1
|
Manyi-Loh CE, Lues R. Listeria monocytogenes and Listeriosis: The Global Enigma. Foods 2025; 14:1266. [PMID: 40238523 PMCID: PMC11989209 DOI: 10.3390/foods14071266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Listeria monocytogenes is an intracellular, Gram-positive, non-spore-forming, non-encapsulated, facultative anaerobic, rod-shaped, and psychrotrophic food-borne pathogen that causes the infection, listeriosis, thus it attracts great attention following listeriosis outbreaks, which are often associated with high mortality rates. The prevalence of listeriosis is quite low globally; however, the most recent and deadliest outbreak occurred in South Africa, during which 216 persons lost their lives. L. monocytogenes is endowed with the potential to multiply through a wide range of harsh environmental conditions, forming biofilms on varying surfaces in the food industry, as well as having persistent and antibiotic-resistant cells, which pose a major threat and burden to the ready-to-eat food industry. A more frustrating characteristic of this bacterium is its strain divergence, alongside an increased level of antibiotic resistance registered among the strains of L. monocytogenes recovered from food, humans, and environmental sources, especially to those antibiotics involved in the treatment of human listeriosis. Antibiotic resistance exerted by and among pathogenic food-borne microbes is an ongoing public health menace that continues to be an issue. Against this background, a thorough search into different databases using various search engines was performed, which led to the gathering of salient information that was organised, chronologically, based on Listeria monocytogenes and listeriosis. Altogether, the findings elaborated in this study present up-to date knowledge on different aspects of this pathogen which will improve our understanding of the mystery associated with it and the ways to prevent and control its dissemination through ready-to-eat foods. In addition, constant monitoring of the antibiotic resistance profiles of strains of L. monocytogenes from varying sources detected changes, giving an update on the trend in antibiotic resistance. Overall, monitoring of bacterial contamination serves as the key aspect in the control of the food safety output in the food industry.
Collapse
Affiliation(s)
- Christy E. Manyi-Loh
- Centre for Applied Food Sustainability and Biotechnology, Central University of Technology, Bloemfontein X9301, South Africa;
| | | |
Collapse
|
2
|
Castro-Cordova P, Lopez-Garcia OK, Orozco J, Montes-Bravo N, Gil F, Pizarro-Guajardo M, Paredes-Sabja D. Clostridioides difficile major toxins remodel the intestinal epithelia, affecting spore adherence/internalization into intestinal tissue and their association with gut vitronectin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635439. [PMID: 39974910 PMCID: PMC11838273 DOI: 10.1101/2025.01.29.635439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The most common cause of healthcare-associated diarrhea and colitis in the U.S., is Clostridioides difficile, a spore-forming pathogen. Two toxins, TcdA and TcdB, are major virulence factors essential for disease manifestations, while C. difficile spores are essential for disease transmission and recurrence. Both toxins cause major damage to the epithelial barrier, trigger massive inflammation, and reshape the microbiome and metabolic composition, facilitating C. difficile colonization. C. difficile spores, essential for transmission and recurrence of the disease, persist adhered and internalized in the intestinal epithelia. Studies have suggested that toxin-neutralization in combination with antibiotic during CDI treatment in humans significantly reduces disease recurrence, suggesting a link between toxin-mediated damage and spore persistence. Here, we show that TcdA/TcdB-intoxication of intestinal epithelial Caco-2 cells leads to remodeling of accessible levels of fibronectin (Fn) and vitronectin (Vn) and their cognate alpha-integrin subunits. While TcdB-intoxication of intestinal tissue had no impact in accessible levels of Fn and Vn, but significantly increased levels of intracellular Vn. We observed that Fn and Vn released to the supernatant readily bind to C. difficile spores in vitro, while TcdB-intoxication of intestinal tissue led to increased association of C. difficile spores with gut Vn. Toxin-intoxication of the intestinal tissue also contributes to increased adherence and internalization of C. difficile spores. However, TcdB-intoxicated ligated loops infected of mice treated with Bezlotoxumanb (monoclonal anti-TcdB antibodies) did not prevent TcdB-mediated increased spore adherence and internalization into intestinal tissue. This study highlights the importance of studying the impact of C. difficile toxins of host tissues has in C. difficile interaction with host surfaces that may contribute to increased persistence and disease recurrence.
Collapse
Affiliation(s)
- Pablo Castro-Cordova
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Chile
| | - Osiris K. Lopez-Garcia
- Interdisciplinary Program in Genetics & Genomics, Texas A&M University, College Station, TX USA
- Department of Biology, Texas A&M University, College Station, TX USA
| | - Josué Orozco
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
| | | | - Fernando Gil
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- Microbiota-Host Interactions & Clostridia Research Group, Universidad Andres Bello, Santiago, Chile
| | - Marjorie Pizarro-Guajardo
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- Interdisciplinary Program in Genetics & Genomics, Texas A&M University, College Station, TX USA
| | - Daniel Paredes-Sabja
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- Interdisciplinary Program in Genetics & Genomics, Texas A&M University, College Station, TX USA
- Department of Biology, Texas A&M University, College Station, TX USA
| |
Collapse
|
3
|
Ma H, Zhao Y, He X, Wang Q, Zhang Y, Xing X, Wu X, Quan G, Bao S. Dihydrolipoamide acetyltransferase is a key factor mediating adhesion and invasion of host cells by Mycoplasma synoviae. Vet Microbiol 2024; 299:110297. [PMID: 39561529 DOI: 10.1016/j.vetmic.2024.110297] [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: 05/23/2024] [Revised: 10/06/2024] [Accepted: 11/02/2024] [Indexed: 11/21/2024]
Abstract
Mycoplasma synoviae is a significant avian pathogen responsible for chronic respiratory diseases, arthritis, and infectious synovitis in chickens and turkeys. These infections result in substantial economic losses to the global poultry industry. Dihydrolipoamide acetyltransferase (E2) is a multifunctional protein that plays an indispensable role in energy metabolism and redox balance and is also a key virulence factor of various pathogens. In this study, we used the avian pathogen M. synoviae as a model to identify the role of the E2 protein in the colonization and invasion of host cells. First, we prepared the polyclonal antibody of recombinant E2 (rE2) protein and found that the rE2 antibody had a strong complement-activating ability. E2 was found to be distributed in the cytoplasm and cell membrane of M. synoviae by immunoelectron microscopy. E2 localized on the cell membrane is a key factor in the adhesion of M. synoviae and has good immunogenicity. Enzyme-linked immunosorbent assay showed that the binding of rE2 to membrane proteins of chicken embryo fibroblasts (DF-1) was dose-dependent, and antiserum effectively inhibited this binding ability. Furthermore, E2 interacted with various components of the host extracellular matrix (ECM) and promoted the conversion of plasminogen to plasmin through terephthalic acid (tPA). In addition, E2 can enhance the ability of M. synoviae to invade DF-1 cells, which was significantly reduced after treatment with anti-E2 serum. These results indicate that E2 is an adhesion- and invasion-related protein and may be involved in the pathogenesis of M. synoviae, which provides new ideas for studying the pathogenesis of M. synoviae and preparing subunit vaccines.
Collapse
Affiliation(s)
- Haiyun Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| | - Yunhai Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| | - Xiaoxiao He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| | - Qing Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| | - Yuting Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| | - Xiaoyong Xing
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| | - Xiaochun Wu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| | - Guomei Quan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| | - Shijun Bao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| |
Collapse
|
4
|
Guo M, Li Y, Tang J, Wang Q, Wang Q, Zhou H, Lin H, Ma Z, Fan H. Glaesserella parasuis serotype 4 exploits fibronectin via RlpA for tracheal colonization following porcine circovirus type 2 infection. PLoS Pathog 2024; 20:e1012513. [PMID: 39264911 PMCID: PMC11392263 DOI: 10.1371/journal.ppat.1012513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024] Open
Abstract
Porcine circovirus type 2 (PCV2) often causes disease through coinfection with other bacterial pathogens, including Glaesserella parasuis (G. parasuis), which causes high morbidity and mortality, but the role played by PCV2 and bacterial and host factors contributing to this process have not been defined. Bacterial attachment is assumed to occur via specific receptor-ligand interactions between adhesins on the bacterial cell and host proteins adsorbed to the implant surface. Mass spectrometry (MS) analysis of PCV2-infected swine tracheal epithelial cells (STEC) revealed that the expression of Extracellular matrix protein (ECM) Fibronectin (Fn) increased significantly on the infected cells surface. Importantly, efficient G. parasuis serotype 4 (GPS4) adherence to STECs was imparted by interactions with Fn. Furthermore, abrogation of adherence was gained by genetic knockout of Fn, Fn and Integrin β1 antibody blocking. Fn is frequently exploited as a receptor for bacterial pathogens. To explore the GPS4 adhesin that interacts with Fn, recombinant Fn N-terminal type I and type II domains were incubated with GPS4, and the interacting proteins were pulled down for MS analysis. Here, we show that rare lipoprotein A (RlpA) directly interacts with host Fibronectin mediating GPS4 adhesion. Finally, we found that PCV2-induced Fibronectin expression and adherence of GPS4 were prevented significantly by TGF-β signaling pathway inhibitor SB431542. Our data suggest the RlpA-Fn interaction to be a potentially promising novel therapeutic target to combat PCV2 and GPS4 coinfection.
Collapse
Affiliation(s)
- Mengru Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuhui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jinsheng Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qing Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Qiancheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hong Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Huixing Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhe Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hongjie Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
| |
Collapse
|
5
|
Abbasnia S, Hashem Asnaashari AM, Sharebiani H, Soleimanpour S, Mosavat A, Rezaee SA. Mycobacterium tuberculosis and host interactions in the manifestation of tuberculosis. J Clin Tuberc Other Mycobact Dis 2024; 36:100458. [PMID: 38983441 PMCID: PMC11231606 DOI: 10.1016/j.jctube.2024.100458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024] Open
Abstract
The final step of epigenetic processes is changing the gene expression in a new microenvironment in the body, such as neuroendocrine changes, active infections, oncogenes, or chemical agents. The case of tuberculosis (TB) is an outcome of Mycobacterium tuberculosis (M.tb) and host interaction in the manifestation of active and latent TB or clearance. This comprehensive review explains and interprets the epigenetics findings regarding gene expressions on the host-pathogen interactions in the development and progression of tuberculosis. This review introduces novel insights into the complicated host-pathogen interactions, discusses the challengeable results, and shows the gaps in the clear understanding of M.tb behavior. Focusing on the biological phenomena of host-pathogen interactions, the epigenetic changes, and their outcomes provides a promising future for developing effective TB immunotherapies when converting gene expression toward appropriate host immune responses gradually becomes attainable. Overall, this review may shed light on the dark sides of TB pathogenesis as a life-threatening disease. Therefore, it may support effective planning and implementation of epigenetics approaches for introducing proper therapies or effective vaccines.
