1
|
Bloise N, Giannaccari M, Guagliano G, Peluso E, Restivo E, Strada S, Volpini C, Petrini P, Visai L. Growing Role of 3D In Vitro Cell Cultures in the Study of Cellular and Molecular Mechanisms: Short Focus on Breast Cancer, Endometriosis, Liver and Infectious Diseases. Cells 2024; 13:1054. [PMID: 38920683 PMCID: PMC11201503 DOI: 10.3390/cells13121054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Over the past decade, the development of three-dimensional (3D) models has increased exponentially, facilitating the unravelling of fundamental and essential cellular mechanisms by which cells communicate with each other, assemble into tissues and organs and respond to biochemical and biophysical stimuli under both physiological and pathological conditions. This section presents a concise overview of the most recent updates on the significant contribution of different types of 3D cell cultures including spheroids, organoids and organ-on-chip and bio-printed tissues in advancing our understanding of cellular and molecular mechanisms. The case studies presented include the 3D cultures of breast cancer (BC), endometriosis, the liver microenvironment and infections. In BC, the establishment of 3D culture models has permitted the visualization of the role of cancer-associated fibroblasts in the delivery of exosomes, as well as the significance of the physical properties of the extracellular matrix in promoting cell proliferation and invasion. This approach has also become a valuable tool in gaining insight into general and specific mechanisms of drug resistance. Given the considerable heterogeneity of endometriosis, 3D models offer a more accurate representation of the in vivo microenvironment, thereby facilitating the identification and translation of novel targeted therapeutic strategies. The advantages provided by 3D models of the hepatic environment, in conjunction with the high throughput characterizing various platforms, have enabled the elucidation of complex molecular mechanisms underlying various threatening hepatic diseases. A limited number of 3D models for gut and skin infections have been developed. However, a more profound comprehension of the spatial and temporal interactions between microbes, the host and their environment may facilitate the advancement of in vitro, ex vivo and in vivo disease models. Additionally, it may pave the way for the development of novel therapeutic approaches in diverse research fields. The interested reader will also find concluding remarks on the challenges and prospects of using 3D cell cultures for discovering cellular and molecular mechanisms in the research areas covered in this review.
Collapse
Affiliation(s)
- Nora Bloise
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of University of Pavia, 27100 Pavia, Italy
| | - Marialaura Giannaccari
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Giuseppe Guagliano
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, P.zza L. Da Vinci 32, 20133 Milan, Italy; (G.G.); (P.P.)
| | - Emanuela Peluso
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Elisa Restivo
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Silvia Strada
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Cristina Volpini
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Paola Petrini
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, P.zza L. Da Vinci 32, 20133 Milan, Italy; (G.G.); (P.P.)
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of Politecnico di Milano, 20133 Milan, Italy
| | - Livia Visai
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
2
|
Kunrath MF, Farina G, Sturmer LBS, Teixeira ER. TiO 2 nanotubes as an antibacterial nanotextured surface for dental implants: Systematic review and meta-analysis. Dent Mater 2024; 40:907-920. [PMID: 38714394 DOI: 10.1016/j.dental.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 05/09/2024]
Abstract
OBJECTIVES Nanotechnology is constantly advancing in dental science, progressing several features aimed at improving dental implants. An alternative for surface treatment of dental implants is electrochemical anodization, which may generate a nanotubular surface (TiO2 nanotubes) with antibacterial potential and osteoinductive features. This systematic review and meta-analysis aims to elucidate the possible antibacterial properties of the surface in question compared to the untreated titanium surface. SOURCES For that purpose, was performed a systematic search on the bases PubMed, Lilacs, Embase, Web Of Science, Cinahl, and Cochrane Central, as well as, manual searches and gray literature. STUDY SELECTION The searches resulted in 742 articles, of which 156 followed for full-text reading. Then, 37 were included in the systematic review and 8 were included in meta-analysis. RESULTS Fifteen studies revealed significant antibacterial protection using TiO2 nanotube surfaces, while 15 studies found no statistical difference between control and nanotextured surfaces. Meta-analysis of in vitro studies demonstrated relevant bacterial reduction only for studies investigating Staphylococcus aureus in a period of 6 h. Meta-analysis of in vivo studies revealed three times lower bacterial adhesion and proliferation on TiO2 nanotube surfaces. CONCLUSIONS TiO2 nanotube topography as a surface for dental implants in preclinical research has demonstrated a positive relationship with antibacterial properties, nevertheless, factors such as anodization protocols, bacteria strains, and mono-culture methods should be taken into consideration, consequently, further studies are necessary to promote clinical translatability.
Collapse
Affiliation(s)
- Marcel F Kunrath
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden; School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; School of Technology, Post-Graduate Program in Materials Technology and Engineering, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Georgia Farina
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luiza B S Sturmer
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Eduardo R Teixeira
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
3
|
Teng JLL, Ma Y, Chen JHK, Luo R, Foo CH, Li TT, Fong JYH, Yao W, Wong SSY, Fung KSC, Lau SKP, Woo PCY. Streptococcus oriscaviae sp. nov. Infection Associated with Guinea Pigs. Microbiol Spectr 2022; 10:e0001422. [PMID: 35510851 PMCID: PMC9241640 DOI: 10.1128/spectrum.00014-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/17/2022] [Indexed: 11/20/2022] Open
Abstract
Pet bite-related infections are commonly caused by the pet's oral flora transmitted to the animal handlers through the bite wounds. In this study, we isolated a streptococcus, HKU75T, in pure culture from the purulent discharge collected from a guinea pig bite wound in a previously healthy young patient. HKU75T was alpha-hemolytic on sheep blood agar and agglutinated with Lancefield group D and group G antisera. API 20 STREP showed that the most likely identity for HKU75T was S. suis I with 85.4% confidence while Vitek 2 showed that HKU75T was unidentifiable. MALDI-TOF MS identified HKU75T as Streptococcus suis (score of 1.86 only). 16S rRNA gene sequencing showed that HKU75T was most closely related to S. parasuis (98.3% nucleotide identity), whereas partial groEL and rpoB gene sequencing showed that it was most closely related to S. suis (81.8% and 89.8% nucleotide identity respectively). Whole genome sequencing and intergenomic distance determined by ANI revealed that there was <85% identity between the genome of HKU75T and those of all other known Streptococcus species. Genome classification using concatenated sequences of 92 bacterial core genes showed that HKU75T belonged to the Suis group. groEL gene sequences identical to that of HKU75T could be directly amplified from the oral cavities of the two guinea pigs owned by the patient. HKU75T is a novel Streptococcus species, which we propose to be named S. oriscaviae. The oral cavity of guinea pigs is presumably a reservoir of S. oriscaviae. Some of the reported S. suis strains isolated from clinical specimens may be S. oriscaviae. IMPORTANCE We reported the discovery of a novel Streptococcus species, propose to be named Streptococcus oriscaviae, from the pus collected from a guinea pig bite wound in a healthy young patient. The bacterium was initially misidentified as S. suis/S. parasuis by biochemical tests, mass spectrometry. and housekeeping genes sequencing. Its novelty was confirmed by whole genome sequencing. Comparative genomic studies showed that S. oriscaviae belongs to the Suis group. S. oriscaviae sequences were detected in the oral cavities of the two guinea pigs owned by the patient, suggesting that the oral cavity of guinea pigs could be a reservoir of S. oriscaviae. Some of the reported S. suis strains may be S. oriscaviae. Further studies are warranted to refine our knowledge on this novel Streptococcus species.
Collapse
Affiliation(s)
- Jade L. L. Teng
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuanchao Ma
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jonathan H. K. Chen
- Department of Microbiology, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Chuen-Hing Foo
- Department of Microbiology, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Tsz Tuen Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jordan Y. H. Fong
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Weiming Yao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Samson S. Y. Wong
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kitty S. C. Fung
- Department of Pathology, United Christian Hospital, Hong Kong, China
| | - Susanna K. P. Lau
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Patrick C. Y. Woo
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
4
|
Novel Developments on Stimuli-Responsive Probiotic Encapsulates: From Smart Hydrogels to Nanostructured Platforms. FERMENTATION 2022. [DOI: 10.3390/fermentation8030117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Biomaterials engineering and biotechnology have advanced significantly towards probiotic encapsulation with encouraging results in assuring sufficient bioactivity. However, some major challenges remain to be addressed, and these include maintaining stability in different compartments of the gastrointestinal tract (GIT), favoring adhesion only at the site of action, and increasing residence times. An alternative to addressing such challenges is to manufacture encapsulates with stimuli-responsive polymers, such that controlled release is achievable by incorporating moieties that respond to chemical and physical stimuli present along the GIT. This review highlights, therefore, such emerging delivery matrices going from a comprehensive description of addressable stimuli in each GIT compartment to novel synthesis and functionalization techniques to currently employed materials used for probiotic’s encapsulation and achieving multi-modal delivery and multi-stimuli responses. Next, we explored the routes for encapsulates design to enhance their performance in terms of degradation kinetics, adsorption, and mucus and gut microbiome interactions. Finally, we present the clinical perspectives of implementing novel probiotics and the challenges to assure scalability and cost-effectiveness, prerequisites for an eventual niche market penetration.
Collapse
|
5
|
Lopes GV, Ramires T, Kleinubing NR, Scheik LK, Fiorentini ÂM, Padilha da Silva W. Virulence factors of foodborne pathogen Campylobacterjejuni. Microb Pathog 2021; 161:105265. [PMID: 34699927 DOI: 10.1016/j.micpath.2021.105265] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/27/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022]
Abstract
Campylobacter jejuni is a highly frequent cause of gastrointestinal foodborne disease in humans throughout the world. Disease outcomes vary from mild to severe diarrhea, and in rare cases the Guillain-Barré syndrome or reactive arthritis can develop as a post-infection complication. Transmission to humans usually occurs via the consumption of a range of foods, especially those associated with the consumption of raw or undercooked poultry meat, unpasteurized milk, and water-based environmental sources. When associated to food or water ingestion, the C. jejuni enters the human host intestine via the oral route and colonizes the distal ileum and colon. When it adheres and colonizes the intestinal cell surfaces, the C. jejuni is expected to express several putative virulence factors, which cause damage to the intestine either directly, by cell invasion and/or production of toxin(s), or indirectly, by triggering inflammatory responses. This review article highlights various C. jejuni characteristics - such as motility and chemotaxis - that contribute to the biological fitness of the pathogen, as well as factors involved in human host cell adhesion and invasion, and their potential role in the development of the disease. We have analyzed and critically discussed nearly 180 scientific articles covering the latest improvements in the field.
