Pseudovibrio, Ruegeria and Aquimarina) and yet-uncultivated (Synechococcus) bacteria found in either sponge-associated or free-living sources. Our analysis revealed only very few genera-specific functions that could be correlated with a sponge-associated lifestyle. Using different sets of sponge-associated and free-living bacteria for each genus, we could however show that the functions identified as 'sponge-associated' are dependent on the reference comparison being made. Using simulation approaches, we show how this influences the robustness of identifying functional signatures and how evolutionary divergence and genomic adaptation can be distinguished. Our results highlight the future need for robust comparative analyses to define genomic signatures of symbiotic lifestyles, whether it is for symbionts of sponges or other host organisms.
Collapse
Affiliation(s)
- Cristina Díez-Vives
- Centre for Marine Bio-Innovation and School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Ana I S Esteves
- Centre for Marine Bio-Innovation and School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Rodrigo Costa
- Department of Bioengineering, Institute for Bioengineering and Biosciences (IBB), IST, Universidade de Lisboa, Lisbon, Portugal
| | - Shaun Nielsen
- Centre for Marine Bio-Innovation and School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Torsten Thomas
- Centre for Marine Bio-Innovation and School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
196
|
Yu LCH, Wei SC, Ni YH. Impact of microbiota in colorectal carcinogenesis: lessons from experimental models. Intest Res 2018; 16:346-357. [PMID: 30090033 PMCID: PMC6077307 DOI: 10.5217/ir.2018.16.3.346] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023] Open
Abstract
A role of gut microbiota in colorectal cancer (CRC) growth was first suggested in germ-free rats almost 50 years ago, and the existence of disease-associated bacteria (termed pathobionts) had becoming increasingly evident from experimental data of fecal transplantation, and microbial gavage or monoassociation. Altered bacterial compositions in fecal and mucosal specimens were observed in CRC patients compared to healthy subjects. Microbial fluctuations were found at various cancer stages; an increase of bacterial diversity was noted in the adenoma specimens, while a reduction of bacterial richness was documented in CRC samples. The bacterial species enriched in the human cancerous tissues included Escherichia coli, Fusobacterium nucleatum, and enterotoxigenic Bacteroides fragilis. The causal relationship of gut bacteria in tumorigenesis was established by introducing particular bacterial strains in in situ mouse CRC models. Detailed experimental protocols of bacterial gavage and the advantages and caveats of different experimental models are summarized in this review. The microbial genotoxins, enterotoxins, and virulence factors implicated in the mechanisms of bacteria-driven tumorigenesis are described. In conclusion, intestinal microbiota is involved in colon tumorigenesis. Bacteria-targeting intervention would be the next challenge for CRC.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shu-Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Hsuan Ni
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
197
|
Weimer BC, Chen P, Desai PT, Chen D, Shah J. Whole Cell Cross-Linking to Discover Host-Microbe Protein Cognate Receptor/Ligand Pairs. Front Microbiol 2018; 9:1585. [PMID: 30072965 PMCID: PMC6060266 DOI: 10.3389/fmicb.2018.01585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 06/26/2018] [Indexed: 12/31/2022] Open
Abstract
Bacterial surface ligands mediate interactions with the host cell during association that determines the specific outcome for the host–microbe association. The association begins with receptors on the host cell binding ligands on the microbial cell to form a partnership that initiates responses in both cells. Methods to determine the specific cognate partnerships are lacking. Determining these molecular interactions between the host and microbial surfaces are difficult, yet crucial in defining biologically important events that are triggered during association of the microbiome, and critical in defining the initiating signal from the host membrane that results in pathology or commensal association. In this study, we designed an approach to discover cognate host–microbe receptor/ligand pairs using a covalent cross-linking strategy with whole cells. Protein/protein cross-linking occurred when the interacting molecules were within 9–12 Å, allowing for identification of specific pairs of proteins from the host and microbe that define the molecular interaction during association. To validate the method three different bacteria with three previously known protein/protein partnerships were examined. The exact interactions were confirmed and led to discovery of additional partnerships that were not recognized as cognate partners, but were previously reported to be involved in bacterial interactions. Additionally, three unknown receptor/ligand partners were discovered and validated with in vitro infection assays by blocking the putative host receptor and deleting the bacterial ligand. Subsequently, Salmonella enterica sv. Typhimurium was cross-linked to differentiated colonic epithelial cells (caco-2) to discover four previously unknown host receptors bound to three previously undefined host ligands for Salmonella. This approach resulted in a priori discovery of previously unknown and biologically important molecules for host/microbe association that were casually reported to mediate bacterial invasion. The whole cell cross-linking approach promises to enable discovery of possible targets to modulate interaction of the microbiome with the host that are important in infection and commensalism, both of with initiate a host response.