Collapse
Affiliation(s)
- Shadi Abbasnia
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hiva Sharebiani
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture, and Research (ACECR), Razavi Khorasan, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Rima M, Villeneuve-Faure C, Soumbo M, El Garah F, Pilloux L, Roques C, Makasheva K. Towards a better understanding of the effect of protein conditioning layers on microbial adhesion: a focused investigation of fibronectin and bovine serum albumin layers on SiO 2 surfaces. Biomater Sci 2024; 12:3086-3099. [PMID: 38716803 DOI: 10.1039/d4bm00099d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
The interaction of foreign implants with their surrounding environment is significantly influenced by the adsorption of proteins on the biomaterial surfaces, playing a role in microbial adhesion. Therefore, understanding protein adsorption on solid surfaces and its effect on microbial adhesion is essential to assess the associated risk of infection. The aim of this study is to evaluate the effect of conditioning by fibronectin (Fn) or bovine serum albumin (BSA) protein layers of silica (SiO2) surfaces on the adhesion and detachment of two pathogenic microorganisms: Pseudomonas aeruginosa PAO1-Tn7-gfp and Candida albicans CIP 48.72. Experiments are conducted under both static and hydrodynamic conditions using a shear stress flow chamber. Through the use of very low wall shear stresses, the study brings the link between the static and dynamic conditions of microbial adhesion. The results reveal that the microbial adhesion critically depends on: (i) the presence of a protein layer conditioning the SiO2 surface, (ii) the type of protein and (iii) the protein conformation and organization in the conditioning layer. In addition, a very distinct adhesion behaviour of P. aeruginosa is observed towards the two tested proteins, Fn and BSA. This effect is reinforced by the amount of proteins adsorbed on the surface and their organization in the layer. The results are discussed in the light of atomic force microscopy analysis of the organization and conformation of proteins in the layers after adsorption on the SiO2 surface, as well as the specificity in bacterial behaviour when interacting with these protein layers. The study also demonstrates the very distinctive behaviours of the prokaryote P. aeruginosa PAO1-Tn7-gfp compared to the eukaryote C. albicans CIP 48.72. This underscores the importance of considering species-specific interactions between the protein conditioning layer and different pathogenic microorganisms, which appear crucial in designing tailored anti-adhesive surfaces.
Collapse
Affiliation(s)
- Maya Rima
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | | | - Marvine Soumbo
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
- LAPLACE, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | - Fatima El Garah
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | - Ludovic Pilloux
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | - Christine Roques
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | - Kremena Makasheva
- LAPLACE, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| |
Collapse
|
7
|
Bonadio JD, Bashiri G, Halligan P, Kegel M, Ahmed F, Wang K. Delivery technologies for therapeutic targeting of fibronectin in autoimmunity and fibrosis applications. Adv Drug Deliv Rev 2024; 209:115303. [PMID: 38588958 PMCID: PMC11111362 DOI: 10.1016/j.addr.2024.115303] [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: 10/02/2023] [Revised: 02/29/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Fibronectin (FN) is a critical component of the extracellular matrix (ECM) contributing to various physiological processes, including tissue repair and immune response regulation. FN regulates various cellular functions such as adhesion, proliferation, migration, differentiation, and cytokine release. Alterations in FN expression, deposition, and molecular structure can profoundly impact its interaction with other ECM proteins, growth factors, cells, and associated signaling pathways, thus influencing the progress of diseases such as fibrosis and autoimmune disorders. Therefore, developing therapeutics that directly target FN or its interaction with cells and other ECM components can be an intriguing approach to address autoimmune and fibrosis pathogenesis.
Collapse
Affiliation(s)
- Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Fatima Ahmed
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
8
|
Ou L, Ye B, Sun M, Qi N, Li J, Lv M, Lin X, Cai H, Hu J, Song Y, Chen X, Zhu Y, Yin L, Zhang J, Liao S, Zhang H. Mechanisms of intestinal epithelial cell damage by Clostridiumperfringens. Anaerobe 2024; 87:102856. [PMID: 38609034 DOI: 10.1016/j.anaerobe.2024.102856] [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: 11/27/2023] [Revised: 03/31/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Clostridium perfringens, a Gram-positive bacterium, causes intestinal diseases in humans and livestock through its toxins, related to alpha toxin (CPA), beta toxin (CPB), C. perfringens enterotoxin (CPE), epsilon toxin (ETX), Iota toxin (ITX), and necrotic enteritis B-like toxin (NetB). These toxins disrupt intestinal barrier, leading to various cell death mechanisms such as necrosis, apoptosis, and necroptosis. Additionally, non-toxin factors like adhesins and degradative enzymes contribute to virulence by enhancing colonization and survival of C. perfringens. A vicious cycle of intestinal barrier breach, misregulated cell death, and subsequent inflammation is at the heart of chronic inflammatory and infectious gastrointestinal diseases. Understanding these mechanisms is essential for developing targeted therapies against C. perfringens-associated intestinal diseases.
Collapse
Affiliation(s)
- Lanxin Ou
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China; College of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Bijin Ye
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China; College of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Mingfei Sun
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Nanshan Qi
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Juan Li
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Minna Lv
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Xuhui Lin
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Haiming Cai
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Junjing Hu
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Yongle Song
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Xiangjie Chen
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Yibin Zhu
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Lijun Yin
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Jianfei Zhang
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Shenquan Liao
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Key Laboratory of Avian Influenza and Other Major Poultry Diseases Prevention and Control, Ministry of Agriculture and Rural Affairs, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China.
| | - Haoji Zhang
- College of Life Science and Engineering, Foshan University, Foshan, 528225, China.
| |
Collapse
|
9
|
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
|
10
|
Lin Q, Lin S, Fan Z, Liu J, Ye D, Guo P. A Review of the Mechanisms of Bacterial Colonization of the Mammal Gut. Microorganisms 2024; 12:1026. [PMID: 38792855 PMCID: PMC11124445 DOI: 10.3390/microorganisms12051026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
A healthy animal intestine hosts a diverse population of bacteria in a symbiotic relationship. These bacteria utilize nutrients in the host's intestinal environment for growth and reproduction. In return, they assist the host in digesting and metabolizing nutrients, fortifying the intestinal barrier, defending against potential pathogens, and maintaining gut health. Bacterial colonization is a crucial aspect of this interaction between bacteria and the intestine and involves the attachment of bacteria to intestinal mucus or epithelial cells through nonspecific or specific interactions. This process primarily relies on adhesins. The binding of bacterial adhesins to host receptors is a prerequisite for the long-term colonization of bacteria and serves as the foundation for the pathogenicity of pathogenic bacteria. Intervening in the adhesion and colonization of bacteria in animal intestines may offer an effective approach to treating gastrointestinal diseases and preventing pathogenic infections. Therefore, this paper reviews the situation and mechanisms of bacterial colonization, the colonization characteristics of various bacteria, and the factors influencing bacterial colonization. The aim of this study was to serve as a reference for further research on bacteria-gut interactions and improving animal gut health.
Collapse
Affiliation(s)
- Qingjie Lin
- College of Animal Science, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Fuzhou 350002, China; (Q.L.); (S.L.); (Z.F.)
| | - Shiying Lin
- College of Animal Science, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Fuzhou 350002, China; (Q.L.); (S.L.); (Z.F.)
| | - Zitao Fan
- College of Animal Science, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Fuzhou 350002, China; (Q.L.); (S.L.); (Z.F.)
| | - Jing Liu
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China;
| | - Dingcheng Ye
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China;
| | - Pingting Guo
- College of Animal Science, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Fuzhou 350002, China; (Q.L.); (S.L.); (Z.F.)
| |
Collapse
|
11
|
Schirmer M, Stražar M, Avila-Pacheco J, Rojas-Tapias DF, Brown EM, Temple E, Deik A, Bullock K, Jeanfavre S, Pierce K, Jin S, Invernizzi R, Pust MM, Costliow Z, Mack DR, Griffiths AM, Walters T, Boyle BM, Kugathasan S, Vlamakis H, Hyams J, Denson L, Clish CB, Xavier RJ. Linking microbial genes to plasma and stool metabolites uncovers host-microbial interactions underlying ulcerative colitis disease course. Cell Host Microbe 2024; 32:209-226.e7. [PMID: 38215740 PMCID: PMC10923022 DOI: 10.1016/j.chom.2023.12.013] [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: 03/13/2023] [Revised: 11/08/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024]
Abstract
Understanding the role of the microbiome in inflammatory diseases requires the identification of microbial effector molecules. We established an approach to link disease-associated microbes to microbial metabolites by integrating paired metagenomics, stool and plasma metabolomics, and culturomics. We identified host-microbial interactions correlated with disease activity, inflammation, and the clinical course of ulcerative colitis (UC) in the Predicting Response to Standardized Colitis Therapy (PROTECT) pediatric inception cohort. In severe disease, metabolite changes included increased dipeptides and tauro-conjugated bile acids (BAs) and decreased amino-acid-conjugated BAs in stool, whereas in plasma polyamines (N-acetylputrescine and N1-acetylspermidine) increased. Using patient samples and Veillonella parvula as a model, we uncovered nitrate- and lactate-dependent metabolic pathways, experimentally linking V. parvula expansion to immunomodulatory tryptophan metabolite production. Additionally, V. parvula metabolizes immunosuppressive thiopurine drugs through xdhA xanthine dehydrogenase, potentially impairing the therapeutic response. Our findings demonstrate that the microbiome contributes to disease-associated metabolite changes, underscoring the importance of these interactions in disease pathology and treatment.
Collapse
Affiliation(s)
- Melanie Schirmer
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Translational Microbiome Data Integration, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany; ZIEL - Institute for Food & Health, Technical University of Munich, 85354 Freising, Germany.
| | - Martin Stražar
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | | | - Eric M Brown
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Emily Temple
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amy Deik
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kevin Bullock
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sarah Jeanfavre
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kerry Pierce
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shen Jin
- Translational Microbiome Data Integration, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | | | - Marie-Madlen Pust
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Zach Costliow
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David R Mack
- Division of Gastroenterology, Hepatology & Nutrition, Children's Hospital of Eastern Ontario and University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Anne M Griffiths
- Division of Gastroenterology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Thomas Walters
- Division of Gastroenterology, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Brendan M Boyle
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Subra Kugathasan
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
| | - Hera Vlamakis
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffrey Hyams
- Connecticut Children's Medical Center, Division of Digestive Diseases, Hartford, CT 06106, USA
| | - Lee Denson
- Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Clary B Clish
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ramnik J Xavier
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
12
|
Bhattacharya M, Horswill AR. The role of human extracellular matrix proteins in defining Staphylococcus aureus biofilm infections. FEMS Microbiol Rev 2024; 48:fuae002. [PMID: 38337187 PMCID: PMC10873506 DOI: 10.1093/femsre/fuae002] [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: 07/19/2023] [Revised: 01/26/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024] Open
Abstract
Twenty to forty one percent of the world's population is either transiently or permanently colonized by the Gram-positive bacterium, Staphylococcus aureus. In 2017, the CDC designated methicillin-resistant S. aureus (MRSA) as a serious threat, reporting ∼300 000 cases of MRSA-associated hospitalizations annually, resulting in over 19 000 deaths, surpassing that of HIV in the USA. S. aureus is a proficient biofilm-forming organism that rapidly acquires resistance to antibiotics, most commonly methicillin (MRSA). This review focuses on a large group of (>30) S. aureus adhesins, either surface-associated or secreted that are designed to specifically bind to 15 or more of the proteins that form key components of the human extracellular matrix (hECM). Importantly, this includes hECM proteins that are pivotal to the homeostasis of almost every tissue environment [collagen (skin), proteoglycans (lung), hemoglobin (blood), elastin, laminin, fibrinogen, fibronectin, and fibrin (multiple organs)]. These adhesins offer S. aureus the potential to establish an infection in every sterile tissue niche. These infections often endure repeated immune onslaught, developing into chronic, biofilm-associated conditions that are tolerant to ∼1000 times the clinically prescribed dose of antibiotics. Depending on the infection and the immune response, this allows S. aureus to seamlessly transition from colonizer to pathogen by subtly manipulating the host against itself while providing the time and stealth that it requires to establish and persist as a biofilm. This is a comprehensive discussion of the interaction between S. aureus biofilms and the hECM. We provide particular focus on the role of these interactions in pathogenesis and, consequently, the clinical implications for the prevention and treatment of S. aureus biofilm infections.