Collapse
Affiliation(s)
- Graciela Volz Lopes
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Tassiana Ramires
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Natalie Rauber Kleinubing
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Letícia Klein Scheik
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Ângela Maria Fiorentini
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Wladimir Padilha da Silva
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil.
| |
Collapse
|
6
|
Xie X, Hao F, Chen R, Wang J, Wei Y, Liu J, Wang H, Zhang Z, Bai Y, Shao G, Xiong Q, Feng Z. Nicotinamide Adenine Dinucleotide-Dependent Flavin Oxidoreductase of Mycoplasma hyopneumoniae Functions as a Potential Novel Virulence Factor and Not Only as a Metabolic Enzyme. Front Microbiol 2021; 12:747421. [PMID: 34671334 PMCID: PMC8521518 DOI: 10.3389/fmicb.2021.747421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
Mycoplasma hyopneumoniae (Mhp) is the main pathogen that causes enzootic pneumonia, a disease that has a significant impact on the pig industry worldwide. The pathogenesis of enzootic pneumonia, especially possible virulence factors of Mhp, has still not been fully elucidated. The transcriptomic and proteomic analyses of different Mhp strains reported in the literature have revealed differences in virulence, and differences in RNA transcription levels between high- and low-virulence strains initially indicated that nicotinamide adenine dinucleotide (NADH)-dependent flavin oxidoreductase (NFOR) was related to Mhp pathogenicity. Prokaryotic expression and purification of the NFOR protein from Mhp were performed, a rabbit-derived polyclonal antibody against NFOR was prepared, and multiple sequence alignment and evolutionary analyses of Mhp NFOR were performed. For the first time, it was found that the NFOR protein was conserved among all Mhp strains, and NFOR was localized to the cell surface and could adhere to immortalized porcine bronchial epithelial cells (hTERT-PBECs). Adhesion to hTERT-PBECs could be specifically inhibited by an anti-NFOR polyclonal antibody, and the rates of adhesion to both high- and low-virulence strains, 168 and 168L, significantly decreased by more than 40%. Moreover, Mhp NFOR not only recognized and interacted with host fibronectin and plasminogen but also induced cellular oxidative stress and apoptosis in hTERT-PBECs. The release of lactate dehydrogenase by hTERT-PBECs incubated with Mhp NFOR was significantly positively correlated with the virulence of Mhp. Overall, in addition to being a metabolic enzyme related to oxidative stress, NFOR may also function as a potential novel virulence factor of Mhp, thus contributing to the pathogenesis of Mhp; these findings provide new ideas and theoretical support for studying the pathogenic mechanisms of other mycoplasmas.
Collapse
Affiliation(s)
- Xing Xie
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Fei Hao
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Rong Chen
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jingjing Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yanna Wei
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jin Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | - Haiyan Wang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Zhenzhen Zhang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Yun Bai
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Guoqing Shao
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Qiyan Xiong
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Zhixin Feng
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| |
Collapse
|
7
|
Han S, Lu Y, Xie J, Fei Y, Zheng G, Wang Z, Liu J, Lv L, Ling Z, Berglund B, Yao M, Li L. Probiotic Gastrointestinal Transit and Colonization After Oral Administration: A Long Journey. Front Cell Infect Microbiol 2021; 11:609722. [PMID: 33791234 PMCID: PMC8006270 DOI: 10.3389/fcimb.2021.609722] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Orally administered probiotics encounter various challenges on their journey through the mouth, stomach, intestine and colon. The health benefits of probiotics are diminished mainly due to the substantial reduction of viable probiotic bacteria under the harsh conditions in the gastrointestinal tract and the colonization resistance caused by commensal bacteria. In this review, we illustrate the factors affecting probiotic viability and their mucoadhesive properties through their journey in the gastrointestinal tract, including a discussion on various mucosadhesion-related proteins on the probiotic cell surface which facilitate colonization.
Collapse
Affiliation(s)
- Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanmeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaojiao Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqiu Fei
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziyuan Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology & Business University (BTBU), Beijing, China
| | - Jie Liu
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology & Business University (BTBU), Beijing, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongxin Ling
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Björn Berglund
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mingfei Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Järvå MA, Hirt H, Dunny GM, Berntsson RPA. Polymer Adhesin Domains in Gram-Positive Cell Surface Proteins. Front Microbiol 2020; 11:599899. [PMID: 33324381 PMCID: PMC7726212 DOI: 10.3389/fmicb.2020.599899] [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: 08/28/2020] [Accepted: 10/28/2020] [Indexed: 01/12/2023] Open
Abstract
Surface proteins in Gram-positive bacteria are often involved in biofilm formation, host-cell interactions, and surface attachment. Here we review a protein module found in surface proteins that are often encoded on various mobile genetic elements like conjugative plasmids. This module binds to different types of polymers like DNA, lipoteichoic acid and glucans, and is here termed polymer adhesin domain. We analyze all proteins that contain a polymer adhesin domain and classify the proteins into distinct classes based on phylogenetic and protein domain analysis. Protein function and ligand binding show class specificity, information that will be useful in determining the function of the large number of so far uncharacterized proteins containing a polymer adhesin domain.
Collapse
Affiliation(s)
- Michael A Järvå
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Helmut Hirt
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Gary M Dunny
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Ronnie P-A Berntsson
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden.,Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
9
|
Hammerschmidt S, Rohde M, Preissner KT. Extracellular Matrix Interactions with Gram-Positive Pathogens. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0041-2018. [PMID: 31004421 PMCID: PMC11590433 DOI: 10.1128/microbiolspec.gpp3-0041-2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Indexed: 01/10/2023] Open
Abstract
The main strategies used by pathogenic bacteria to infect eukaryotic tissue include their adherence to cells and the extracellular matrix (ECM), the subsequent colonization and invasion as well as the evasion of immune defences. A variety of structurally and functionally characterized adhesins and binding proteins of gram-positive bacteria facilitate these processes by specifically recognizing and interacting with various components of the host ECM, including different collagens, fibronectin and other macromolecules. The ECM affects the cellular physiology of our body and is critical for adhesion, migration, proliferation, and differentiation of many host cell types, but also provides the support for infiltrating pathogens, particularly under conditions of injury and trauma. Moreover, microbial binding to a variety of adhesive components in host tissue fluids leads to structural and/or functional alterations of host proteins and to the activation of cellular mechanisms that influence tissue and cell invasion of pathogens. Since the diverse interactions of gram-positive bacteria with the ECM represent important pathogenicity mechanisms, their characterization not only allows a better understanding of microbial invasion but also provides clues for the design of novel therapeutic strategies to manage infectious diseases.
Collapse
Affiliation(s)
- Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, University of Greifswald, D-17487 Greifswald, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz-Center for Infection Research, D-38124 Braunschweig, Germany
| | - Klaus T Preissner
- Institute for Biochemistry, Medical School, Justus-Liebig-University, D-35392 Giessen, Germany
| |
Collapse
|
10
|
Ortiz Franyuti D, Mitsi M, Vogel V. Mechanical Stretching of Fibronectin Fibers Upregulates Binding of Interleukin-7. NANO LETTERS 2018; 18:15-25. [PMID: 28845674 DOI: 10.1021/acs.nanolett.7b01617] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Since evidence is rising that extracellular matrix (ECM) fibers might serve as reservoirs for growth factors and cytokines, we investigated the interaction between fibronectin (FN) and interleukin-7 (IL-7), a cytokine of immunological significance and a target of several immunotherapies. By employing a FN fiber stretch assay and Förster resonance energy transfer (FRET) confocal microscopy, we found that stretching of FN fibers increased IL-7 binding. We localized the FN binding site on the CD loop of IL-7, since a synthetic CD loop peptide also bound stronger to stretched than to relaxed FN fibers. On the basis of a structural model, we propose that the CD loop can bind to FN, while IL-7 is bound to its cognate cell surface receptors. Sequence alignment with bacterial adhesins, which also bind the FN N-terminus, suggests that a conserved motif on the CD loop (110TKSLEEN116 and the truncated 112SLEE115 in human and mouse IL-7, respectively) might bind to the second FN type I module (FnI2) and that additional epitopes enhance the stretch-upregulated binding. FN fiber stretching might thus serve as a mechano-regulated mechanism to locally concentrate IL-7 in an ECM-bound state, thereby upregulating the potency of IL-7 signaling. A feedback model mechanism is proposed that could explain the well-known, but poorly understood, function of IL-7 in ECM homeostasis. Understanding how local IL-7 availability and signaling might be modulated by the tensional state of the ECM niche, which is adjusted by residing stroma cells, is highly relevant for basic science but also for advancing IL-7 based immunotherapies.
Collapse
Affiliation(s)
- Daniela Ortiz Franyuti
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department for Health Sciences and Technology (D-HEST), ETH Zurich , Vladimir-Prelog-Weg 4, HCI F443 CH-8093 Zürich, Switzerland
| | - Maria Mitsi
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department for Health Sciences and Technology (D-HEST), ETH Zurich , Vladimir-Prelog-Weg 4, HCI F443 CH-8093 Zürich, Switzerland
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department for Health Sciences and Technology (D-HEST), ETH Zurich , Vladimir-Prelog-Weg 4, HCI F443 CH-8093 Zürich, Switzerland
| |
Collapse
|
11
|
Multiple Cryptic Binding Sites are Necessary for Robust Fibronectin Assembly: An In Silico Study. Sci Rep 2017; 7:18061. [PMID: 29273802 PMCID: PMC5741729 DOI: 10.1038/s41598-017-18328-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/08/2017] [Indexed: 12/31/2022] Open
Abstract
The mechanism of assembly of the extracellular matrix protein fibronectin (FN) into elastic, insoluble fibrils is still poorly understood. FN fibrillogenesis requires cell-generated forces, which expose cryptic FN-FN binding sites buried in FN Type III domains. The number and location of cryptic binding sites have been debated, but experimental evidence suggests multiple domains may contain FN-FN binding sites. The requirement of cell-dependent forces to generate FN fibrils restricts investigation of the mechanism of assembly. To address this, we use a recently developed biophysical model of fibrillogenesis to test competing hypotheses for the location and number of cryptic FN-FN binding sites and quantify the effect of these molecular alterations on assembled FN fibril properties. Simulations predict that a single FN-FN binding site facilitates either negligible fibrillogenesis or produces FN fibrils that are neither robust nor physiological. However, inclusion of multiple FN-FN binding sites predicts robust fibrillogenesis, which minimally depends on individual domain properties. Multiple FN-FN binding site models predict a heterogeneous fibril population that contains two distinct phenotypes with unique viscoelastic properties, which we speculate may play a key role in generating heterogeneous mechanical signaling in the extracellular matrix of developing and regenerating tissues.
Collapse
|
12
|
Metasecretome Phage Display. Methods Mol Biol 2017. [PMID: 29116525 DOI: 10.1007/978-1-4939-7447-4_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Metasecretome is a collection of cell-surface and secreted proteins that mediate interactions between microbial communities and their environment. These include adhesins, enzymes, surface structures such as pili or flagella, vaccine targets or proteins responsible for immune evasion. Traditional approaches to exploring matasecretome of complex microbial communities via cultivation of microorganisms and screening of individual strains fail to sample extraordinary diversity in these communities, since only a limited fraction of microorganisms are represented by cultures. Advances in culture-independent sequence analysis methods, collectively referred to as metagenomics, offer an alternative approach that enables the direct analysis of collective microbial genomes (metagenome) recovered from environmental samples. This protocol describes a method, metasecretome phage display, which selectively displays the metasecretome portion of the metagenome. The metasecretome library can then be used for two purposes: (1) to sequence the entire metasecretome (using PacBio technology); (2) to identify metasecretome proteins that have a specific function of interest by affinity-screening (bio-panning) using a variety of methods described in other chapters of this volume.