Collapse
Affiliation(s)
- Bart C Weimer
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Poyin Chen
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Prerak T Desai
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States.,Department of Dietetics, Nutrition and Food Sciences, Utah State University, Logan, UT, United States
| | - Dong Chen
- Department of Biology, Utah State University, Logan, UT, United States
| | - Jigna Shah
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States.,Department of Dietetics, Nutrition and Food Sciences, Utah State University, Logan, UT, United States
| |
Collapse
|
198
|
Mi G, Shi D, Wang M, Webster TJ. Reducing Bacterial Infections and Biofilm Formation Using Nanoparticles and Nanostructured Antibacterial Surfaces. Adv Healthc Mater 2018; 7:e1800103. [PMID: 29790304 DOI: 10.1002/adhm.201800103] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/29/2018] [Indexed: 02/02/2023]
Abstract
With the rapid spreading of resistance among common bacterial pathogens, bacterial infections, especially antibiotic-resistant bacterial infections, have drawn much attention worldwide. In light of this, nanoparticles, including metal and metal oxide nanoparticles, liposomes, polymersomes, and solid lipid nanoparticles, have been increasingly exploited as both efficient antimicrobials themselves or as delivery platforms to enhance the effectiveness of existing antibiotics. In addition to the emergence of widespread antibiotic resistance, of equal concern are implantable device-associated infections, which result from bacterial adhesion and subsequent biofilm formation at the site of implantation. The ineffectiveness of conventional antibiotics against these biofilms often leads to revision surgery, which is both debilitating to the patient and expensive. Toward this end, micro- and nanotopographies, especially those that resemble natural surfaces, and nonfouling chemistries represent a promising combination for long-term antibacterial activity. Collectively, the use of nanoparticles and nanostructured surfaces to combat bacterial growth and infections is a promising solution to the growing problem of antibiotic resistance and biofilm-related device infections.
Collapse
Affiliation(s)
- Gujie Mi
- Department of Chemical Engineering; 313 Snell Engineering Center; Northeastern University; 360 Huntington Avenue Boston MA 02115 USA
| | - Di Shi
- Department of Chemical Engineering; 313 Snell Engineering Center; Northeastern University; 360 Huntington Avenue Boston MA 02115 USA
| | - Mian Wang
- Department of Chemical Engineering; 313 Snell Engineering Center; Northeastern University; 360 Huntington Avenue Boston MA 02115 USA
| | - Thomas J. Webster
- Department of Chemical Engineering; 313 Snell Engineering Center; Northeastern University; 360 Huntington Avenue Boston MA 02115 USA
| |
Collapse
|
199
|
Pais IS, Valente RS, Sporniak M, Teixeira L. Drosophila melanogaster establishes a species-specific mutualistic interaction with stable gut-colonizing bacteria. PLoS Biol 2018; 16:e2005710. [PMID: 29975680 PMCID: PMC6049943 DOI: 10.1371/journal.pbio.2005710] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/17/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023] Open
Abstract
Animals live together with diverse bacteria that can impact their biology. In Drosophila melanogaster, gut-associated bacterial communities are relatively simple in composition but also have a strong impact on host development and physiology. It is generally assumed that gut bacteria in D. melanogaster are transient and their constant ingestion with food is required to maintain their presence in the gut. Here, we identify bacterial species from wild-caught D. melanogaster that stably associate with the host independently of continuous inoculation. Moreover, we show that specific Acetobacter wild isolates can proliferate in the gut. We further demonstrate that the interaction between D. melanogaster and the wild isolated Acetobacter thailandicus is mutually beneficial and that the stability of the gut association is key to this mutualism. The stable population in the gut of D. melanogaster allows continuous bacterial spreading into the environment, which is advantageous to the bacterium itself. The bacterial dissemination is in turn advantageous to the host because the next generation of flies develops in the presence of this particularly beneficial bacterium. A. thailandicus leads to a faster host development and higher fertility of emerging adults when compared to other bacteria isolated from wild-caught flies. Furthermore, A. thailandicus is sufficient and advantageous when D. melanogaster develops in axenic or freshly collected figs, respectively. This isolate of A. thailandicus colonizes several genotypes of D. melanogaster but not the closely related D. simulans, indicating that the stable association is host specific. This work establishes a new conceptual model to understand D. melanogaster-gut microbiota interactions in an ecological context; stable interactions can be mutualistic through microbial farming, a common strategy in insects. Moreover, these results develop the use of D. melanogaster as a model to study gut microbiota proliferation and colonization.
Collapse
Affiliation(s)
- Inês S. Pais
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | - Luis Teixeira
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
200
|
Adhesive interactions between milk fat globule membrane and Lactobacillus rhamnosus GG inhibit bacterial attachment to Caco-2 TC7 intestinal cell. Colloids Surf B Biointerfaces 2018; 167:44-53. [DOI: 10.1016/j.colsurfb.2018.03.044] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 03/07/2018] [Accepted: 03/27/2018] [Indexed: 12/17/2022]
|