Collapse
Affiliation(s)
- Mohini Bhattacharya
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
- Department of Veterans Affairs, Eastern Colorado Health Care System, Aurora, CO 80045, United States
| |
Collapse
|
13
|
Han S, Wang Y, Wang L, Chang W, Wen B, Fang J, Hou X, Qi X, Wang J. Mycoplasma synoviae LP78 is a fibronectin/plasminogen binding protein, putative adhesion, and potential diagnostic antigen. Front Microbiol 2024; 14:1335658. [PMID: 38264482 PMCID: PMC10803467 DOI: 10.3389/fmicb.2023.1335658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Mycoplasma synoviae (M. synoviae) is one of the major poultry pathogens causing infectious synovitis, airsacculitis, a high incidence of shell breakage, and egg production loss. However, the pathogenesis of M. synoviae remains unclear. Adhesion of mycoplasmas to host cells is a crucial step in infection and colonization. The purpose of this study was to determine the adhesive function of a putative P80 family lipoprotein (LP78) and evaluate its application in the detection of antibodies against M. synoviae. Recombinant LP78 (rLP78) was expressed in the supernatant component of Escherichia coli and mouse anti-rLP78 serum was prepared. Bioinformatic analysis and western blotting results revealed that LP78 was conservative among M. synoviae strains. It was distributed not only in the cytoplasm but also on the membrane of M. synoviae through western blotting and indirect immunofluorescence (IFA). The adherence of M. synoviae to DF-1 cells was significantly inhibited by mouse anti-rLP78 serum (p < 0.01). IFA revealed that rLP78 adhered to DF-1 cells, and this adherence was prevented by mouse anti-rLP78 serum. Furthermore, rLP78 was found to bind to the DF-1 cells membrane proteins in a dose-dependent manner by enzyme-linked immunosorbent assay (ELISA). Screening of DF-1 cells membrane proteins by western blotting showed that proteins with molecular weight of 35-40 kDa and 55-70 kDa bound to rLP78. Moreover, rLP78 was identified to be a fibronectin/plasminogen binding protein. The sensitivity and specificity of rLP78-based iELISA were 85.7 and 94.1%, respectively. The maximum dilution of positive serum (HI titer, 1:128) detected via rLP78-based iELISA was 1:6,400, whereas that detected using a commercial ELISA kit was 1:12,800-1:25,600. Both rLP78-based iELISA and the commercial ELISA kit detected seroconversion after 7 days of challenge and immunization. No cross-reactivity with positive sera against other avian pathogens was observed in rLP78-based iELISA. Collectively, these results indicate that LP78 is a fibronectin/plasminogen-binding adhesion protein of M. synoviae and a potential diagnostic antigen. The present study will facilitate a better understanding of the pathogenesis of M. synoviae and the development of new diagnostic.
Collapse
Affiliation(s)
- Shuizhong Han
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ying Wang
- College of Food and Drugs, Luoyang Polytechnic, Luoyang, China
| | - Lizhen Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wenchi Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Bo Wen
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Junyang Fang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xiaolan Hou
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
14
|
Badawi AH, Mohamad NA, Stanslas J, Kirby BP, Neela VK, Ramasamy R, Basri H. In Vitro Blood-Brain Barrier Models for Neuroinfectious Diseases: A Narrative Review. Curr Neuropharmacol 2024; 22:1344-1373. [PMID: 38073104 PMCID: PMC11092920 DOI: 10.2174/1570159x22666231207114346] [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: 09/12/2022] [Revised: 11/04/2022] [Accepted: 11/25/2022] [Indexed: 05/16/2024] Open
Abstract
The blood-brain barrier (BBB) is a complex, dynamic, and adaptable barrier between the peripheral blood system and the central nervous system. While this barrier protects the brain and spinal cord from inflammation and infection, it prevents most drugs from reaching the brain tissue. With the expanding interest in the pathophysiology of BBB, the development of in vitro BBB models has dramatically evolved. However, due to the lack of a standard model, a range of experimental protocols, BBB-phenotype markers, and permeability flux markers was utilized to construct in vitro BBB models. Several neuroinfectious diseases are associated with BBB dysfunction. To conduct neuroinfectious disease research effectively, there stems a need to design representative in vitro human BBB models that mimic the BBB's functional and molecular properties. The highest necessity is for an in vitro standardised BBB model that accurately represents all the complexities of an intact brain barrier. Thus, this in-depth review aims to describe the optimization and validation parameters for building BBB models and to discuss previous research on neuroinfectious diseases that have utilized in vitro BBB models. The findings in this review may serve as a basis for more efficient optimisation, validation, and maintenance of a structurally- and functionally intact BBB model, particularly for future studies on neuroinfectious diseases.
Collapse
Affiliation(s)
- Ahmad Hussein Badawi
- Department of Neurology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Nur Afiqah Mohamad
- Department of Neurology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Centre for Foundation Studies, Lincoln University College, 47301, Petaling Jaya, Selangor, Malaysia
| | - Johnson Stanslas
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Brian Patrick Kirby
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Vasantha Kumari Neela
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Hamidon Basri
- Department of Neurology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| |
Collapse
|
15
|
Liu S, Li Z, Lan S, Hao H, Jin X, Liang J, Baz AA, Yan X, Gao P, Chen S, Chu Y. LppA is a novel plasminogen receptor of Mycoplasma bovis that contributes to adhesion by binding the host extracellular matrix and Annexin A2. Vet Res 2023; 54:107. [PMID: 37978536 PMCID: PMC10657132 DOI: 10.1186/s13567-023-01242-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023] Open
Abstract
Mycoplasma bovis is responsible for various inflammatory diseases in cattle. The prevention and control of M. bovis are complicated by the absence of effective vaccines and the emergence of multidrug-resistant strains, resulting in substantial economic losses worldwide in the cattle industry. Lipoproteins, vital components of the Mycoplasmas cell membrane, are deemed potent antigens for eliciting immune responses in the host upon infection. However, the functions of lipoproteins in M. bovis remain underexplored due to their low sequence similarity with those of other bacteria and the scarcity of genetic manipulation tools for M. bovis. In this study, the lipoprotein LppA was identified in all examined M. bovis strains. Utilizing immunoelectron microscopy and Western blotting, it was observed that LppA localizes to the surface membrane. Recombinant LppA demonstrated dose-dependent adherence to the membrane of embryonic bovine lung (EBL) cells, and this adhesion was inhibited by anti-LppA serum. In vitro binding assays confirmed LppA's ability to associate with fibronectin, collagen IV, laminin, vitronectin, plasminogen, and tPA, thereby facilitating the conversion of plasminogen to plasmin. Moreover, LppA was found to bind and enhance the accumulation of Annexin A2 (ANXA2) on the cell membrane. Disrupting LppA in M. bovis significantly diminished the bacterium's capacity to adhere to EBL cells, underscoring LppA's function as a bacterial adhesin. In conclusion, LppA emerges as a novel adhesion protein that interacts with multiple host extracellular matrix proteins and ANXA2, playing a crucial role in M. bovis's adherence to host cells and dissemination. These insights substantially deepen our comprehension of the molecular pathogenesis of M. bovis.
Collapse
Affiliation(s)
- Shuang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Zhangcheng Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Shimei Lan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Huafang Hao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Xiangrui Jin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Jinjia Liang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Ahmed Adel Baz
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Xinmin Yan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Pengcheng Gao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China
| | - Shengli Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China.
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China.
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China.
- Key Laboratory of Veterinary Etilogoical Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 730046, China.
| |
Collapse
|
16
|
Cruz-Samperio R, Hicks CL, Scott A, Gispert Contamina I, Elani Y, Richardson RJ, Perriman AW. Modular Bioorthogonal Lipid Nanoparticle Modification Platforms for Cardiac Homing. J Am Chem Soc 2023; 145:22659-22670. [PMID: 37812759 PMCID: PMC10591475 DOI: 10.1021/jacs.3c07811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Indexed: 10/11/2023]
Abstract
Lipid nanoparticles (LNPs) are becoming widely adopted as vectors for the delivery of therapeutic payloads but generally lack intrinsic tissue-homing properties. These extracellular vesicle (EV) mimetics can be targeted toward the liver, lung, or spleen via charge modification of their lipid headgroups. Homing to other tissues has only been achieved via covalent surface modification strategies using small-molecule ligands, peptides, or monoclonal antibodies─methods that are challenging to couple with large-scale manufacturing. Herein, we design a novel modular artificial membrane-binding protein (AMBP) platform for the modification of LNPs postformation. The system is composed of two protein modules that can be readily coupled using bioorthogonal chemistry to yield the AMBP. The first is a membrane anchor module comprising a supercharged green fluorescent protein (scGFP) electrostatically conjugated to a dynamic polymer surfactant corona. The second is a functional module containing a cardiac tissue fibronectin homing sequence from the bacterial adhesin CshA. We demonstrate that LNPs modified using the AMBP exhibit a 20-fold increase in uptake by fibronectin-rich C2C12 cells under static conditions and a 10-fold increase under physiologically relevant shear stresses, with no loss of cell viability. Moreover, we show targeted localization of the AMBP-modified LNPs in zebrafish hearts, highlighting their therapeutic potential as a vector for the treatment of cardiac disease and, more generally, as a smart vector.
Collapse
Affiliation(s)
- Raquel Cruz-Samperio
- School
of Cellular and Molecular Medicine, University
of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, U.K.
| | - Corrigan L. Hicks
- School
of Cellular and Molecular Medicine, University
of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, U.K.
| | - Aaron Scott
- School
of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, U.K.
| | | | - Yuval Elani
- Department
of Chemical Engineering, Imperial College
London, South Kensington, London SW7 2AZ, U.K.
| | - Rebecca J. Richardson
- School
of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, U.K.
| | - Adam W. Perriman
- School
of Cellular and Molecular Medicine, University
of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, U.K.
- Research
School of Chemistry, Australian National
University, Canberra ACT 2601, Australia
- John
Curtin School of Medical Research, Australian
National University, Canberra ACT 2601, Australia
| |
Collapse
|
17
|
Vaca DJ, Frenzel F, Ballhorn W, Torres SG, Leisegang MS, Günther S, Bender D, Kraiczy P, Göttig S, Kempf VAJ. Adhesion of human pathogenic bacteria to endothelial cells is facilitated by fibronectin interaction. Microbes Infect 2023; 25:105172. [PMID: 37343664 DOI: 10.1016/j.micinf.2023.105172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 06/07/2023] [Accepted: 06/10/2023] [Indexed: 06/23/2023]
Abstract
Human pathogenic bacteria circulating in the bloodstream need to find a way to interact with endothelial cells (ECs) lining the blood vessels to infect and colonise the host. The extracellular matrix (ECM) of ECs might represent an attractive initial target for bacterial interaction, as many bacterial adhesins have reported affinities to ECM proteins, in particular to fibronectin (Fn). Here, we analysed the general role of EC-expressed Fn for bacterial adhesion. For this, we evaluated the expression levels of ECM coding genes in different ECs, revealing that Fn is the highest expressed gene and thereby, it is highly abundant in the ECM environment of ECs. The role of Fn as a mediator in bacterial cell-host adhesion was evaluated in adhesion assays of Acinetobacter baumannii, Bartonella henselae, Borrelia burgdorferi, and Staphylococcus aureus to ECs. The assays demonstrated that bacteria colocalised with Fn fibres, as observed by confocal laser scanning microscopy. Fn removal from the ECM environment (FN1 knockout ECs) diminished bacterial adherence to ECs in both static and dynamic adhesion assays to varying extents, as evaluated via absolute quantification using qPCR. Interactions between adhesins and Fn might represent the crucial step for the adhesion of human-pathogenic Gram-negative and Gram-positive bacteria targeting the ECs as a niche of infection.