Collapse
|
13
|
Murai M, Moriyama H, Hata E, Takeuchi F, Amemura-Maekawa J. Variation and association of fibronectin-binding protein genes fnbA and fnbB in Staphylococcus aureus Japanese isolates. Microbiol Immunol 2017; 60:312-25. [PMID: 26990092 DOI: 10.1111/1348-0421.12377] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 02/28/2016] [Accepted: 03/10/2016] [Indexed: 01/02/2023]
Abstract
Fibronectin-binding proteins A and B (FnBPA and FnBPB) mediate adhesion of Staphylococcus aureus to fibrinogen, elastin and fibronectin. FnBPA and FnBPB are encoded by two closely linked genes, fnbA and fnbB, respectively. With the exception of the N-terminal regions, the amino acid sequences of FnBPA and FnBPB are highly conserved. To investigate the genetics and evolution of fnbA and fnbB, the most variable regions, which code for the 67th amino acids of the A through B regions (A67-B) of fnbA and fnbB, were focused upon. Eighty isolates of S. aureus in Japan were sequenced and 19 and 18 types in fnbA and fnbB, respectively, identified. Although the phylogeny of fnbA and fnbB were found to be quite different, each fnbA type connected with a specific fnbB type, indicating that fnbA and fnbB mutate independently, whereas the combination of both genes after recombination is stable. Hence those fnbA-fnbB combinations were defined as FnBP sequence types (FnSTs). Representative isolates of each FnST were assigned distinct STs by multilocus sequence typing, suggesting correspondence of FnST with genome lineage. Linkage disequilibrium (LD) analysis of the A67-B region revealed that subdomains N2, N3 and FnBR1 form a LD block in fnbA, whereas N2 and N3 form two independent LD blocks in fnbB. N2-N3 three-dimensional structural models indicated that not only the variable amino acid residues, but also well-conserved amino acid residues between FnBPA and FnBPB, are located on the surface of the protein. These results highlight a molecular process of the FnBP that has evolved by mingled mutation and recombination with retention of functions.
Collapse
Affiliation(s)
- Miyo Murai
- Department of Health Sciences, Saitama Prefectural University,820, Sannomiya, Koshigaya-shi, Saitama 343-8540
| | - Hideaki Moriyama
- School of Biological Sciences, University of Nebraska-Lincoln, 243 Manter Hall, Lincoln, Nebraska 68588, USA
| | - Eiji Hata
- Dairy Hygiene Research Division, National Institute of Animal Health, 4 Hitsujigaoka, Toyohira-ku, Sapporo, Hokkaido 062-0045
| | - Fumihiko Takeuchi
- Pathogen Genomics Center, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8640
| | - Junko Amemura-Maekawa
- Department of Bacteriology I, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| |
Collapse
|
14
|
Foster TJ. The remarkably multifunctional fibronectin binding proteins of Staphylococcus aureus. Eur J Clin Microbiol Infect Dis 2016; 35:1923-1931. [PMID: 27604831 DOI: 10.1007/s10096-016-2763-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/16/2016] [Indexed: 12/01/2022]
Abstract
Staphylococcus aureus expresses two distinct but closely related multifunctional cell wall-anchored (CWA) proteins that bind to the host glycoprotein fibronectin. The fibronectin binding proteins FnBPA and FnBPB comprise two distinct domains. The C-terminal domain comprises a tandem array of repeats that bind to the N-terminal type I modules of fibronectin by the tandem β-zipper mechanism. This causes allosteric activation of a cryptic integrin binding domain, allowing fibronectin to act as a bridge between bacterial cells and the α5β1 integrin on host cells, triggering bacterial uptake by endocytosis. Variants of FnBPA with polymorphisms in fibronectin binding repeats (FnBRs) that increase affinity for the ligand are associated with strains that infect cardiac devices and cause endocarditis, suggesting that binding affinity is particularly important in intravascular infections. The N-terminal A domains of FnBPA and FnBPB have diverged into seven antigenically distinct isoforms. Each binds fibrinogen by the 'dock, lock and latch' mechanism characteristic of clumping factor A. However, FnBPs can also bind to elastin, which is probably important in adhesion to connective tissue in vivo. In addition, they can capture plasminogen from plasma, which can be activated to plasmin by host and bacterial plasminogen activators. The bacterial cells become armed with a host protease which destroys opsonins, contributing to immune evasion and promotes spreading during skin infection. Finally, some methicillin-resistant S. aureus (MRSA) strains form biofilm that depends on the elaboration of FnBPs rather than polysaccharide. The A domains of the FnBPs can interact homophilically, allowing cells to bind together as the biofilm accumulates.
Collapse
Affiliation(s)
- T J Foster
- Microbiology Department, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
15
|
Musyoki AM, Shi Z, Xuan C, Lu G, Qi J, Gao F, Zheng B, Zhang Q, Li Y, Haywood J, Liu C, Yan J, Shi Y, Gao GF. Structural and functional analysis of an anchorless fibronectin-binding protein FBPS from Gram-positive bacterium Streptococcus suis. Proc Natl Acad Sci U S A 2016; 113:13869-13874. [PMID: 27834729 PMCID: PMC5137682 DOI: 10.1073/pnas.1608406113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The anchorless fibronectin-binding proteins (FnBPs) are a group of important virulence factors for which the structures are not available and the functions are not well defined. In this study we performed comprehensive studies on a prototypic member of this group: the fibronectin-/fibrinogen-binding protein from Streptococcus suis (FBPS). The structures of the N- and C-terminal halves (FBPS-N and FBPS-C), which together cover the full-length protein in sequence, were solved at a resolution of 2.1 and 2.6 Å, respectively, and each was found to be composed of two domains with unique folds. Furthermore, we have elucidated the organization of these domains by small-angle X-ray scattering. We further showed that the fibronectin-binding site is located in FBPS-C and that FBPS promotes the adherence of S suis to host cells by attaching the bacteria via FBPS-N. Finally, we demonstrated that FBPS functions both as an adhesin, promoting S suis attachment to host cells, and as a bacterial factor, activating signaling pathways via β1 integrin receptors to induce chemokine production.
Collapse
Affiliation(s)
- Abednego Moki Musyoki
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongyu Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chunling Xuan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Guangwen Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China
- Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Gao
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Beiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qiangmin Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Joel Haywood
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Cuihua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jinghua Yan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
- Research Network of Immunity and Health, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China;
- University of the Chinese Academy of Sciences, Beijing 100049, China
- Research Network of Immunity and Health, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
16
|
Abstract
Fibronectin is a large vertebrate glycoprotein that is found in soluble and insoluble forms and involved in diverse processes. Protomeric fibronectin is a dimer of subunits, each of which comprises 29-31 modules - 12 type I, two type II and 15-17 type III. Plasma fibronectin is secreted by hepatocytes and circulates in a compact conformation before it binds to cell surfaces, converts to an extended conformation and is assembled into fibronectin fibrils. Here we review biophysical and structural studies that have shed light on how plasma fibronectin transitions from the compact to the extended conformation. The three types of modules each have a well-organized secondary and tertiary structure as defined by NMR and crystallography and have been likened to "beads on a string". There are flexible sequences in the N-terminal tail, between the fifth and sixth type I modules, between the first two and last two of the type III modules, and at the C-terminus. Several specific module-module interactions have been identified that likely maintain the compact quaternary structure of circulating fibronectin. The quaternary structure is perturbed in response to binding events, including binding of fibronectin to the surface of vertebrate cells for fibril assembly and to bacterial adhesins.
Collapse
Affiliation(s)
- Lisa M Maurer
- a Departments of Biomolecular Chemistry and Medicine , University of Wisconsin-Madison , Madison , WI , United States
| | - Wenjiang Ma
- a Departments of Biomolecular Chemistry and Medicine , University of Wisconsin-Madison , Madison , WI , United States
| | - Deane F Mosher
- a Departments of Biomolecular Chemistry and Medicine , University of Wisconsin-Madison , Madison , WI , United States
| |
Collapse
|
17
|
Xu X, Steffensen B, Robichaud TK, Mikhailova M, Lai V, Montgomery R, Chu L. Fibronectin-binding protein TDE1579 affects cytotoxicity of Treponema denticola. Anaerobe 2015; 36:39-48. [PMID: 26456217 DOI: 10.1016/j.anaerobe.2015.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/16/2015] [Accepted: 09/29/2015] [Indexed: 11/25/2022]
Abstract
While FbpA, a family of bacterial fibronectin (FN) binding proteins has been studied in several gram-positive bacteria, the gram-negative Treponema denticola, an anaerobic periodontal pathogen, also has an overlooked fbp gene (tde1579). In this research, we confirm that recombinant Fbp protein (rFbp) of T. denticola binds human FN with a Kdapp of 1.5 × 10(-7) M and blocks the binding of T. denticola to FN in a concentration-dependent manner to a level of 42%. The fbp gene was expressed in T. denticola. To reveal the roles of fbp in T. denticola pathogenesis, an fbp isogenic mutant was constructed. The fbp mutant had 51% reduced binding ability to human gingival fibroblasts (hGF). When hGF were challenged with T. denticola, the fbp mutant caused less cell morphology change, had 50% reduced cytotoxicity to hGF, and had less influence on the growth of hGF cells.