Collapse
Affiliation(s)
- Diana J Vaca
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Fabienne Frenzel
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Wibke Ballhorn
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Sara Garcia Torres
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Matthias S Leisegang
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Parkstraße 1, 61231, Bad Nauheim, Germany.
| | - Daniela Bender
- Federal Institute for Vaccines and Biomedicines, Department of Virology, Paul Ehrlich Institute, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany.
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Stephan Göttig
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| | - Volkhard A J Kempf
- Institute of Medical Microbiology and Infection Control, Goethe University, Paul Ehrlich Straße 40, 60596, Frankfurt, Germany.
| |
Collapse
|
18
|
Wang J, Liang K, Chen L, Su X, Liao D, Yu J, He J. Unveiling the stealthy tactics: mycoplasma's immune evasion strategies. Front Cell Infect Microbiol 2023; 13:1247182. [PMID: 37719671 PMCID: PMC10502178 DOI: 10.3389/fcimb.2023.1247182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/09/2023] [Indexed: 09/19/2023] Open
Abstract
Mycoplasmas, the smallest known self-replicating organisms, possess a simple structure, lack a cell wall, and have limited metabolic pathways. They are responsible for causing acute or chronic infections in humans and animals, with a significant number of species exhibiting pathogenicity. Although the innate and adaptive immune responses can effectively combat this pathogen, mycoplasmas are capable of persisting in the host, indicating that the immune system fails to eliminate them completely. Recent studies have shed light on the intricate and sophisticated defense mechanisms developed by mycoplasmas during their long-term co-evolution with the host. These evasion strategies encompass various tactics, including invasion, biofilm formation, and modulation of immune responses, such as inhibition of immune cell activity, suppression of immune cell function, and resistance against immune molecules. Additionally, antigen variation and molecular mimicry are also crucial immune evasion strategies. This review comprehensively summarizes the evasion mechanisms employed by mycoplasmas, providing valuable insights into the pathogenesis of mycoplasma infections.
Collapse
Affiliation(s)
- Jingyun Wang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Keying Liang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Chen
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaoling Su
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Daoyong Liao
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jianwei Yu
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jun He
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
19
|
Hayashida A, Saeed HN, Zhang F, Song Y, Liu J, Parks WC, Bispo PJM, Park PW. Sulfated motifs in heparan sulfate inhibit Streptococcus pneumoniae adhesion onto fibronectin and attenuate corneal infection. PROTEOGLYCAN RESEARCH 2023; 1:e9. [PMID: 38957622 PMCID: PMC11218895 DOI: 10.1002/pgr2.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/26/2023] [Indexed: 07/04/2024]
Abstract
A large number of bacterial pathogens bind to host extracellular matrix (ECM) components. For example, many Gram-negative and Gram-positive pathogens express binding proteins for fibronectin (FN) on their cell surface. Mutagenesis studies of bacterial FN-binding proteins have demonstrated their importance in pathogenesis in preclinical animal models. However, means to draw on these findings to design therapeutic approaches that specifically target FN-bacteria interactions have not been successful because bacterial pathogens can elaborate several FN-binding proteins and also because FN is an essential protein and likely a nondruggable target. Here we report that select heparan compounds potently inhibit Streptococcus pneumoniae infection of injured corneas in mice. Using intact heparan sulfate (HS) and heparin (HP), heparinase-digested fragments of HS, HP oligosaccharides, and chemically or chemoenzymatically modified heparan compounds, we found that inhibition of S. pneumoniae corneal infection by heparan compounds is not mediated by simple charge effects but by a selective sulfate group. Removal of 2-O-sulfates significantly inhibited the ability of HP to inhibit S. pneumoniae corneal infection, whereas the addition of 2-O-sulfates to heparosan (H) significantly increased H's ability to inhibit bacterial corneal infection. Proximity ligation assays indicated that S. pneumoniae attaches directly to FN fibrils in the corneal epithelial ECM and that HS and HP specifically inhibit this binding interaction in a 2-O-sulfate-dependent manner. These data suggest that heparan compounds containing 2-O-sulfate groups protect against S. pneumoniae corneal infection by inhibiting bacterial attachment to FN fibrils in the subepithelial ECM of injured corneas. Moreover, 2-O-sulfated heparan compounds significantly inhibited corneal infection in immunocompromised hosts, by a clinical keratitis isolate of S. pneumoniae, and also when topically administered in a therapeutic manner. These findings suggest that the administration of nonanticoagulant 2-O-sulfated heparan compounds may represent a plausible approach to the treatment of S. pneumoniae keratitis.
Collapse
Affiliation(s)
- Atsuko Hayashida
- Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Hajirah N. Saeed
- Department of Ophthalmology, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Ophthalmology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Yuefan Song
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Jian Liu
- Division of Medicinal Chemistry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - William C. Parks
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Paulo J. M. Bispo
- Department of Ophthalmology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Pyong Woo Park
- Department of Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Alekseeva MG, Dyakov IN, Bushkova KK, Mavletova DA, Yunes RA, Chernyshova IN, Masalitin IA, Koshenko TA, Nezametdinova VZ, Danilenko VN. Study of the binding of ΔFN3.1 fragments of the Bifidobacterium longum GT15 with TNFα and prevalence of domain-containing proteins in groups of bacteria of the human gut microbiota. MICROBIOME RESEARCH REPORTS 2023; 2:10. [PMID: 38047275 PMCID: PMC10688814 DOI: 10.20517/mrr.2023.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/25/2023] [Accepted: 03/31/2023] [Indexed: 12/05/2023]
Abstract
Aim: This study is mainly devoted to determining the ability of ∆FN3.1 protein fragments of Bifidobacterium (B.) longum subsp. longum GT15, namely two FN3 domains (2D FN3) and a C-terminal domain (CD FN3), to bind to tumor necrosis factor-alpha (TNF-α). Methods: Fragments of the fn3 gene encoding the 2D FN3 and CD FN3 were cloned in Escherichia (E.) coli. In order to assess the binding specificity between 2D FN3 and CD FN3 to TNFα, we employed the previously developed sandwich ELISA system to detect any specific interactions between the purified protein and any of the studied cytokines. The trRosetta software was used to build 3D models of the ∆FN3.1, 2D FN3, and CD FN3 proteins. The detection of polymorphism in the amino acid sequences of the studied proteins and the analysis of human gut-derived bacterial proteins carrying FN3 domains were performed in silico. Results: We experimentally showed that neither 2D FN3 nor CD FN3 alone can bind to TNFα. Prediction of the 3D structures of ΔFN3.1, 2D FN3, and CD FN3 suggested that only ΔFN3.1 can form a pocket allowing binding with TNFα to occur. Polymorphism analysis of amino acid sequences of ΔFN3.1 proteins in B. longum strains uncovered substitutions that can alter the conformation of the spatial structure of the ΔFN3.1 protein. We also analyzed human gut-derived bacterial proteins harboring FN3 domains which allowed us to differentiate between those containing motifs of cytokine receptors (MCRs) in their FN3 domains and those lacking them. Conclusion: Only the complete ∆FN3.1 protein can selectively bind to TNFα. Analysis of 3D models of the 2D FN3, CD FN3, and ΔFN3.1 proteins showed that only the ΔFN3.1 protein is potentially capable of forming a pocket allowing TNFα binding to occur. Only FN3 domains containing MCRs exhibited sequence homology with FN3 domains of human proteins.
Collapse
Affiliation(s)
- Maria G. Alekseeva
- Laboratory of Genetics of Microorganisms, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Ilya N. Dyakov
- Laboratory of Immunoglobulin biosynthesis, Mechnikov Research Institute of Vaccines and Sera, Moscow 105064, Russia
| | - Kristina K. Bushkova
- Laboratory of Immunoglobulin biosynthesis, Mechnikov Research Institute of Vaccines and Sera, Moscow 105064, Russia
| | - Dilara A. Mavletova
- Laboratory of Genetics of Microorganisms, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Roman A. Yunes
- Laboratory of Genetics of Microorganisms, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Irina N. Chernyshova
- Laboratory of Immunoglobulin biosynthesis, Mechnikov Research Institute of Vaccines and Sera, Moscow 105064, Russia
| | - Ilya A. Masalitin
- Laboratory of Immunoglobulin biosynthesis, Mechnikov Research Institute of Vaccines and Sera, Moscow 105064, Russia
| | - Tatiana A. Koshenko
- Laboratory of Genetics of Microorganisms, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Venera Z. Nezametdinova
- Laboratory of Genetics of Microorganisms, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Valery N. Danilenko
- Laboratory of Genetics of Microorganisms, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
- Caspian International School of Medicine, Caspian University, Almaty 050000, Kazakhstan
| |
Collapse
|
21
|
Khoury J, Zhang T, Earle DB, Forrest ML. Accelerated neutral atom beam (ANAB) and gas clustered ion beam (GCIB) treatment of implantable device polymers leads to decreased bacterial attachment in vitro and decreased inflammation in vivo. ENGINEERED REGENERATION 2023. [DOI: 10.1016/j.engreg.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023] Open
|
22
|
Mohindra V, Chowdhury LM, Chauhan N, Paul A, Singh RK, Kushwaha B, Maurya RK, Lal KK, Jena JK. Transcriptome Analysis Revealed Osmoregulation Related Regulatory Networks and Hub Genes in the Gills of Hilsa shad, Tenualosa ilisha, during the Migratory Osmotic Stress. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:161-173. [PMID: 36631626 DOI: 10.1007/s10126-022-10190-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Tenualosa ilisha (Hilsa shad), an anadromous fish, usually inhabits coastal and estuarine waters, and migrates to freshwater for spawning. In this study, large-scale gill transcriptome analyses from three salinity regions, i.e., fresh, brackish and marine water, revealed 3277 differentially expressed genes (DEGs), out of which 232 were found to be common between marine vs freshwater and brackish vs freshwater. These genes were mapped into 54 KEGG Pathways, and the most significant of these were focal adhesion, adherens junction, tight junction, and PI3K-Akt signaling pathways. A total of 24 osmoregulatory genes were found to be differentially expressed in different habitats. The gene members of slc16 and slc2 families showed a dissimilar pattern of expressions, while two claudin genes (cldn11 & cldn10), transmembrane tm56b, and voltage-gated potassium channel gene kcna10 were downregulated in freshwater samples, as compared to that of brackish and marine environment. Protein-protein interaction (PPI) network analysis of 232 DEGs showed 101 genes to be involved in PPI, while fn1 gene was found to be interacting with the highest number of genes (36). Twenty-five hub genes belonged to 12 functional groups, with muscle structure development with seven genes, forming the major group. These results provided valuable information about the genes, potentially involved in the molecular mechanisms regulating water homeostasis in gills, during migration for spawning and low-salinity adaptation in Hilsa shad. These genes may form the basis for the bio-marker development for adaptation to the stress levied by major environmental changes, due to hatchery/culture conditions.