Collapse
Affiliation(s)
- Xiaoping Xu
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| | - Bjorn Steffensen
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Periodontology, Tufts University School of Dental Medicine, Boston, MD, 02111, USA
| | - Trista K Robichaud
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Margarita Mikhailova
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Veronica Lai
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Ryan Montgomery
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Lianrui Chu
- Department of Developmental Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| |
Collapse
|
18
|
Nishiyama K, Nakazato A, Ueno S, Seto Y, Kakuda T, Takai S, Yamamoto Y, Mukai T. Cell surface-associated aggregation-promoting factor fromLactobacillus gasseri SBT2055 facilitates host colonization and competitive exclusion ofCampylobacter jejuni. Mol Microbiol 2015; 98:712-26. [DOI: 10.1111/mmi.13153] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2015] [Indexed: 02/04/2023]
Affiliation(s)
- Keita Nishiyama
- Department of Animal Science; School of Veterinary Medicine; Kitasato University; Towada Aomori 034-8628 Japan
| | - Akiko Nakazato
- Department of Animal Science; School of Veterinary Medicine; Kitasato University; Towada Aomori 034-8628 Japan
| | - Shintaro Ueno
- Department of Animal Science; School of Veterinary Medicine; Kitasato University; Towada Aomori 034-8628 Japan
| | - Yasuyuki Seto
- Milk Science Research Institute; Megmilk Snow Brand Co. Ltd.; Kawagoe Saitama 350-1165 Japan
| | - Tsutomu Kakuda
- Faculty of Veterinary Medicine; School of Veterinary Medicine; Kitasato University; Towada Aomori 034-8628 Japan
| | - Shinji Takai
- Faculty of Veterinary Medicine; School of Veterinary Medicine; Kitasato University; Towada Aomori 034-8628 Japan
| | - Yuji Yamamoto
- Department of Animal Science; School of Veterinary Medicine; Kitasato University; Towada Aomori 034-8628 Japan
| | - Takao Mukai
- Department of Animal Science; School of Veterinary Medicine; Kitasato University; Towada Aomori 034-8628 Japan
| |
Collapse
|
19
|
Katayama S, Tagomori M, Morita N, Yamasaki T, Nariya H, Okada M, Watanabe M, Hitsumoto Y. Determination of the Clostridium perfringens-binding site on fibronectin. Anaerobe 2015; 34:174-81. [DOI: 10.1016/j.anaerobe.2014.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 10/23/2014] [Accepted: 11/12/2014] [Indexed: 10/24/2022]
|
20
|
Walden M, Edwards JM, Dziewulska AM, Bergmann R, Saalbach G, Kan SY, Miller OK, Weckener M, Jackson RJ, Shirran SL, Botting CH, Florence GJ, Rohde M, Banfield MJ, Schwarz-Linek U. An internal thioester in a pathogen surface protein mediates covalent host binding. eLife 2015; 4. [PMID: 26032562 PMCID: PMC4450167 DOI: 10.7554/elife.06638] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/01/2015] [Indexed: 01/19/2023] Open
Abstract
To cause disease and persist in a host, pathogenic and commensal microbes must adhere to tissues. Colonization and infection depend on specific molecular interactions at the host-microbe interface that involve microbial surface proteins, or adhesins. To date, adhesins are only known to bind to host receptors non-covalently. Here we show that the streptococcal surface protein SfbI mediates covalent interaction with the host protein fibrinogen using an unusual internal thioester bond as a ‘chemical harpoon’. This cross-linking reaction allows bacterial attachment to fibrin and SfbI binding to human cells in a model of inflammation. Thioester-containing domains are unexpectedly prevalent in Gram-positive bacteria, including many clinically relevant pathogens. Our findings support bacterial-encoded covalent binding as a new molecular principle in host-microbe interactions. This represents an as yet unexploited target to treat bacterial infection and may also offer novel opportunities for engineering beneficial interactions. DOI:http://dx.doi.org/10.7554/eLife.06638.001 The human body is home to many trillions of microbes; most are harmless, but some may cause disease. To live inside a host, microbes must first attach to host tissues. This process involves multiple proteins on each microbe's surface, called adhesins, which interact with the molecules that make up these tissues. Like all proteins, adhesins are long chains of simpler building blocks called amino acids, and each amino acid is connected to the next via a strong ‘covalent’ bond. Adhesins, however, typically attach bacteria to host molecules through the combined strength of many weak ‘non-covalent’ interactions. It was recently discovered that one adhesin from a bacterium called Streptococcus pyogenes contains a rare, extra covalent bond—called a thioester—in an unusual location between two of its amino acids. S. pyogenes is a common cause of throat infections in humans, and can also cause the life-threatening ‘flesh-eating disease’. Walden, Edwards et al. have now used a range of computational, biochemical, structural biology and cell-based techniques to study other adhesins that have thioester bonds in more detail. Computational searches identified hundreds of bacterial proteins containing similar bonds. These included many from bacteria that infect humans: such as Streptococcus pneumoniae, which is the most common cause of pneumonia in adults; and Clostridium difficile, which is notorious for causing severe gut infections in hospital patients. Closer examination of the three-dimensional structures of three of these proteins—including one called SfbI from S. pyogenes—revealed that each had a clear thioester bond. Biochemical tests of an additional nine of the identified proteins strongly suggested they too contained thioester bonds. Walden, Edwards et al. then showed that SfbI was able to not only attach to tissues like conventional adhesins, but also chemically react with fibrinogen: a human protein that is essential for blood clotting and commonly found in inflamed tissues and healing wounds. This chemical reaction results in the formation of a covalent bond between SfbI and fibrinogen, which is as stable as the bonds that link the amino acids in a protein chain. Further experiments revealed that SfbI strongly binds to human cells grown in the lab under conditions that mimic tissue inflammation. Finally, Walden, Edwards et al. made a mutant version of SfbI that did not contain a thioester, and found that it could not interact with fibrinogen nor bind to human cells. Together, these findings suggest that thioesters in bacterial adhesins act like ‘chemical harpoons’, which microbes can use to irreversibly attach themselves to molecules within their host's tissues. This attachment mechanism has not been seen before in host-microbe interactions, and further research is now needed to explore whether interfering with this process could represent a new way to treat bacterial infections. DOI:http://dx.doi.org/10.7554/eLife.06638.002
Collapse
Affiliation(s)
- Miriam Walden
- Department of Biological Chemistry, John Innes Centre, Norwich, United Kingdom
| | - John M Edwards
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | | | - Rene Bergmann
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Gerhard Saalbach
- Department of Biological Chemistry, John Innes Centre, Norwich, United Kingdom
| | - Su-Yin Kan
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Ona K Miller
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Miriam Weckener
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Rosemary J Jackson
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Sally L Shirran
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Catherine H Botting
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Gordon J Florence
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Mark J Banfield
- Department of Biological Chemistry, John Innes Centre, Norwich, United Kingdom
| | - Ulrich Schwarz-Linek
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
21
|
Ma W, Ma H, Mosher DF. On-Off Kinetics of Engagement of FNI Modules of Soluble Fibronectin by β-Strand Addition. PLoS One 2015; 10:e0124941. [PMID: 25919138 PMCID: PMC4412574 DOI: 10.1371/journal.pone.0124941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/20/2015] [Indexed: 02/07/2023] Open
Abstract
Intrinsically disordered sequences within bacterial adhesins bind to E-strands in the β-sheets of multiple FNI modules of fibronectin (FN) by anti-parallel β-strand addition, also called tandem β-zipper formation. The FUD segment of SfbI of Streptococcus pyogenes and Bbk32 segment of BBK32 of Borrelia burgdorferi, despite being imbedded in different adhesins from different bacteria, target the same 2-5,8-9 FNI modules, 2-5,8-9 FNI, in the N-terminal 70-kDa region (FN70K) of FN. To facilitate further comparisons, FUD, Bbk32, two other polypeptides based on SfbI that target 1-5 FNI (HADD) and 2-5 FNI (FRD), and mutant Bbk32 (ΔBbk32) were produced with fluorochromes placed just outside of the binding sequences. Unlabeled FUD competed ~ 1000-fold better for binding of labeled Bbk32 to FN than unlabeled Bbk32 competed for binding of labeled FUD to FN. Binding kinetics were determined by fluorescence polarization in a stopped-flow apparatus. On-rates for FUD, Bbk32, HADD, and FRD were similar, and all bound more rapidly to FN70K fragment than to full length FN. In stopped-flow displacement and size exclusion chromatographic assays, however, k off for FUD or HADD to FN70K or FN was considerably lower compared to k off of FRD or Bbk32. FUD and Bbk32 differ in the spacing between sequences that interact with 3FNI and 4FNI or with 5FNI and 8FNI. ΔBbk32, in which 2 residues were removed from Bbk32 to make the spacing more like FUD, had a k off intermediate between that of Bbk32 and FUD. These results indicate a "folding-after-binding" process after initial association of certain polypeptide sequences to FN that results in formation of a stable complex and is a function of number of FNI modules engaged by the polypeptide, spacing of engagement sites, and perhaps flexibility within the polypeptide-FN complex. We suggest that contributions of SfbI and BBK32 adhesins to bacterial pathogenicity may be determined in part by stability of adhesin-FN complexes.
Collapse
Affiliation(s)
- Wenjiang Ma
- Departments of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Hanqing Ma
- Departments of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Deane F. Mosher
- Departments of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
22
|
Sattar S, Bennett NJ, Wen WX, Guthrie JM, Blackwell LF, Conway JF, Rakonjac J. Ff-nano, short functionalized nanorods derived from Ff (f1, fd, or M13) filamentous bacteriophage. Front Microbiol 2015; 6:316. [PMID: 25941520 PMCID: PMC4403547 DOI: 10.3389/fmicb.2015.00316] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/30/2015] [Indexed: 11/22/2022] Open
Abstract
F-specific filamentous phage of Escherichia coli (Ff: f1, M13, or fd) are long thin filaments (860 nm × 6 nm). They have been a major workhorse in display technologies and bionanotechnology; however, some applications are limited by the high length-to-diameter ratio of Ff. Furthermore, use of functionalized Ff outside of laboratory containment is in part hampered by the fact that they are genetically modified viruses. We have now developed a system for production and purification of very short functionalized Ff-phage-derived nanorods, named Ff-nano, that are only 50 nm in length. In contrast to standard Ff-derived vectors that replicate in E. coli and contain antibiotic-resistance genes, Ff-nano are protein-DNA complexes that cannot replicate on their own and do not contain any coding sequences. These nanorods show an increased resistance to heating at 70∘C in 1% SDS in comparison to the full-length Ff phage of the same coat composition. We demonstrate that functionalized Ff-nano particles are suitable for application as detection particles in sensitive and quantitative “dipstick” lateral flow diagnostic assay for human plasma fibronectin.
Collapse
Affiliation(s)
- Sadia Sattar
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand
| | - Nicholas J Bennett
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand
| | - Wesley X Wen
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand
| | - Jenness M Guthrie
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand ; Science Haven Limited, Palmerston North New Zealand
| | - Len F Blackwell
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand ; Science Haven Limited, Palmerston North New Zealand
| | - James F Conway
- University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Jasna Rakonjac
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand
| |
Collapse
|
23
|
Ke W, Molini BJ, Lukehart SA, Giacani L. Treponema pallidum subsp. pallidum TP0136 protein is heterogeneous among isolates and binds cellular and plasma fibronectin via its NH2-terminal end. PLoS Negl Trop Dis 2015; 9:e0003662. [PMID: 25793702 PMCID: PMC4368718 DOI: 10.1371/journal.pntd.0003662] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/28/2015] [Indexed: 11/19/2022] Open
Abstract
Adherence-mediated colonization plays an important role in pathogenesis of microbial infections, particularly those caused by extracellular pathogens responsible for systemic diseases, such as Treponema pallidum subsp. pallidum (T. pallidum), the agent of syphilis. Among T. pallidum adhesins, TP0136 is known to bind fibronectin (Fn), an important constituent of the host extracellular matrix. To deepen our understanding of the TP0136-Fn interaction dynamics, we used two naturally-occurring sequence variants of the TP0136 protein to investigate which region of the protein is responsible for Fn binding, and whether TP0136 would adhere to human cellular Fn in addition to plasma Fn and super Fn as previously reported. Fn binding assays were performed with recombinant proteins representing the two full-length TP0136 variants and their discrete regions. As a complementary approach, we tested inhibition of T. pallidum binding to Fn by recombinant full-length TP0136 proteins and fragments, as well as by anti-TP0136 immune sera. Our results show that TP0136 adheres more efficiently to cellular Fn than to plasma Fn, that the TP0136 NH2-terminal conserved region of the protein is primarily responsible for binding to plasma Fn but that binding sites for cellular Fn are also present in the protein’s central and COOH-terminal regions. Additionally, message quantification studies show that tp0136 is highly transcribed during experimental infection, and that its message level increases in parallel to the host immune pressure on the pathogen, which suggests a possible role for this protein in T. pallidum persistence. In a time where syphilis incidence is high, our data will help in the quest to identify suitable targets for development of a much needed vaccine against this important disease. The study of Treponema pallidum subsp. pallidum (T. pallidum) proteins that mediate adhesion to host tissue components is pivotal to understand how the syphilis agent establishes infection and is able to invade virtually every organ system following dissemination from the site of entry. This study focuses on T. pallidum TP0136, a known plasma fibronectin (Fn) and super Fn binding protein that is heterogeneous in sequence among T. pallidum isolates. This study shows that TP0136 also mediates attachment to human cellular Fn, that TP0136 conserved NH2-terminus is primarily responsible for binding to plasma Fn, but that cellular Fn binding sites appears to be scattered throughout the molecule. Message quantification experiments reveal that tp0136 transcription is high during experimental syphilis and increases at the time of bacterial immune clearance, suggesting a role for this antigen in counteracting the host defenses during infection, as reported for other Fn binding proteins in other pathogens. Our data deepen the current knowledge of the function of T. pallidum TP0136 and further support a role for this virulence factor in syphilis pathogenesis.