Collapse
Affiliation(s)
- Vindhya Mohindra
- ICAR-National Bureau of Fish Genetic Resources (NBFGR), Canal Ring Road, Dilkusha, Lucknow, 226002, India.
| | - Labrechai Mog Chowdhury
- ICAR-National Bureau of Fish Genetic Resources (NBFGR), Canal Ring Road, Dilkusha, Lucknow, 226002, India
| | - Nishita Chauhan
- ICAR-National Bureau of Fish Genetic Resources (NBFGR), Canal Ring Road, Dilkusha, Lucknow, 226002, India
| | - Alisha Paul
- ICAR-National Bureau of Fish Genetic Resources (NBFGR), Canal Ring Road, Dilkusha, Lucknow, 226002, India
| | - Rajeev Kumar Singh
- ICAR-National Bureau of Fish Genetic Resources (NBFGR), Canal Ring Road, Dilkusha, Lucknow, 226002, India
| | - Basdeo Kushwaha
- ICAR-National Bureau of Fish Genetic Resources (NBFGR), Canal Ring Road, Dilkusha, Lucknow, 226002, India
| | - Rajesh Kumar Maurya
- ICAR-National Bureau of Fish Genetic Resources (NBFGR), Canal Ring Road, Dilkusha, Lucknow, 226002, India
| | - Kuldeep K Lal
- ICAR-National Bureau of Fish Genetic Resources (NBFGR), Canal Ring Road, Dilkusha, Lucknow, 226002, India
| | - J K Jena
- Indian Council of Agricultural Research (ICAR), Krishi Anusandhan Bhawan-II, New Delhi, 110 012, India
| |
Collapse
|
23
|
Wrighton S, Ahnlide VK, André O, Bahnan W, Nordenfelt P. Group A streptococci induce stronger M protein-fibronectin interaction when specific human antibodies are bound. Front Microbiol 2023; 14:1069789. [PMID: 36778879 PMCID: PMC9909010 DOI: 10.3389/fmicb.2023.1069789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023] Open
Abstract
Group A streptococcus (GAS) is a highly adapted, human-specific pathogen that is known to manipulate the immune system through various mechanisms. GAS' M protein constitutes a primary target of the immune system due to its spatial configuration and dominance on the bacterial surface. Antibody responses targeting the M protein have been shown to favor the conserved C region. Such antibodies (Abs) circumvent antigenic escape and efficiently bind to various M types. The ability of GAS to bind to fibronectin (Fn), a high molecular weight glycoprotein of the extracellular matrix, has long been known to be essential for the pathogen's evolutionary success and fitness. However, some strains lack the ability to efficiently bind Fn. Instead, they have been found to additionally bind Fn via the A-B domains of their M proteins. Here, we show that human Abs can induce increased Fn-binding affinity in M proteins, likely by enhancing the weak A-B domain binding. We found that this enhanced Fn binding leads to a reduction in Ab-mediated phagocytosis, indicating that this constitutes a GAS immune escape mechanism. We could show that the Fc domain of Abs is necessary to trigger this phenomenon and that Ab flexibility may also play a key role. We, moreover, saw that our Abs could enhance Fn binding in 3 out of 5 emm type strains tested, belonging to different clades, making it likely that this is a more generalizable phenomenon. Together our results suggest a novel synergistic interplay of GAS and host proteins which ultimately benefits the bacterium.
Collapse
|
24
|
Redistribution of the Novel Clostridioides difficile Spore Adherence Receptor E-Cadherin by TcdA and TcdB Increases Spore Binding to Adherens Junctions. Infect Immun 2023; 91:e0047622. [PMID: 36448839 PMCID: PMC9872679 DOI: 10.1128/iai.00476-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Clostridioides difficile causes antibiotic-associated diseases in humans, ranging from mild diarrhea to severe pseudomembranous colitis and death. A major clinical challenge is the prevention of disease recurrence, which affects nearly ~20 to 30% of the patients with a primary C. difficile infection (CDI). During CDI, C. difficile forms metabolically dormant spores that are essential for recurrence of CDI (R-CDI). In prior studies, we have shown that C. difficile spores interact with intestinal epithelial cells (IECs), which contribute to R-CDI. However, this interaction remains poorly understood. Here, we provide evidence that C. difficile spores interact with E-cadherin, contributing to spore adherence and internalization into IECs. C. difficile toxins TcdA and TcdB lead to adherens junctions opening and increase spore adherence to IECs. Confocal micrographs demonstrate that C. difficile spores associate with accessible E-cadherin; spore-E-cadherin association increases upon TcdA and TcdB intoxication. The presence of anti-E-cadherin antibodies decreased spore adherence and entry into IECs. By enzyme-linked immunosorbent assay (ELISA), immunofluorescence, and immunogold labeling, we observed that E-cadherin binds to C. difficile spores, specifically to the hairlike projections of the spore, reducing spore adherence to IECs. Overall, these results expand our knowledge of how C. difficile spores bind to IECs by providing evidence that E-cadherin acts as a spore adherence receptor to IECs and by revealing how toxin-mediated damage affects spore interactions with IECs.
Collapse
|
25
|
Wang Y, Gu Z, Zhang S, Li P. Complete Genome Sequencing Revealed the Potential Application of a Novel Weizmannia coagulans PL-W Production with Promising Bacteriocins in Food Preservative. Foods 2023; 12:216. [PMID: 36613432 PMCID: PMC9818457 DOI: 10.3390/foods12010216] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/01/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023] Open
Abstract
Weizmannia coagulans is an important potential probiotic with dual characteristics of Bacillus and Lactobacillus. This study describes a novel Weizmannia coagulans PL-W with excellent antibacterial activity isolated from Mongolian traditional cheese, in which safety and probiotic potential were evaluated by complete genome sequencing. The crude bacteriocins of W. coagulans PL-W showed antibacterial activity against various foodborne pathogens, including Listeria monocytogenes CMCC 54,004, Bacillus cereus ATCC 14,579, and Staphylococcus aureus ATCC 25,923. Moreover, the crude bacteriocins have outstanding stability against pH, temperature, surfactants, and are sensitive to protease. The complete genome sequencing revealed W. coagulans PL-W consists of 3,666,052-base pair (bp) circular chromosomes with a GC content of 46.24% and 3485 protein-coding genes. It contains 84 tRNA, 10 23S rRNA, 10 16S rRNA, and 10 5S rRNA. In addition, no risk-related genes such as acquired antibiotic resistance genes, virulence, and pathogenic factors were identified, demonstrating that W. coagulans PL-W is safe to use. Furthermore, the presence of gene clusters involved in bacteriocin synthesis, adhesion-related genes, and genes contributing to acid and bile tolerance indicate that W. coagulans PL-W is a potential candidate probiotic. Thus, antimicrobial activity and genome characterization of W. coagulans PL-W demonstrate that it has extensive potential applications as a food protective culture.
Collapse
Affiliation(s)
| | | | | | - Pinglan Li
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
26
|
Hu Z, Li H, Zhao Y, Wang G, Shang Y, Chen Y, Wang S, Tian M, Qi J, Yu S. NADH oxidase of Mycoplasma synoviae is a potential diagnostic antigen, plasminogen/fibronectin binding protein and a putative adhesin. BMC Vet Res 2022; 18:455. [PMID: 36581820 PMCID: PMC9798693 DOI: 10.1186/s12917-022-03556-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Mycoplasma synoviae (MS) is an important pathogen causing respiratory diseases and arthritis in chickens and turkeys, thus, resulting in serious economic losses to the poultry industry. Membrane-associated proteins are thought to play important roles in cytoadherence and pathogenesis. NADH oxidase (NOX) is an oxidoreductase involved in glycolysis, which is thought to be a multifunctional protein and potential virulence factor in some pathogens. However, little is known regarding the NOX of MS (MSNOX). We previously demonstrated that MSNOX was a metabolic enzyme distributed in not only the cytoplasm but also the MS membrane. This study was aimed at exploring NOX's potential as a diagnostic antigen and its role in MS cytoadherence. RESULTS Western blots and ELISAs indicated that recombinant MSNOX (rMSNOX) protein reacted with sera positive for various MS isolates, but not MG isolates or other avian pathogens, thus, suggesting that rMSNOX is a potential diagnostic antigen. In addition, rabbit anti-rMSNOX serum showed substantial complement-dependent mycoplasmacidal activity toward various MS isolates and MG Rlow. MSNOX protein was found not only in the cytoplasm but also on the membrane of MS through suspension immunofluorescence and immunogold electron microscopy assays. Indirect immunofluorescence assays indicated that rMSNOX adhered to DF-1 cells, and this adherence was inhibited by rabbit anti-rMSNOX, but not anti-MG serum. Furthermore, indirect immunofluorescence and colony counting assays confirmed that the rabbit anti-rMSNOX serum inhibited the adherence of various MS isolates but not MG Rlow to DF-1 cells. Moreover, plasminogen (Plg)- and fibronectin (Fn)-binding assays demonstrated that rMSNOX bound Plg and Fn in a dose-dependent manner, thereby further confirming that MSNOX may be a putative adhesin. CONCLUSION MSNOX was identified to be a surface immunogenic protein that has good immunoreactivity and specificity in Western blot and ELISA, and therefore, may be used as a potential diagnostic antigen in the future. In addition, rMSNOX adhered to DF-1 cells, an effect inhibited by rabbit anti-rMSNOX, but not anti-MG serum, and anti-rMSNOX serum inhibited the adherence of various MS isolates, but not MG Rlow, to DF-1 cells, thus indicating that the inhibition of adherence by anti-MSNOX serum was MS specific. Moreover, rMSNOX adhered to extracellular matrix proteins including Plg and Fn, thus suggesting that NOX may play important roles in MS cytoadherence and pathogenesis. Besides, rabbit anti-rMSNOX serum presented complement-dependent mycoplasmacidal activity toward both MS and MG, indicating the MSNOX may be further studied as a potential protective vaccine candidate.
Collapse
Affiliation(s)
- Zengjin Hu
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China ,grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiangxilu, Hefei, Anhui 230061 People’s Republic of China
| | - Haoran Li
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China ,grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiangxilu, Hefei, Anhui 230061 People’s Republic of China
| | - Yuxin Zhao
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China ,grid.268415.cCollege of Veterinary Medicine, Yangzhou University, No. 88 University South Road, Yangzhou, Jiangsu 225009 People’s Republic of China
| | - Guijun Wang
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiangxilu, Hefei, Anhui 230061 People’s Republic of China
| | - Yuanbing Shang
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Yuetong Chen
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Shaohui Wang
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Mingxing Tian
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Jingjing Qi
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Shengqing Yu
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| |
Collapse
|
27
|
Xu QY, Pan Q, Wu Q, Xin JQ. Mycoplasma Bovis adhesins and their target proteins. Front Immunol 2022; 13:1016641. [PMID: 36341375 PMCID: PMC9630594 DOI: 10.3389/fimmu.2022.1016641] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
Bovine mycoplasmosis is an important infectious disease of cattle caused by Mycoplasma bovis (M. bovis) which poses a serious threat to the breeding industry. Adhesin is involved in the initial process of M. bovis colonization, which is closely related to the infection, cell invasion, immune escape and virulence of this pathogenic microorganism. For the reason that M. bovis lacks a cell wall, its adhesin is predominantly located on the surface of the cell membrane. The adhesins of M. bovis are usually identified by adhesion and adhesion inhibition analysis, and more than 10 adhesins have been identified so far. These adhesins primarily bind to plasminogen, fibronectin, heparin and amyloid precursor-like protein-2 of host cells. This review aims to concisely summarize the current knowledge regarding the adhesins of M. bovis and their target proteins of the host cell. Additionally, the biological characteristics of the adhesin will be briefly analyzed.