Collapse
Affiliation(s)
- Wujian Ke
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Harborview Medical Center, Seattle, Washington, United States of America
- Graduate School, Southern Medical University, Guangzhou, PR China
- Division of STD, Guangdong Provincial Center for STI & Skin Diseases Control and Prevention, Guangzhou, PR China
| | - Barbara J. Molini
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Harborview Medical Center, Seattle, Washington, United States of America
| | - Sheila A. Lukehart
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Harborview Medical Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Lorenzo Giacani
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Harborview Medical Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
24
|
Galloway-Peña JR, Liang X, Singh KV, Yadav P, Chang C, La Rosa SL, Shelburne S, Ton-That H, Höök M, Murray BE. The identification and functional characterization of WxL proteins from Enterococcus faecium reveal surface proteins involved in extracellular matrix interactions. J Bacteriol 2015; 197:882-92. [PMID: 25512313 PMCID: PMC4325096 DOI: 10.1128/jb.02288-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/10/2014] [Indexed: 11/20/2022] Open
Abstract
The WxL domain recently has been identified as a novel cell wall binding domain found in numerous predicted proteins within multiple Gram-positive bacterial species. However, little is known about the function of proteins containing this novel domain. Here, we identify and characterize 6 Enterococcus faecium proteins containing the WxL domain which, by reverse transcription-PCR (RT-PCR) and genomic analyses, are located in three similarly organized operons, deemed WxL loci A, B, and C. Western blotting, electron microscopy, and enzyme-linked immunosorbent assays (ELISAs) determined that genes of WxL loci A and C encode antigenic, cell surface proteins exposed at higher levels in clinical isolates than in commensal isolates. Secondary structural analyses of locus A recombinant WxL domain-containing proteins found they are rich in β-sheet structure and disordered segments. Using Biacore analyses, we discovered that recombinant WxL proteins from locus A bind human extracellular matrix proteins, specifically type I collagen and fibronectin. Proteins encoded by locus A also were found to bind to each other, suggesting a novel cell surface complex. Furthermore, bile salt survival assays and animal models using a mutant from which all three WxL loci were deleted revealed the involvement of WxL operons in bile salt stress and endocarditis pathogenesis. In summary, these studies extend our understanding of proteins containing the WxL domain and their potential impact on colonization and virulence in E. faecium and possibly other Gram-positive bacterial species.
Collapse
Affiliation(s)
- Jessica R Galloway-Peña
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, Texas, USA Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas, USA Center for the Study of Emerging and Re-emerging Pathogens, University of Texas Health Science Center, Houston, Texas, USA
| | - Xiaowen Liang
- Center for Infectious and Inflammatory Diseases, Institute for Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA
| | - Kavindra V Singh
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas, USA Center for the Study of Emerging and Re-emerging Pathogens, University of Texas Health Science Center, Houston, Texas, USA
| | - Puja Yadav
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, Texas, USA Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas, USA Center for the Study of Emerging and Re-emerging Pathogens, University of Texas Health Science Center, Houston, Texas, USA
| | - Chungyu Chang
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, Texas, USA
| | - Sabina Leanti La Rosa
- Department of Chemistry, Biotechnology and Food Science, Laboratory of Microbial Gene Technology and Food Microbiology, The Norwegian University of Life Sciences, Aas, Norway
| | - Samuel Shelburne
- Department of Infectious Diseases, Infection Control and Employee Health, M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Hung Ton-That
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, Texas, USA
| | - Magnus Höök
- Center for Infectious and Inflammatory Diseases, Institute for Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA
| | - Barbara E Murray
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, Houston, Texas, USA Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas, USA Center for the Study of Emerging and Re-emerging Pathogens, University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
25
|
Antunes CA, Sanches dos Santos L, Hacker E, Köhler S, Bösl K, Ott L, de Luna MDG, Hirata R, Azevedo VADC, Mattos-Guaraldi AL, Burkovski A. Characterization of DIP0733, a multi-functional virulence factor of Corynebacterium diphtheriae. MICROBIOLOGY-SGM 2015; 161:639-47. [PMID: 25635272 DOI: 10.1099/mic.0.000020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Corynebacterium diphtheriae is typically recognized as an extracellular pathogen. However, a number of studies revealed its ability to invade epithelial cells, indicating a more complex pathogen-host interaction. The molecular mechanisms controlling and facilitating internalization of Cor. diphtheriae are poorly understood. In this study, we investigated the role of DIP0733 as virulence factor to elucidate how it contributes to the process of pathogen-host cell interaction. Based on in vitro experiments, it was suggested recently that the DIP0733 protein might be involved in adhesion, invasion of epithelial cells and induction of apoptosis. A corresponding Cor. diphtheriae mutant strain generated in this study was attenuated in its ability to colonize and kill the host in a Caenorhabditis elegans infection model system. Furthermore, the mutant showed an altered adhesion pattern and a drastically reduced ability to adhere and invade epithelial cells. Subsequent experiments showed an influence of DIP0733 on binding of Cor. diphtheriae to extracellular matrix proteins such as collagen and fibronectin. Furthermore, based on its fibrinogen-binding activity, DIP0733 may play a role in avoiding recognition of Cor. diphtheriae by the immune system. In summary, our findings support the idea that DIP0733 is a multi-functional virulence factor of Cor. diphtheriae.
Collapse
Affiliation(s)
- Camila Azevedo Antunes
- Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany Departmento de Biologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Elena Hacker
- Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stefanie Köhler
- Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Korbinian Bösl
- Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Ott
- Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Raphael Hirata
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vasco Ariston de Carvalho Azevedo
- Departmento de Biologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Andreas Burkovski
- Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
26
|
Brittan JL, Nobbs AH. Group B Streptococcus pili mediate adherence to salivary glycoproteins. Microbes Infect 2015; 17:360-8. [PMID: 25576026 DOI: 10.1016/j.micinf.2014.12.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/23/2014] [Accepted: 12/30/2014] [Indexed: 12/23/2022]
Abstract
Group B Streptococcus (GBS) is a leading cause of neonatal sepsis, pneumonia and meningitis, and is responsible for a rising number of severe invasive infections in adults. For all disease manifestations, colonisation is a critical first step. GBS has frequently been isolated from the oropharynx of neonates and adults. However, little is understood about the mechanisms of GBS colonisation at this site. In this study it is shown that three GBS strains (COH1, NEM316, 515) have capacity to adhere to human salivary pellicle. Heterologous expression of GBS pilus island (PI) genes in Lactococcus lactis to form surface-expressed pili demonstrated that GBS PI-2a and PI-1 pili bound glycoprotein-340 (gp340), a component of salivary pellicle. By contrast, PI-2b pili did not interact with gp340. The variation was attributable to differences in capacities for backbone and ancillary protein subunits of each pilus to bind gp340. Furthermore, while GBS strains were aggregated by fluid-phase gp340, this mechanism was not mediated by pili, which displayed specificity for immobilised gp340. Thus pili may enable GBS to colonise the soft and hard tissues of the oropharynx, while evading an innate mucosal defence, with implications for risk of progression to severe diseases such as meningitis and sepsis.
Collapse
Affiliation(s)
- Jane L Brittan
- School of Oral & Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol, BS1 2LY, UK
| | - Angela H Nobbs
- School of Oral & Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol, BS1 2LY, UK.
| |
Collapse
|
27
|
Cell Wall-Anchored Surface Proteins of Staphylococcus aureus: Many Proteins, Multiple Functions. Curr Top Microbiol Immunol 2015; 409:95-120. [PMID: 26667044 DOI: 10.1007/82_2015_5002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Staphylococcus aureus persistently colonizes about 20 % of the population and is intermittently associated with the remainder. The organism can cause superficial skin infections and life-threatening invasive diseases. The surface of the bacterial cell displays a variety of proteins that are covalently anchored to peptidoglycan. They perform many functions including adhesion to host cells and tissues, invasion of non-phagocytic cells, and evasion of innate immune responses. The proteins have been categorized into distinct classes based on structural and functional analysis. Many surface proteins are multifunctional. Cell wall-anchored proteins perform essential functions supporting survival and proliferation during the commensal state and during invasive infections. The ability of cell wall-anchored proteins to bind to desquamated epithelial cells is important during colonization, and the binding to fibrinogen is of particular significance in pathogenesis.
Collapse
|
28
|
Ma W, Ma H, Fogerty FJ, Mosher DF. Bivalent ligation of the collagen-binding modules of fibronectin by SFS, a non-anchored bacterial protein of Streptococcus equi. J Biol Chem 2014; 290:4866-4876. [PMID: 25525266 DOI: 10.1074/jbc.m114.612259] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SFS is a non-anchored protein of Streptococcus equi subspecies equi that causes upper respiratory infection in horses. SFS has been shown to bind to fibronectin (FN) and block interaction of FN with type I collagen. We have characterized interactions of a recombinant 60-mer polypeptide, R1R2, with FN. R1R2 contains two copies of collagen-like 19-residue repeats. Experiments utilizing various FN fragments and epitope-mapped anti-FN monoclonal antibodies located the binding site to (8-9)FNI modules of the gelatin-binding domain. Fluorescence polarization and competitive enzyme-linked assays demonstrated that R1R2 binds preferentially to compact dimeric FN rather than monomeric constructs containing (8-9)FNI or a large dimeric FN construct that is constitutively in an extended conformation. In contrast to bacterial peptides that bind (2-5)FNI in addition to (8-9)FNI, R1R2 did not cause conformational extension of FN as assessed by a conformationally sensitive antibody. Equilibrium and stopped-flow binding assays and size exclusion chromatography were compatible with a two-step binding reaction in which each of the repeats of R1R2 interacts with one of the subunits of dimeric FN, resulting in a stable complex with a slow koff. In addition to not binding to type I collagen, the R1R2·FN complex incorporated less efficiently into extracellular matrix than free FN. Thus, R1R2 binds to FN utilizing features of compact soluble FN and in doing so interferes with the organization of the extracellular matrix. A similar bivalent binding strategy may underlie the collagen-FN interaction.
Collapse
Affiliation(s)
- Wenjiang Ma
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | - Hanqing Ma
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | - Frances J Fogerty
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | - Deane F Mosher
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, Wisconsin 53706.
| |
Collapse
|
29
|
Ahmad F, Emrizal, Sirat HM, Jamaludin F, Mustapha NM, Ali RM, Arbain D, Aboul-Enein HY. Antimicrobial and anti-inflammatory activities of Piper porphyrophyllum (Fam. Piperaceae). ARAB J CHEM 2014. [DOI: 10.1016/j.arabjc.2010.12.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
30
|
Ballhausen B, Jung P, Kriegeskorte A, Makgotlho PE, Ruffing U, von Müller L, Köck R, Peters G, Herrmann M, Ziebuhr W, Becker K, Bischoff M. LA-MRSA CC398 differ from classical community acquired-MRSA and hospital acquired-MRSA lineages: functional analysis of infection and colonization processes. Int J Med Microbiol 2014; 304:777-86. [PMID: 25034858 DOI: 10.1016/j.ijmm.2014.06.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) of the clonal complex (CC) 398 became primarily known as colonizers of livestock animals. In the past few years, they have been increasingly introduced into hospitals with subsequent emergence of human infections. However, the (re-)adaptation to the human host is only incompletely understood. This study aimed to assess virulence properties of LA-MRSA CC398 by functional modeling of infection and colonization processes. A selection of 15 human LA-MRSA CC398 isolates and 11 pig-colonizing isolates were characterized regarding their virulence capacities and compared with human isolates of hospital-acquired (HA)-MRSA (CC5, CC22 and CC45) and community-associated (CA)-MRSA (CC8, CC30 and CC80) clonal lineages. Our investigations demonstrated that LA-MRSA CC398 adhered less efficient to human cells and human/bovine plasma fibronectin than CA-MRSA and HA-MRSA isolates. In contrast, the LA-MRSA CC398 isolates revealed a high cytotoxic potential comparable to certain CA-MRSA. Comparing the most prevalent LA-MRSA CC398 spa types (t011, t034, t108), isolates associated with spa t108 showed an increased adhesive and invasive potential paired with an increased ability to evade phagocytosis. The results underline both the pathogenic potential of LA-MRSA in general and the heterogeneity within the CC398 clade regarding the virulence characteristics of CC398 subpopulations. Assuming an ongoing (re-)adaptation to the human host combined with a huge reservoir of LA-MRSA CC398 in livestock and constant zoonotic transmission, the LA-MRSA CC398 lineage has the potential to pose a serious threat to human health.
Collapse
Affiliation(s)
- Britta Ballhausen
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Philipp Jung
- Institute of Medical Microbiology and Hygiene, University of Saarland Hospital, Homburg/Saar, Germany
| | - André Kriegeskorte
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | | | - Ulla Ruffing
- Institute of Medical Microbiology and Hygiene, University of Saarland Hospital, Homburg/Saar, Germany
| | - Lutz von Müller
- Institute of Medical Microbiology and Hygiene, University of Saarland Hospital, Homburg/Saar, Germany
| | - Robin Köck
- Institute of Hygiene, University Hospital of Münster, Münster, Germany
| | - Georg Peters
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Mathias Herrmann
- Institute of Medical Microbiology and Hygiene, University of Saarland Hospital, Homburg/Saar, Germany
| | - Wilma Ziebuhr
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Karsten Becker
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany.
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene, University of Saarland Hospital, Homburg/Saar, Germany
| |
Collapse
|
31
|
Harris G, Ma W, Maurer LM, Potts JR, Mosher DF. Borrelia burgdorferi protein BBK32 binds to soluble fibronectin via the N-terminal 70-kDa region, causing fibronectin to undergo conformational extension. J Biol Chem 2014; 289:22490-9. [PMID: 24962582 DOI: 10.1074/jbc.m114.578419] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BBK32 is a fibronectin (FN)-binding protein expressed on the cell surface of Borrelia burgdorferi, the causative agent of Lyme disease. There is conflicting information about where and how BBK32 interacts with FN. We have characterized interactions of a recombinant 86-mer polypeptide, "Bbk32," comprising the unstructured FN-binding region of BBK32. Competitive enzyme-linked assays utilizing various FN fragments and epitope-mapped anti-FN monoclonal antibodies showed that Bbk32 binding involves both the fibrin-binding and the gelatin-binding domains of the 70-kDa N-terminal region (FN70K). Crystallographic and NMR analyses of smaller Bbk32 peptides complexed, respectively, with (2-3)FNI and (8-9)FNI, demonstrated that binding occurs by β-strand addition. Isothermal titration calorimetry indicated that Bbk32 binds to isolated FN70K more tightly than to intact FN. In a competitive enzyme-linked binding assay, complex formation with Bbk32 enhanced binding of FN with mAbIII-10 to the (10)FNIII module. Thus, Bbk32 binds to multiple FN type 1 modules of the FN70K region by a tandem β-zipper mechanism, and in doing so increases accessibility of FNIII modules that interact with other ligands. The similarity in the FN-binding mechanism of BBK32 and previously studied streptococcal proteins suggests that the binding and associated conformational change of FN play a role in infection.
Collapse
Affiliation(s)
- Gemma Harris
- From the Department of Biology, University of York, York YO10 5DD, United Kingdom and
| | - Wenjiang Ma
- the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | - Lisa M Maurer
- the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | - Jennifer R Potts
- From the Department of Biology, University of York, York YO10 5DD, United Kingdom and
| | - Deane F Mosher
- the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
32
|
Muszanska AK, Rochford ETJ, Gruszka A, Bastian AA, Busscher HJ, Norde W, van der Mei HC, Herrmann A. Antiadhesive polymer brush coating functionalized with antimicrobial and RGD peptides to reduce biofilm formation and enhance tissue integration. Biomacromolecules 2014; 15:2019-26. [PMID: 24833130 DOI: 10.1021/bm500168s] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This paper describes the synthesis and characterization of polymer-peptide conjugates to be used as infection-resistant coating for biomaterial implants and devices. Antiadhesive polymer brushes composed of block copolymer Pluronic F-127 (PF127) were functionalized with antimicrobial peptides (AMP), able to kill bacteria on contact, and arginine-glycine-aspartate (RGD) peptides to promote the adhesion and spreading of host tissue cells. The antiadhesive and antibacterial properties of the coating were investigated with three bacterial strains: Staphylococcus aureus, Staphylococcus epidermidis, and Pseudomonas aeruginosa. The ability of the coating to support mammalian cell growth was determined using human fibroblast cells. Coatings composed of the appropriate ratio of the functional components: PF127, PF127 modified with AMP, and PF127 modified with RGD showed good antiadhesive and bactericidal properties without hampering tissue compatibility.
Collapse
Affiliation(s)
- Agnieszka K Muszanska
- University of Groningen and University Medical Center Groningen , Department of Biomedical Engineering, P.O. Box 196, 9700 AD Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Brissette CA, Gaultney RA. That's my story, and I'm sticking to it--an update on B. burgdorferi adhesins. Front Cell Infect Microbiol 2014; 4:41. [PMID: 24772392 PMCID: PMC3982108 DOI: 10.3389/fcimb.2014.00041] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/18/2014] [Indexed: 11/25/2022] Open
Abstract
Adhesion is the initial event in the establishment of any infection. Borrelia burgdorferi, the etiological agent of Lyme disease, possesses myriad proteins termed adhesins that facilitate contact with its vertebrate hosts. B. burgdorferi adheres to host tissues through interactions with host cells and extracellular matrix, as well as other molecules present in serum and extracellular fluids. These interactions, both general and specific, are critical in the establishment of infection. Modulation of borrelial adhesion to host tissues affects the microorganisms's ability to colonize, disseminate, and persist. In this review, we update the current knowledge on structure, function, and role in pathogenesis of these “sticky” B. burgdorferi infection-associated proteins.
Collapse
Affiliation(s)
- Catherine A Brissette
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences Grand Forks, ND, USA
| | - Robert A Gaultney
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences Grand Forks, ND, USA
| |
Collapse
|
34
|
Fibronectin-binding protein of Borrelia hermsii expressed in the blood of mice with relapsing fever. Infect Immun 2014; 82:2520-31. [PMID: 24686059 DOI: 10.1128/iai.01582-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
To identify and characterize surface proteins expressed by the relapsing fever (RF) agent Borrelia hermsii in the blood of infected mice, we used a cell-free filtrate of their blood to immunize congenic naive mice. The resultant antiserum was used for Western blotting of cell lysates, and gel slices corresponding to reactive bands were subjected to liquid chromatography-tandem mass spectrometry, followed by a search of the proteome database with the peptides. One of the immunogens was identified as the BHA007 protein, which is encoded by a 174-kb linear plasmid. BHA007 had sequence features of lipoproteins, was surface exposed by the criteria of in situ protease susceptibility and agglutination of Vtp(-) cells by anti-BHA007 antibodies, and was not essential for in vitro growth. BHA007 elicited antibodies during experimental infection of mice, but immunization with recombinant protein did not confer protection against needle-delivered infection. Open reading frames (ORFs) orthologous to BHA007 were found on large plasmids of other RF species, including the coding sequences for the CihC proteins of Borrelia duttonii and B. recurrentis, but not in Lyme disease Borrelia species. Recombinant BHA007 bound both human and bovine fibronectin with Kd (dissociation constant) values of 22 and 33 nM, respectively, and bound to C4-binding protein with less affinity. The distant homology of BHA007 and its orthologs to BBK32 proteins of Lyme disease species, as well as to previously described BBK32-like proteins in relapsing fever species, indicates that BHA007 is a member of a large family of multifunctional proteins in Borrelia species that bind to fibronectin as well as other host proteins.
Collapse
|
35
|
Han X, Tong Y, Tian M, Sun X, Wang S, Ding C, Yu S. Characterization of the immunogenicity and pathogenicity of malate dehydrogenase in Brucella abortus. World J Microbiol Biotechnol 2014; 30:2063-70. [PMID: 24609497 DOI: 10.1007/s11274-014-1631-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/04/2014] [Indexed: 01/24/2023]
Abstract
Brucella abortus is a gram-negative, facultative intracellular pathogen that causes brucellosis, a chronic zoonotic disease resulting in abortion in pregnant cattle and undulant fever in humans. Malate dehydrogenase (MDH), a key enzyme in the tricarboxylic acid cycle, plays important metabolic roles in aerobic energy producing pathways and in malate shuttle. In this study, the MDH-encoding gene for malate dehydrogenase mdh of B. abortus S2308 was cloned, sequenced and expressed. Western blot analysis demonstrated that MDH is an immunogenic membrane-associated protein. In addition, recombinant MDH showed sero-reactivity with 30 individual bovine B. abortus-positive sera by enzyme-linked immunosorbent assay, indicates that MDH may be used as a candidate marker for sero-diagnosis of brucellosis. Furthermore, MDH exhibits fibronectin and plasminogen-binding ability in immunoblotting assay. Inhibition assays on HeLa cells demonstrated that rabbit anti-serum against MDH significantly reduced both bacterial adherence and invasion abilities (p < 0.05), suggesting that MDH play a role in B. abortus colonization. Our results indicated that MDH is not only an immunogenic protein, but is also related to bacterial pathogenesis and may act as a new virulent factor, which will benefit for further understanding the MDH's roles in B. abortus metabolism, pathogenesis and immunity.