Collapse
|
28
|
Mondal R, Saldaña-Ahuactzi Z, Soria-Bustos J, Schultz A, Yañez-Santos JA, Laguna YM, Cedillo-Ramírez ML, Girón JA. The EcpD Tip Adhesin of the Escherichia coli Common Pilus Mediates Binding of Enteropathogenic E. coli to Extracellular Matrix Proteins. Int J Mol Sci 2022; 23:ijms231810350. [PMID: 36142263 PMCID: PMC9499635 DOI: 10.3390/ijms231810350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/04/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
The attachment of enteropathogenic Escherichia coli (EPEC) to intestinal epithelial cells is facilitated by several adhesins; however, the individual host-cell receptors for pili-mediated adherence have not been fully characterized. In this study, we evaluated the hypothesis that the E. coli common pilus (ECP) tip adhesin protein EcpD mediates attachment of EPEC to several extracellular matrix (ECM) glycoproteins (fibronectin, laminin, collagens I and IV, and mucin). We found that the ΔecpA mutant, which lacks production of the EcpA filament but retains EcpD on the surface, adhered to these glycoproteins below the wild-type levels, while the ΔecpD mutant, which does not display EcpA or EcpD, bound significantly less to these host glycoproteins. In agreement, a purified recombinant EcpD subunit bound significantly more than EcpA to laminin, fibronectin, collagens I and IV, and mucin in a dose-dependent manner. These are compelling data that strongly suggest that ECP-producing EPEC may bind to host ECM glycoproteins and mucins through the tip adhesin protein EcpD. This study highlights the versatility of EPEC to bind to different host proteins and suggests that the interaction of ECP with the host’s ECM glycoproteins may facilitate colonization of the intestinal mucosal epithelium.
Collapse
Affiliation(s)
- Rajesh Mondal
- ICMR-Bhopal Memorial Hospital and Research Center, Bhopal 462038, India
| | - Zeus Saldaña-Ahuactzi
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Jorge Soria-Bustos
- Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21100, Mexico
| | - Andrew Schultz
- Department of Microbiology and Molecular Genetics, University of Florida, Gainesville, FL 32611, USA
| | - Jorge A. Yañez-Santos
- Facultad de Estomatología, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico
| | - Ygnacio Martínez Laguna
- Centro de Investigación en Ciencias Microbiológicas, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico
| | - María L. Cedillo-Ramírez
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico
| | - Jorge A. Girón
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico
- Correspondence:
| |
Collapse
|
29
|
Dietrich MA, Adamek M, Teitge F, Teich L, Jung-Schroers V, Malinowska A, Świderska B, Rakus K, Kodzik N, Chadzińska M, Karol H, Liszewska E, Ciereszko A. Proteomic analysis of carp seminal plasma provides insights into the immune response to bacterial infection of the male reproductive system. FISH & SHELLFISH IMMUNOLOGY 2022; 127:822-835. [PMID: 35840052 DOI: 10.1016/j.fsi.2022.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023]
Abstract
Aeromonas salmonicida is recognized as a significant bacterial pathogen in ulcerative disease of cyprinid fish. However, the mechanism of immunity to these bacteria in common carp is still not well understood, especially the immune regulation in the gonad to bacterial infection. The aims of our study were to analyze changes in the seminal plasma proteome following A. salmonicida infection in carp males. The observed pathological changes in the tissue (liver, spleen, kidney and testis) morphology and upregulation of immune-related genes (tnfa2, il6a) confirmed the successful infection challenge. Using mass spectrometry-based label-free quantitative proteomics, we identified 1402 seminal plasma proteins, and 44 proteins (20 up- and 24 downregulated) were found to be differentially abundant between infected and control males. Most differentially abundant proteins were involved in the immune response mechanisms, such as acute phase response, complement activation and coagulation, inflammation, lipid metabolism, cell-cell and cell-matrix adhesion, creatine-phosphate biosynthesis and germ cell-Sertoli cell junction signaling. Bacterial infection also caused profound changes in expression of selected genes in the testis and hematopoietic organs, which contributed to changes in seminal proteins. The altered seminal proteins and bacterial proteins in seminal plasma may serve as valuable markers of infection in the testis.
Collapse
Affiliation(s)
- Mariola A Dietrich
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - Mikołaj Adamek
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559, Hannover, Germany
| | - Felix Teitge
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559, Hannover, Germany
| | - Lukas Teich
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559, Hannover, Germany
| | - Verena Jung-Schroers
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559, Hannover, Germany
| | - Agata Malinowska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Pawinskiego 5a, 02-106, Warszawa, Poland
| | - Bianka Świderska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Pawinskiego 5a, 02-106, Warszawa, Poland
| | - Krzysztof Rakus
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Natalia Kodzik
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Magdalena Chadzińska
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Halina Karol
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Ewa Liszewska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Andrzej Ciereszko
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| |
Collapse
|
30
|
Sibanda T, Buys EM. Listeria monocytogenes Pathogenesis: The Role of Stress Adaptation. Microorganisms 2022; 10:microorganisms10081522. [PMID: 36013940 PMCID: PMC9416357 DOI: 10.3390/microorganisms10081522] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 12/13/2022] Open
Abstract
Adaptive stress tolerance responses are the driving force behind the survival ability of Listeria monocytogenes in different environmental niches, within foods, and ultimately, the ability to cause human infections. Although the bacterial stress adaptive responses are primarily a necessity for survival in foods and the environment, some aspects of the stress responses are linked to bacterial pathogenesis. Food stress-induced adaptive tolerance responses to acid and osmotic stresses can protect the pathogen against similar stresses in the gastrointestinal tract (GIT) and, thus, directly aid its virulence potential. Moreover, once in the GIT, the reprogramming of gene expression from the stress survival-related genes to virulence-related genes allows L. monocytogenes to switch from an avirulent to a virulent state. This transition is controlled by two overlapping and interlinked transcriptional networks for general stress response (regulated by Sigma factor B, (SigB)) and virulence (regulated by the positive regulatory factor A (PrfA)). This review explores the current knowledge on the molecular basis of the connection between stress tolerance responses and the pathogenesis of L. monocytogenes. The review gives a detailed background on the currently known mechanisms of pathogenesis and stress adaptation. Furthermore, the paper looks at the current literature and theories on the overlaps and connections between the regulatory networks for SigB and PrfA.
Collapse
Affiliation(s)
- Thulani Sibanda
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa;
- Department of Applied Biology and Biochemistry, National University of Science and Technology, Bulawayo P.O. Box AC939, Zimbabwe
| | - Elna M. Buys
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa;
- Correspondence:
| |
Collapse
|
31
|
Khateb H, Sørensen RS, Cramer K, Eklund AS, Kjems J, Meyer RL, Jungmann R, Sutherland DS. The Role of Nanoscale Distribution of Fibronectin in the Adhesion of Staphylococcus aureus Studied by Protein Patterning and DNA-PAINT. ACS NANO 2022; 16:10392-10403. [PMID: 35801826 PMCID: PMC9330902 DOI: 10.1021/acsnano.2c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Staphylococcus aureus is a widespread and highly virulent pathogen that can cause superficial and invasive infections. Interactions between S. aureus surface receptors and the extracellular matrix protein fibronectin mediate the bacterial invasion of host cells and is implicated in the colonization of medical implant surfaces. In this study, we investigate the role of distribution of both fibronectin and cellular receptors on the adhesion of S. aureus to interfaces as a model for primary adhesion at tissue interfaces or biomaterials. We present fibronectin in patches of systematically varied size (100-1000 nm) in a background of protein and bacteria rejecting chemistry based on PLL-g-PEG and studied S. aureus adhesion under flow. We developed a single molecule imaging assay for localizing fibronectin binding receptors on the surface of S. aureus via the super-resolution DNA points accumulation for imaging in nanoscale topography (DNA-PAINT) technique. Our results indicate that S. aureus adhesion to fibronectin biointerfaces is regulated by the size of available ligand patterns, with an adhesion threshold of 300 nm and larger. DNA-PAINT was used to visualize fibronectin binding receptor organization in situ at ∼7 nm localization precision and with a surface density of 38-46 μm-2, revealing that the engagement of two or more receptors is required for strong S. aureus adhesion to fibronectin biointerfaces.
Collapse
Affiliation(s)
- Heba Khateb
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
| | - Rasmus S. Sørensen
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
| | - Kimberly Cramer
- Max
Planck Institute of Biochemistry, Martinsried 82152, Germany
| | | | - Jorgen Kjems
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
- Department
of Molecular Biology and Genetics Aarhus
University Aarhus
C 8000, Denmark
| | - Rikke L. Meyer
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
| | - Ralf Jungmann
- Max
Planck Institute of Biochemistry, Martinsried 82152, Germany
- Faculty
of Physics and Center for Nanoscience, Ludwig
Maximilian University, Munich 80539, Germany
| | - Duncan S. Sutherland
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
| |
Collapse
|
32
|
Nathawat R, Maku RV, Patel HK, Sankaranarayanan R, Sonti RV. Role of the FnIII domain associated with a cell wall-degrading enzyme cellobiosidase of Xanthomonas oryzae pv. oryzae. MOLECULAR PLANT PATHOLOGY 2022; 23:1011-1021. [PMID: 35278018 PMCID: PMC9190976 DOI: 10.1111/mpp.13205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Cellobiosidase (CbsA) is an important secreted virulence factor of Xanthomonas oryzae pv. oryzae (Xoo), which causes bacterial blight of rice. CbsA is one of several cell wall-degrading enzymes secreted by Xoo via the type II secretion system (T2SS). CbsA is considered a fundamental virulence factor for vascular pathogenesis. CbsA has an N-terminal glycosyl hydrolase domain and a C-terminal fibronectin type III (FnIII) domain. Interestingly, the secreted form of CbsA lacks the FnIII domain during in planta growth. Here we show that the presence of the FnIII domain inhibits the enzyme activity of CbsA on polysaccharide substrates like carboxymethylcellulose. The FnIII domain is required for the interaction of CbsA with SecB chaperone, and this interaction is crucial for the stability and efficient transport of CbsA across the inner membrane. Deletion of the FnIII domain reduced virulence similar to ΔcbsA Xoo, which corroborates the importance of the FnIII domain in CbsA. Our work elucidates a hitherto unknown function of the FnIII domain in enabling the virulence-promoting activity of CbsA.
Collapse
Affiliation(s)
| | - Roshan V. Maku
- CSIR – Centre for Cellular and Molecular BiologyHyderabadIndia
- Present address:
DBT – National Institute of Animal BiotechnologyHyderabadIndia
| | | | | | - Ramesh V. Sonti
- CSIR – Centre for Cellular and Molecular BiologyHyderabadIndia
- Present address:
Indian Institute of Science Education and Research TirupatiTirupatiIndia
| |
Collapse
|
33
|
Interaction of Bartonella henselae with Fibronectin Represents the Molecular Basis for Adhesion to Host Cells. Microbiol Spectr 2022; 10:e0059822. [PMID: 35435766 PMCID: PMC9241615 DOI: 10.1128/spectrum.00598-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Deciphering the mechanisms of bacterial host cell adhesion is a clue for preventing infections. We describe the underestimated role that the extracellular matrix protein fibronectin plays in the adhesion of human-pathogenic
Bartonella henselae
to host cells.