Collapse
Affiliation(s)
- Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
36
|
Foster TJ, Geoghegan JA, Ganesh VK, Höök M. Adhesion, invasion and evasion: the many functions of the surface proteins of Staphylococcus aureus. Nat Rev Microbiol 2014; 12:49-62. [PMID: 24336184 DOI: 10.1038/nrmicro3161] [Citation(s) in RCA: 1015] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Staphylococcus aureus is an important opportunistic pathogen and persistently colonizes about 20% of the human population. Its surface is 'decorated' with proteins that are covalently anchored to the cell wall peptidoglycan. Structural and functional analysis has identified four distinct classes of surface proteins, of which microbial surface component recognizing adhesive matrix molecules (MSCRAMMs) are the largest class. These surface proteins have numerous functions, including adhesion to and invasion of host cells and tissues, evasion of immune responses and biofilm formation. Thus, cell wall-anchored proteins are essential virulence factors for the survival of S. aureus in the commensal state and during invasive infections, and targeting them with vaccines could combat S. aureus infections.
Collapse
Affiliation(s)
- Timothy J Foster
- Microbiology Department, Moyne Institute of Preventive Medicine, Trinity College, Dublin 2, Ireland
| | - Joan A Geoghegan
- Microbiology Department, Moyne Institute of Preventive Medicine, Trinity College, Dublin 2, Ireland
| | - Vannakambadi K Ganesh
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A & M University Health Science Center, Houston, Texas 77030, USA
| | - Magnus Höök
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A & M University Health Science Center, Houston, Texas 77030, USA
| |
Collapse
|
37
|
Abstract
Streptococcus agalactiae (group B Streptococcus [GBS]) is a leading cause of neonatal sepsis and meningitis, peripartum infections in women, and invasive infections in chronically ill or elderly individuals. GBS can be isolated from the gastrointestinal or genital tracts of up to 30% of healthy adults, and infection is thought to arise from invasion from a colonized mucosal site. Accordingly, bacterial surface components that mediate attachment of GBS to host cells or the extracellular matrix represent key factors in the colonization and infection of the human host. We identified a conserved GBS gene of unknown function that was predicted to encode a cell wall-anchored surface protein. Deletion of the gene and a cotranscribed upstream open reading frame (ORF) in GBS strain 515 reduced bacterial adherence to VK2 vaginal epithelial cells in vitro and reduced GBS binding to fibronectin-coated microtiter wells. Expression of the gene product in Lactococcus lactis conferred the ability to adhere to VK2 cells, to fibronectin and laminin, and to fibronectin-coated ME-180 cervical epithelial cells. Expression of the recombinant protein in L. lactis also markedly increased biofilm formation. The adherence function of the protein, named bacterial surface adhesin of GBS (BsaB), depended both on a central BID1 domain found in bacterial intimin-like proteins and on the C-terminal portion of the BsaB protein. Expression of BsaB in GBS, like that of several other adhesins, was regulated by the CsrRS two-component system. We conclude that BsaB represents a newly identified adhesin that participates in GBS attachment to epithelial cells and the extracellular matrix.
Collapse
|
38
|
Gaultney RA, Gonzalez T, Floden AM, Brissette CA. BB0347, from the lyme disease spirochete Borrelia burgdorferi, is surface exposed and interacts with the CS1 heparin-binding domain of human fibronectin. PLoS One 2013; 8:e75643. [PMID: 24086600 PMCID: PMC3785480 DOI: 10.1371/journal.pone.0075643] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 08/16/2013] [Indexed: 11/18/2022] Open
Abstract
The causative agent of Lyme disease, Borrelia burgdorferi, codes for several known fibronectin-binding proteins. Fibronectin a common the target of diverse bacterial pathogens, and has been shown to be essential in allowing for the development of certain disease states. Another borrelial protein, BB0347, has sequence similarity with these other known fibronectin-binding proteins, and may be important in Lyme disease pathogenesis. Herein, we perform an initial characterization of BB0347 via the use of molecular and biochemical techniques. We found that BB0347 is expressed, produced, and presented on the outer surface of intact B. burgdorferi. We also demonstrate that BB0347 has the potential to be important in Lyme disease progression, and have begun to characterize the nature of the interaction between human fibronectin and this bacterial protein. Further work is needed to define the role of this protein in the borrelial infection process.
Collapse
Affiliation(s)
- Robert A. Gaultney
- Department of Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Edwin C. James Medical Research Facility Grand Forks, North Dakota, United States of America
| | - Tammy Gonzalez
- Department of Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Edwin C. James Medical Research Facility Grand Forks, North Dakota, United States of America
| | - Angela M. Floden
- Department of Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Edwin C. James Medical Research Facility Grand Forks, North Dakota, United States of America
| | - Catherine A. Brissette
- Department of Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Edwin C. James Medical Research Facility Grand Forks, North Dakota, United States of America
- * E-mail:
| |
Collapse
|
39
|
Hertig S, Chabria M, Vogel V. Engineering mechanosensitive multivalent receptor-ligand interactions: why the nanolinker regions of bacterial adhesins matter. NANO LETTERS 2012; 12:5162-5168. [PMID: 22938173 DOI: 10.1021/nl302153h] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Inspired by bacterial adhesins, we present a promising strategy of how to engineer peptides to probe various mechanical strains of extracellular matrix fibers. Functional sequence alignment of bacterial adhesins reveals that the bacterial linkers connecting the multivalent binding motifs recognizing fibronectin show considerable heterogeneity in length. Their length regulates the tunable affinities for fibronectin fibrils when stretched into different mechanical strain states. This platform has potential applications in probing extracellular matrix fiber strains in tissues.
Collapse
Affiliation(s)
- Samuel Hertig
- Department of Health Sciences and Technology, ETH Zurich, CH-8093, Zürich, Switzerland
| | | | | |
Collapse
|
40
|
Ribeiro M, Monteiro FJ, Ferraz MP. Infection of orthopedic implants with emphasis on bacterial adhesion process and techniques used in studying bacterial-material interactions. BIOMATTER 2012; 2:176-94. [PMID: 23507884 PMCID: PMC3568104 DOI: 10.4161/biom.22905] [Citation(s) in RCA: 470] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Staphylococcus comprises up to two-thirds of all pathogens in orthopedic implant infections and they are the principal causative agents of two major types of infection affecting bone: septic arthritis and osteomyelitis, which involve the inflammatory destruction of joint and bone. Bacterial adhesion is the first and most important step in implant infection. It is a complex process influenced by environmental factors, bacterial properties, material surface properties and by the presence of serum or tissue proteins. Properties of the substrate, such as chemical composition of the material, surface charge, hydrophobicity, surface roughness and the presence of specific proteins at the surface, are all thought to be important in the initial cell attachment process. The biofilm mode of growth of infecting bacteria on an implant surface protects the organisms from the host immune system and antibiotic therapy. The research for novel therapeutic strategies is incited by the emergence of antibiotic-resistant bacteria. This work will provide an overview of the mechanisms and factors involved in bacterial adhesion, the techniques that are currently being used studying bacterial-material interactions as well as provide insight into future directions in the field.
Collapse
Affiliation(s)
- Marta Ribeiro
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | | | | |
Collapse
|
41
|
Li W, Wan Y, Tao Z, Chen H, Zhou R. A novel fibronectin-binding protein of Streptococcus suis serotype 2 contributes to epithelial cell invasion and in vivo dissemination. Vet Microbiol 2012; 162:186-94. [PMID: 23021642 DOI: 10.1016/j.vetmic.2012.09.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Revised: 09/04/2012] [Accepted: 09/04/2012] [Indexed: 12/30/2022]
Abstract
Streptococcus suis serotype 2 (SS2) is an important pathogen with zoonotic potential. In this study, a novel in vivo induced protein Ssa, encoded by the functionally unknown gene SSU05_1311, was identified as a surface anchored fibronectin-binding protein. The recombinant Ssa as well as its truncated proteins harboring the N-terminal domain (residues 33-153) could bind to human fibronectin with high-affinity. Isogenic knockout of ssa in SS2 led to decrease of bacterial binding to immobilized fibronectin. SS2 Δssa mutant showed reduced adherence and invasion of human pharyngeal epithelial (HEp-2) cells compared to the wild type strain. Heterologous surface display of Ssa fibronectin-binding domain in Escherichia coli enhanced the bacterial attachment and entry to HEp-2 cells. Less SS2 Δssa mutants than the wild type strain were recovered from the blood and brains of the Balb/c mice infected intranasally. But there was no significant difference between the wild type and the mutant on phagocytosis by macrophages (RAW264.7) and bacterial killing by murine PMNs under opsonizing condition. Our data suggest that Ssa is an important virulence factor for SS2 crossing the mucosal epithelia to disseminate in vivo.
Collapse
Affiliation(s)
- Wei Li
- Division of Animal Infectious Diseases in State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | | | | | | | | |
Collapse
|
42
|
Hauck CR, Borisova M, Muenzner P. Exploitation of integrin function by pathogenic microbes. Curr Opin Cell Biol 2012; 24:637-44. [PMID: 22884865 DOI: 10.1016/j.ceb.2012.07.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 01/26/2023]
Abstract
Numerous pathogens express adhesive proteins to directly or indirectly associate with integrins. It is well established that by targeting integrins, microbes not only establish an intimate contact with host tissues, but also trigger cellular responses including bacterial internalization. This review will summarize current knowledge about the role of these integrin-dependent processes during infection and how bacteria assure that they efficiently connect to integrins for host cell invasion or translocation of effector molecules. Furthermore, we will discuss recent insight demonstrating that bacteria can harness the physiological, matrix-binding function of integrins for promoting host colonization. From these combined studies, it is becoming evident that integrins are a common nexus for the manipulation of cellular functions by bacterial pathogens. Approaches to disrupt this connection might be an appropriate means to obtain broad-acting tools to modulate a spectrum of infectious diseases.
Collapse
Affiliation(s)
- Christof R Hauck
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Germany.
| | | | | |
Collapse
|
43
|
Torelli R, Serror P, Bugli F, Paroni Sterbini F, Florio AR, Stringaro A, Colone M, De Carolis E, Martini C, Giard JC, Sanguinetti M, Posteraro B. The PavA-like fibronectin-binding protein of Enterococcus faecalis, EfbA, is important for virulence in a mouse model of ascending urinary tract infection. J Infect Dis 2012; 206:952-60. [PMID: 22782954 DOI: 10.1093/infdis/jis440] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Enterococcus faecalis is an established nosocomial pathogen, yet the pathogenesis of enterococcal infections, particularly of urinary tract infections (UTIs), remains to be fully elucidated. Fibronectin-binding proteins have been identified as potent adhesins in pathogenic Gram-positive cocci. Here, we characterized EfbA, which is encoded by the enterococcal orthologue of Streptococcus pneumoniae pavA. Similar to PavA, the anchorless EfbA protein was localized to the enterococcal cell outer surface and bound to immobilized human fibronectin. In addition to abrogated EfbA expression, deletion of the efbA gene eliminated EfbA from the cell surface and drastically reduced the enterococcal cell binding to immobilized fibronectin. The ΔefbA deletion mutant was highly attenuated vs wild-type in a murine ascending UTI model, consistent with an increased tropism for the kidney relative to the bladder. These results provide the first evidence that EfbA of E. faecalis plays a role in UTIs, probably contributing to the pathogenesis in this site.