Collapse
|
34
|
Hao F, Xie X, Feng Z, Chen R, Wei Y, Liu J, Xiong Q, Shao G, Lin J. NADH oxidase of Mycoplasma hyopneumoniae functions as a potential mediator of virulence. BMC Vet Res 2022; 18:126. [PMID: 35366872 PMCID: PMC8976378 DOI: 10.1186/s12917-022-03230-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/25/2022] [Indexed: 12/03/2022] Open
Abstract
Background Mycoplasma hyopneumoniae (M. hyopneumoniae) is the etiological agent of enzootic pneumonia, a highly infectious swine respiratory disease that distributed worldwide. The pathogenesis and virulence factors of M. hyopneumoniae are not fully clarified. As an important virulence factor of bacteria, nicotinamide adenine dinucleotide (NADH) oxidase (NOX) participates in host-pathogen interaction, however, the function of NOX involved in the pathogenesis of M. hyopneumoniae is not clear. Results In this study, significant differences in NOX transcription expression levels among different strains of M. hyopneumoniae differed in virulence were identified, suggesting that NOX may be correlated with M. hyopneumoniae virulence. The nox gene of M. hyopneumoniae was cloned and expressed in Escherichia coli, and polyclonal antibodies against recombinant NOX (rNOX) were prepared. We confirmed the enzymatic activity of rNOX based on its capacity to oxidize NADH to NAD+. Flow cytometry analysis demonstrated the surface localization of NOX, and subcellular localization analysis further demonstrated that NOX exists in both the cytoplasm and cell membrane. rNOX was depicted to mediate adhesion to immortalized porcine bronchial epithelial cells (hTERT-PBECs). Pre-neutralizing M. hyopneumoniae with anti-rNOX antibody resulted in a more than 55% reduction in the adhesion rate of high- and low-virulence M. hyopneumoniae strains to hTERT-PBECs. Moreover, a significant difference appeared in the decline in CCU50 titer between virulent (168) and virulence-attenuated (168L) strains. NOX not only recognized and interacted with host fibronectin but also induced cellular oxidative stress and apoptosis in hTERT-PBECs. The release of lactate dehydrogenase by NOX in hTERT-PBECs was positively correlated with the virulence of M. hyopneumoniae strains. Conclusions NOX is considered to be a potential virulence factor of M. hyopneumoniae and may play a significant role in mediating its pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-022-03230-7.
Collapse
|
35
|
Huang R, Wu F, Zhou Q, Wei W, Yue J, Xiao B, Luo Z. Lactobacillus and intestinal diseases: mechanisms of action and clinical applications. Microbiol Res 2022; 260:127019. [DOI: 10.1016/j.micres.2022.127019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
|
36
|
Lam M, Falentin-Daudré C. Characterization of plasmatic proteins adsorption on poly(styrene sodium sulfonate) functionalized silicone surfaces. Biophys Chem 2022; 285:106804. [PMID: 35339945 DOI: 10.1016/j.bpc.2022.106804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/28/2022]
Abstract
Proteins adsorption occurs spontaneously on biomaterial upon insertion within the body. The resulting protein layer influences biomaterial biocompatibility through enhanced bio-integration or, on the contrary, adverse reactions. Furthermore, upon adsorption, proteins can undergo modifications of their structure and, ultimately, their physicochemical properties and activity. Hence, the understanding of protein adsorption on implanted materials appears essential, as exemplified by silicone breast prostheses that might lead to serious health issues. Surface modifications with a bioactive polymer, poly(styrene sodium sulfonate)-polyNaSS, on a hydrophobic silicone surface that composes breast implants, have been successfully performed under UV irradiation by a radical surface polymerization. This strategy enhances cell biocompatibility and antibacterial features. Although detailed insights related to the mechanism are still scarce, polyNaSS is supposed to promote changes in the conformation and/or orientation of adsorbed plasma proteins, reducing the odd for a biofilm to form. The present work addresses more in-depth structural investigations of the adsorbed state of two plasma proteins: Bovine Serum Albumin (BSA), as a model protein, and fibronectin (FN), for its role in cell adhesion. Using Atomic force microscopy (AFM), we report that polyNaSS showed no significant impact on the BSA structure conversely to the FN one. However, imaging findings with AFM clearly outlined a change in the structural organization of FN, going from a nano fibrillar assembly with an average length of 130 nm to a globular one when the surface was grafted. Thus, it is highlighted that polyNaSS interacts specifically with FN. In addition, cell spreading assay of L929 fibroblasts on FN-coated surfaces with optical microscopy indicated no significant impact of the change in FN structure upon fibroblasts adhesion, which displayed active elongated shapes. The present features are crucial for understanding the cell adhesion mechanism induced by surface modification.
Collapse
Affiliation(s)
- M Lam
- LBPS/CSPBAT, UMR CNRS 7244, Institut Galilée, Université Sorbonne Paris Nord, 99 avenue JB Clément, 93430 Villetaneuse, France
| | - C Falentin-Daudré
- LBPS/CSPBAT, UMR CNRS 7244, Institut Galilée, Université Sorbonne Paris Nord, 99 avenue JB Clément, 93430 Villetaneuse, France.
| |
Collapse
|
37
|
Srivastava A, Mohan S, Davies KG. Exploring Bacillus thuringiensis as a model for endospore adhesion and its potential to investigate adhesins in Pasteuria penetrans. J Appl Microbiol 2022; 132:4371-4387. [PMID: 35286009 PMCID: PMC9311801 DOI: 10.1111/jam.15522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 11/30/2022]
Abstract
Aims Phytonematodes are a constraint on crop production and have been controlled using nematicides; these are highly toxic and legislation in Europe and elsewhere is prohibiting their use and alternatives are being sought. Pasteuria penetrans is a hyperparasitic bacterium that form endospores and have potential to control root‐knot nematodes (Meloidogyne spp.), but their attachment to the nematode cuticle is host‐specific. Understanding host specificity has relied upon endospore inhibition bioassays using immunological and biochemical approaches. Phylogenetic analysis of survey sequences has shown P. penetrans to be closely related to Bacillus and to have a diverse range of collagen‐like fibres which we hypothesise to be involved in the endospore adhesion. However, due to the obligately hyperparasitic nature of Pasteuria species, identifying and characterizing these collagenous‐like proteins through gain of function has proved difficult and new approaches are required. Methods and Results Using antibodies raised to synthetic peptides based on Pasteuria collagen‐like genes we show similarities between P. penetrans and the more easily cultured bacterium Bacillus thuringiensis and suggest it be used as a gain of function platform/model. Using immunological approaches similar proteins between P. penetrans and B. thuringiensis are identified and characterized, one >250 kDa and another ~72 kDa are glycosylated with N‐acetylglucosamine and both of which are digested if treated with collagenase. These treatments also affected endospore attachment and suggest these proteins are involved in adhesion of endospores to nematode cuticle. Conclusion There are conserved similarities in the collagen‐like proteins present on the surface of endospores of both P. penetrans and B. thuringiensis. Significance and Impact of Study As B. thuringiensis is relatively easy to culture and can be transformed, it could be developed as a platform for studying the role of the collagen‐like adhesins from Pasteuria in endospore adhesion.
Collapse
Affiliation(s)
- Arohi Srivastava
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, AL10 9AB, UK
| | - Sharad Mohan
- Division of Nematology, Indian Agricultural Research Institute, Pusa Campus, New Delhi110012, India
| | - Keith G Davies
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, AL10 9AB, UK
| |
Collapse
|
38
|
Kosik K, Sowińska A, Seremak-Mrozikiewicz A, Abu-Amara JA, Al-Saad SR, Karbowski LM, Gryczka K, Kurzawińska G, Szymankiewicz-Bręborowicz M, Drews K, Szpecht D. Polymorphisms of fibronectin-1 (rs3796123; rs1968510; rs10202709; rs6725958; and rs35343655) are not associated with bronchopulmonary dysplasia in preterm infants. Mol Cell Biochem 2022; 477:1645-1652. [PMID: 35230604 DOI: 10.1007/s11010-022-04397-1] [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: 10/06/2021] [Accepted: 02/16/2022] [Indexed: 11/27/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease that mainly affects premature newborns. Many different factors, increasingly genetic, are involved in the pathogenesis of BPD. The aim of the study is to investigate the possible influence of fibronectin SNP on the occurrence of BPD. The study included 108 infants born between 24 and 32 weeks of gestation. BPD was diagnosed based on the National Institutes of Health Consensus definition. The 5 FN1 gene polymorphisms assessed in the study were the following: rs3796123; rs1968510; rs10202709; rs6725958; and rs35343655. BPD developed in 30 (27.8%) out of the 108 preterm infants. Incidence of BPD was higher in infants with lower APGAR scores and low birthweight. Investigation did not confirm any significant prevalence for BPD development in any genotypes and alleles of FN1. Further studies should be performed to confirm the role of genetic factors in etiology and pathogenesis of BPD.
Collapse
Affiliation(s)
- Katarzyna Kosik
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Anna Sowińska
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | | - Katarzyna Gryczka
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Grażyna Kurzawińska
- Department of Perinatology and Women's Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Krzysztof Drews
- Department of Perinatology and Women's Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Dawid Szpecht
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
39
|
Development of a Phage Cocktail to Target Salmonella Strains Associated with Swine. Pharmaceuticals (Basel) 2022; 15:ph15010058. [PMID: 35056115 PMCID: PMC8777603 DOI: 10.3390/ph15010058] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023] Open
Abstract
Infections caused by multidrug resistant Salmonella strains are problematic in swine and are entering human food chains. Bacteriophages (phages) could be used to complement or replace antibiotics to reduce infection within swine. Here, we extensively characterised six broad host range lytic Salmonella phages, with the aim of developing a phage cocktail to prevent or treat infection. Intriguingly, the phages tested differed by one to five single nucleotide polymorphisms. However, there were clear phenotypic differences between them, especially in their heat and pH sensitivity. In vitro killing assays were conducted to determine the efficacy of phages alone and when combined, and three cocktails reduced bacterial numbers by ~2 × 103 CFU/mL within two hours. These cocktails were tested in larvae challenge studies, and prophylactic treatment with phage cocktail SPFM10-SPFM14 was the most efficient. Phage treatment improved larvae survival to 90% after 72 h versus 3% in the infected untreated group. In 65% of the phage-treated larvae, Salmonella counts were below the detection limit, whereas it was isolated from 100% of the infected, untreated larvae group. This study demonstrates that phages effectively reduce Salmonella colonisation in larvae, which supports their ability to similarly protect swine.