Collapse
Affiliation(s)
- Riccardo Torelli
- Institute of Microbiology, Università Cattolica del Sacro Cuore, Largo F Vito, 1 00168 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Voss S, Gámez G, Hammerschmidt S. Impact of pneumococcal microbial surface components recognizing adhesive matrix molecules on colonization. Mol Oral Microbiol 2012; 27:246-56. [PMID: 22759310 DOI: 10.1111/j.2041-1014.2012.00654.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Microorganisms have evolved elaborate strategies to adhere to host cells and to evade the host complement and immune attack, ensuring survival in various host niches and dissemination into sterile parts of the human body. Streptococcus pneumoniae (the pneumococcus) is not only a commensal of the human respiratory tract but also the etiological agent of severe and life-threatening diseases. Pneumococcal attachment to mucosal surfaces is a highly dynamic process requiring the contact of pneumococcal surface-exposed proteins with soluble or immobilized host factors. These avid interactions may trigger proteolytic cascades or result in engagement of cell surface receptors and intracellularly associated signaling machineries for subsequent uptake of pneumococci into host cells. In the present review, the intimate communication of S. pneumoniae molecules recognizing adhesive matrix molecules (microbial surface components recognizing adhesive matrix molecules) with their host counterparts and their individual role in pneumococcal colonization is discussed.
Collapse
Affiliation(s)
- S Voss
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | | | | |
Collapse
|
45
|
Maurer LM, Ma W, Eickstaedt NL, Johnson IA, Tomasini-Johansson BR, Annis DS, Mosher DF. Ligation of the fibrin-binding domain by β-strand addition is sufficient for expansion of soluble fibronectin. J Biol Chem 2012; 287:13303-12. [PMID: 22351755 PMCID: PMC3339936 DOI: 10.1074/jbc.m111.294041] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 02/02/2012] [Indexed: 11/06/2022] Open
Abstract
How fibronectin (FN) converts from a compact plasma protein to a fibrillar component of extracellular matrix is not understood. "Functional upstream domain" (FUD), a polypeptide based on F1 adhesin of Streptococcus pyogenes, binds by anti-parallel β-strand addition to discontinuous sets of N-terminal FN type I modules, (2-5)FNI of the fibrin-binding domain and (8-9)FNI of the gelatin-binding domain. Such binding blocks assembly of FN. To learn whether ligation of (2-5)FNI, (8-9)FNI, or the two sets in combination is important for inhibition, we tested "high affinity downstream domain" (HADD), which binds by β-strand addition to the continuous set of FNI modules, (1-5)FNI, comprising the fibrin-binding domain. HADD and FUD were similarly active in blocking fibronectin assembly. Binding of HADD or FUD to soluble plasma FN exposed the epitope to monoclonal antibody mAbIII-10 in the tenth FN type III module ((10)FNIII) and caused expansion of FN as assessed by dynamic light scattering. Soluble N-terminal constructs truncated after (9)FNI or (3)FNIII competed better than soluble FN for binding of FUD or HADD to adsorbed FN, indicating that interactions involving type III modules more C-terminal than (3)FNIII limit β-strand addition to (1-5)FNI within intact soluble FN. Preincubation of FN with mAbIII-10 or heparin modestly increased binding to HADD or FUD. Thus, ligation of FNIII modules involved in binding of integrins and glycosaminoglycans, (10)FNIII and (12-14)FNIII, increases accessibility of (1-5)FNI. Allosteric loss of constraining interactions among (1-5)FNI, (10)FNIII, and (12-14)FNIII likely enables assembly of FN into extracellular fibrils.
Collapse
Affiliation(s)
- Lisa M. Maurer
- From the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Wenjiang Ma
- From the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Nathan L. Eickstaedt
- From the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Ian A. Johnson
- From the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Bianca R. Tomasini-Johansson
- From the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Douglas S. Annis
- From the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Deane F. Mosher
- From the Departments of Biomolecular Chemistry and Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
46
|
Tsurupa G, Pechik I, Litvinov RI, Hantgan RR, Tjandra N, Weisel JW, Medved L. On the mechanism of αC polymer formation in fibrin. Biochemistry 2012; 51:2526-38. [PMID: 22397628 DOI: 10.1021/bi2017848] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Our previous studies revealed that the fibrinogen αC-domains undergo conformational changes and adopt a physiologically active conformation upon their self-association into αC polymers in fibrin. In the present study, we analyzed the mechanism of αC polymer formation and tested our hypothesis that self-association of the αC-domains occurs through the interaction between their N-terminal subdomains and may include β-hairpin swapping. Our binding experiments performed by size-exclusion chromatography and optical trap-based force spectroscopy revealed that the αC-domains self-associate exclusively through their N-terminal subdomains, while their C-terminal subdomains were found to interact with the αC-connectors that tether the αC-domains to the bulk of the molecule. This interaction should reinforce the structure of αC polymers and provide the proper orientation of their reactive residues for efficient cross-linking by factor XIIIa. Molecular modeling of self-association of the N-terminal subdomains confirmed that the hypothesized β-hairpin swapping does not impose any steric hindrance. To "freeze" the conformation of the N-terminal subdomain and prevent the hypothesized β-hairpin swapping, we introduced by site-directed mutagenesis an extra disulfide bond between two β-hairpins of the bovine Aα406-483 fragment corresponding to this subdomain. The experiments performed by circular dichroism revealed that Aα406-483 mutant containing Lys429Cys/Thr463Cys mutations preserved its β-sheet structure. However, in contrast to wild-type Aα406-483, this mutant had lower tendency for oligomerization, and its structure was not stabilized upon oligomerization, in agreement with the above hypothesis. On the basis of the results obtained and our previous findings, we propose a model of fibrin αC polymer structure and molecular mechanism of assembly.
Collapse
Affiliation(s)
- Galina Tsurupa
- Center for Vascular and Inflammatory Diseases and the Department of Biochemistry, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | | | | | | | | | | | | |
Collapse
|
47
|
Kneidl J, Löffler B, Erat MC, Kalinka J, Peters G, Roth J, Barczyk K. Soluble CD163 promotes recognition, phagocytosis and killing of Staphylococcus aureus via binding of specific fibronectin peptides. Cell Microbiol 2012; 14:914-36. [PMID: 22309204 DOI: 10.1111/j.1462-5822.2012.01766.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CD163 is a multi-ligand scavenger receptor exclusively expressed by monocytes and macrophages, which is released after their activation during sepsis (sCD163). The biological relevance of sCD163, however, is not yet clear. We now demonstrate that sCD163 exhibits direct antimicrobial effects by recognizing a specific subfragment ((6) F1(1) F2(2) F2(7) F1) of fibronectin (FN) bound to staphylococcal surface molecules. Moreover, contact with staphylococci promotes sCD163-shedding from monocyte surface via induction of metalloproteinases ADAM10 and ADAM17. sCD163 subsequently binds to Staphylococcus aureus via FN peptides and strongly amplifies phagocytosis as well as killing by monocytes and to a lesser extend by neutrophils. This mechanism exhibits additional paracrine effects because staphylococci additionally opsonized by sCD163 induce higher activation and more efficient killing activity of non-professional phagocytes like endothelial cells. Targeting pathogen-bound FN by sCD163 would be a very sophisticated strategy to attack S. aureus as any attempt of the pathogen to avoid this defence mechanism will automatically bring about loss of adherence to the host protein FN, which is a pivotal patho-mechanism of highly invasive staphylococcal strains. Thus, we report a novel function for sCD163 that is of particular importance for immune defence of the host against S. aureus infections.
Collapse
Affiliation(s)
- Jessica Kneidl
- Institute of Immunology, University of Münster, 48149 Münster, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Zoonotic infections caused by Streptococcus spp. have been neglected in spite of the fact that frequency and severity of outbreaks increased dramatically in recent years. This may be due to non-identification since respective species are often not considered in human medical diagnostic procedures. On the other hand, an expanding human population concomitant with an increasing demand for food and the increased number of companion animals favour conditions for host species adaptation of animal streptococci. This review aims to give an overview on streptococcal zoonoses with focus on epidemiology and pathogenicity of four major zoonotic species, Streptococcus canis, Streptococcus equi sub. zooepidemicus, Streptococcus iniae and Streptococcus suis.
Collapse
|
49
|
Wang Y, Subbiahdoss G, de Vries J, Libera M, van der Mei HC, Busscher HJ. Effect of adsorbed fibronectin on the differential adhesion of osteoblast-like cells and Staphylococcus aureus with and without fibronectin-binding proteins. BIOFOULING 2012; 28:1011-1021. [PMID: 23004018 DOI: 10.1080/08927014.2012.725471] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The influence of fibronectin (Fn) coated surfaces patterned with poly(ethylene glycol) microgels having inter-gel spacings between 0.5 and 3.0 μm on the adhesion of Staphylococcus aureus strains with and without Fn-binding proteins and cellular adhesion/spreading was investigated. Quantitative force measurements between a S. aureus cell and a patterned surface showed that the adhesion force between the bacterium and the patterned surface increased substantially after Fn adsorption, regardless of the strain used, but decreased with decreasing inter-gel spacing. In flow-chamber experiments, the Fn-binding strain adhered at a higher rate after Fn adsorption than the strain lacking Fn-binding proteins. In both cases, the adhesion rates decreased with decreasing inter-gel spacing. Osteoblast-like cells could bind to patterned surfaces despite the microgels, and adsorbed Fn substantially amplified this effect. Even under highly non-adhesive conditions associated with closely spaced microgels, adsorbed Fn preserves a window of inter-gel spacing around 1 μm where the adhesion of staphylococcal cells is hindered while cells can still adhere and spread.
Collapse
Affiliation(s)
- Y Wang
- Department of Biomedical Engineering, W.J. Kolff Institute, University Medical Center and University of Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
50
|
Krachler AM, Ham H, Orth K. Turnabout is fair play: use of the bacterial Multivalent Adhesion Molecule 7 as an antimicrobial agent. Virulence 2012; 3:68-71. [PMID: 22086133 DOI: 10.4161/viru.3.1.18172] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Pathogen attachment to host tissues is one of the initial and most crucial events during the establishment of bacterial infections and thus interference with this step could be an efficient strategy to fight bacterial colonization. Our recent work has identified one of the factors involved in initial binding of host cells by a wide range of Gram-negative pathogens, Multivalent Adhesion Molecule (MAM) 7. Interference with MAM7-mediated attachment, for example by pre-incubation of host cells with recombinant MAM7, significantly delays the onset of hallmarks of infection, such as pathogen-mediated cytotoxicity or the development of other adhesive structures such as actin pedestals. Thus, we are trying to develop tools based on MAM7 that can be used to prevent or diminish certain Gram-negative bacterial infections. Herein, we describe the use of bead-coupled MAM7 as an inhibitor of infection with the clinically relevant pathogen Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Anne Marie Krachler
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | | |
Collapse
|