Collapse
|
40
|
Qi J, Wang Y, Li H, Shang Y, Gao S, Ding C, Liu X, Wang S, Li T, Tian M, Yu S. Mycoplasma synoviae dihydrolipoamide dehydrogenase is an immunogenic fibronectin/plasminogen binding protein and a putative adhesin. Vet Microbiol 2021; 265:109328. [PMID: 35032790 DOI: 10.1016/j.vetmic.2021.109328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 11/23/2020] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
Abstract
Mycoplasma synoviae (M. synoviae) is an important avian pathogen that causes arthritis and airsacculitis in young chickens and turkeys. Infection by M. synoviae results in considerable economic losses to the poultry industry worldwide. Cytoadherence is a crucial stage during mycoplasma infection. Dihydrolipoamide dehydrogenase (PdhD) is a flavin-dependent enzyme that is critical for energy metabolism and redox balance. To date, its role in cytoadherence is poorly understood. In this study, recombinant PdhD from M. synoviae (rMSPdhD) was expressed in the supernatant component of E. coli BL21 and rabbit anti-rMSPdhD serum was prepared. rMSPdhD was shown to be an immunogenic protein by immunoblot assays, while the mycoplasmacidal assay revealed that the rabbit anti-rMSPdhD serum had a high complement-dependent mycoplasmacidal rate (88.5 %). Using a suspension immunofluorescence assay and subcellular localization analysis, MSPdhD was shown to be a surface-localized protein distributed in both the cytoplasm and cell membrane of M. synoviae. The enzymatic activity of rMSPdhD was determined by measuring its ability to reduce lipoamide to dihydrolipoamide and convert NADH to NAD+. Using an indirect immunofluorescence assay, rMSPdhD was shown to adhere to DF-1 chicken embryo fibroblast cells. Furthermore, the attachment of M. synoviae to DF-1 cells was significantly inhibited by rabbit anti-rMSPdhD serum. Western blot and ELISA binding assays confirmed that rMSPdhD also bound to fibronectin (Fn) and plasminogen (Plg) in a dose-dependent manner. In conclusion, our data show that MSPdhD is not only a biological enzyme, but also an immunogenic surface-exposed protein that can bind to Fn and Plg as well as adhere to host cells. In addition, we show that rabbit anti-rMSPdhD serum can inhibit the adhesion of M. synoviae to DF-1 cells and has a significant complement-dependent bactericidal activity. Our findings suggest that MSPdhD may be involved in the pathogenesis of M. synoviae.
Collapse
Affiliation(s)
- Jingjing Qi
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China
| | - Yu Wang
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China; College of Veterinary Medicine, Yangzhou University, No. 88 University South Road, Yangzhou, Jiangsu 225009, PR China
| | - Haoran Li
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China; College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiangxilu, Hefei, Anhui, PR China
| | - Yuanbing Shang
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China; College of Animal Husbandry and Veterinary Medicine, Jin Zhou Medical University, No. 40 Section 3 Songpo Road, Linghe District, Jinzhou City, Liaoning 121001, PR China
| | - Song Gao
- College of Veterinary Medicine, Yangzhou University, No. 88 University South Road, Yangzhou, Jiangsu 225009, PR China
| | - Chan Ding
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China
| | - Xiaohan Liu
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China
| | - Shaohui Wang
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China
| | - Tao Li
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China
| | - Mingxing Tian
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China
| | - Shengqing Yu
- Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241, PR China.
| |
Collapse
|
41
|
Bacterial Outer Membrane Protein OmpX Regulates β1 Integrin and Epidermal Growth Factor Receptor (EGFR) Involved in Invasion of M-HeLa Cells by Serratia proteamaculans. Int J Mol Sci 2021; 22:ijms222413246. [PMID: 34948042 PMCID: PMC8703988 DOI: 10.3390/ijms222413246] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 01/31/2023] Open
Abstract
Opportunistic pathogen Serratia proteamaculans are able to penetrate the eukaryotic cells. The penetration rate can be regulated by bacterial surface protein OmpX. OmpX family proteins are able to bind to host cell surface to the epidermal growth factor receptor (EGFR) and the extracellular matrix protein fibronectin, whose receptors are in return the α5 β1 integrins. Here we elucidated the involvement of these host cell proteins in S. proteamaculans invasion. We have shown that, despite the absence of fibronectin contribution to S. proteamaculans invasion, β1 integrin was directly involved in invasion of M-HeLa cells. Herewith β1 integrin was not the only receptor that determines sensitivity of host cells to bacterial invasion. Signal transfer from EGFR was also involved in the penetration of these bacteria into M-HeLa cells. However, M-HeLa cells have not been characterized by large number of these receptors. It turned out that S. proteamaculans attachment to the host cell surface resulted in an increment of EGFR and β1 integrin genes expression. Such gene expression increment also caused Escherichia coli attachment, transformed with a plasmid encoding OmpX from S. proteamaculans. Thus, an OmpX binding to the host cell surface caused an increase in the EGFR and β1 integrin expression involved in S. proteamaculans invasion.
Collapse
|
42
|
Mehdizadeh Gohari I, A. Navarro M, Li J, Shrestha A, Uzal F, A. McClane B. Pathogenicity and virulence of Clostridium perfringens. Virulence 2021; 12:723-753. [PMID: 33843463 PMCID: PMC8043184 DOI: 10.1080/21505594.2021.1886777] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Clostridium perfringens is an extremely versatile pathogen of humans and livestock, causing wound infections like gas gangrene (clostridial myonecrosis), enteritis/enterocolitis (including one of the most common human food-borne illnesses), and enterotoxemia (where toxins produced in the intestine are absorbed and damage distant organs such as the brain). The virulence of this Gram-positive, spore-forming, anaerobe is largely attributable to its copious toxin production; the diverse actions and roles in infection of these toxins are now becoming established. Most C. perfringens toxin genes are encoded on conjugative plasmids, including the pCW3-like and the recently discovered pCP13-like plasmid families. Production of C. perfringens toxins is highly regulated via processes involving two-component regulatory systems, quorum sensing and/or sporulation-related alternative sigma factors. Non-toxin factors, such as degradative enzymes like sialidases, are also now being implicated in the pathogenicity of this bacterium. These factors can promote toxin action in vitro and, perhaps in vivo, and also enhance C. perfringens intestinal colonization, e.g. NanI sialidase increases C. perfringens adherence to intestinal tissue and generates nutrients for its growth, at least in vitro. The possible virulence contributions of many other factors, such as adhesins, the capsule and biofilms, largely await future study.
Collapse
Affiliation(s)
- Iman Mehdizadeh Gohari
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mauricio A. Navarro
- California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, University of California Davis, San Bernardino, CA, USA
| | - Jihong Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Archana Shrestha
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Francisco Uzal
- California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, University of California Davis, San Bernardino, CA, USA
| | - Bruce A. McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
43
|
Casillas-Ituarte NN, Staats AM, Lower BH, Stoodley P, Lower SK. Host blood proteins as bridging ligand in bacterial aggregation as well as anchor point for adhesion in the molecular pathogenesis of Staphylococcus aureus infections. Micron 2021; 150:103137. [PMID: 34392091 PMCID: PMC8484042 DOI: 10.1016/j.micron.2021.103137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/09/2021] [Accepted: 08/03/2021] [Indexed: 11/20/2022]
Abstract
Fibronectin (Fn) and fibrinogen (Fg) are major host proteins present in the extracellular matrix, blood, and coatings on indwelling medical devices. The ability of Staphylococcus aureus to cause infections in humans depends on favorable interactions with these host ligands. Closely related bacterial adhesins, fibronectin-binding proteins A and B (FnBPA, FnBPB) were evaluated for two key steps in pathogenesis: clumping and adhesion. Experiments utilized optical spectrophotometry, flow cytometry, and atomic force microscopy to probe FnBPA/B alone or in combination in seven different strains of S. aureus and Lactococcus lactis, a Gram-positive surrogate that naturally lacks adhesins to mammalian ligands. In the absence of soluble ligands, both FnBPA and FnBPB were capable of interacting with adjacent FnBPs from neighboring bacteria to mediate clumping. In the presence of soluble host ligands, clumping was enhanced particularly under shear stress and with Fn present in the media. FnBPB exhibited greater ability to clump compared to FnBPA. The strength of adhesion was similar for immobilized Fn to FnBPA and FnBPB. These findings suggest that these two distinct but closely related bacterial adhesins, have different functional capabilities to interact with host ligands in different settings (e.g., soluble vs. immobilized). Survival and persistence of S. aureus in a human host may depend on complementary roles of FnBPA and FnBPB as they interact with different conformations of Fn or Fg (compact in solution vs. extended on a surface) present in different physiological spaces.
Collapse
Affiliation(s)
- Nadia N Casillas-Ituarte
- School of Earth Sciences, The Ohio State University, Columbus, OH, 43210, USA; School of Environment and Natural Resources, The Ohio State University, Columbus, OH, 43210, USA.
| | - Amelia M Staats
- Departments of Microbiology and Microbial Infection and Immunity, The Ohio State University, 43210, Columbus, OH, USA
| | - Brian H Lower
- School of Environment and Natural Resources, The Ohio State University, Columbus, OH, 43210, USA
| | - Paul Stoodley
- Departments of Microbiology and Microbial Infection and Immunity, The Ohio State University, 43210, Columbus, OH, USA; Department of Orthopaedics, The Ohio State University, Columbus, OH, 43210, USA
| | - Steven K Lower
- School of Earth Sciences, The Ohio State University, Columbus, OH, 43210, USA; School of Environment and Natural Resources, The Ohio State University, Columbus, OH, 43210, USA; Departments of Microbiology and Microbial Infection and Immunity, The Ohio State University, 43210, Columbus, OH, USA
| |
Collapse
|
44
|
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
|
45
|
Shi Y, Berking A, Baade T, Legate KR, Fässler R, Hauck CR. PIP5KIγ90-generated phosphatidylinositol-4,5-bisphosphate promotes the uptake of Staphylococcus aureus by host cells. Mol Microbiol 2021; 116:1249-1267. [PMID: 34519119 DOI: 10.1111/mmi.14807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022]
Abstract
Staphylococcus aureus, a Gram-positive pathogen, invades cells mainly in an integrin-dependent manner. As the activity or conformation of several integrin-associated proteins can be regulated by phosphatidylinositol-4,5-bisphosphate (PI-4,5-P2 ), we investigated the roles of PI-4,5-P2 and PI-4,5-P2 -producing enzymes in cellular invasion by S. aureus. PI-4,5-P2 accumulated upon contact of S. aureus with the host cell, and targeting of an active PI-4,5-P2 phosphatase to the plasma membrane reduced bacterial invasion. Knockdown of individual phosphatidylinositol-4-phosphate 5-kinases revealed that phosphatidylinositol-4-phosphate 5-kinase γ (PIP5KIγ) plays an important role in bacterial internalization. Specific ablation of the talin and FAK-binding motif in PIP5KIγ90 reduced bacterial invasion, which could be rescued by reexpression of an active, but not inactive PIP5KIγ90. Furthermore, PIP5KIγ90-deficient cells showed normal basal PI-4,5-P2 levels in the plasma membrane but reduced the accumulation of PI-4,5-P2 and talin at sites of S. aureus attachment and overall lower levels of FAK phosphorylation. These results highlight the importance of local synthesis of PI-4,5-P2 by a focal adhesion-associated lipid kinase for integrin-mediated internalization of S. aureus.
Collapse
Affiliation(s)
- Yong Shi
- Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany
| | - Anne Berking
- Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany
| | - Timo Baade
- Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany.,Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| | | | | | - Christof R Hauck
- Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany.,Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| |
Collapse
|
46
|
Abstract
Translation of the genetic information into proteins, performed by the ribosome, is a key cellular process in all organisms. Translation usually proceeds smoothly, but, unfortunately, undesirable events can lead to stalling of translating ribosomes. To rescue these faulty arrested ribosomes, bacterial cells possess three well-characterized quality control systems, tmRNA, ArfA, and ArfB. Recently, an additional ribosome rescue mechanism has been discovered in Bacillus subtilis. In contrast to the "canonical" systems targeting the 70S bacterial ribosome, this latter mechanism operates by first splitting the ribosome into the small (30S) and large (50S) subunits to then clearing the resultant jammed large subunit from the incomplete nascent polypeptide. Here, I will discuss the recent microbiological, biochemical, and structural data regarding functioning of this novel rescue system.
Collapse
Affiliation(s)
- Maxim S Svetlov
- Center for Biomolecular Sciences, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
47
|
Functional Genomic Insights into Probiotic Bacillus siamensis Strain B28 from Traditional Korean Fermented Kimchi. Foods 2021; 10:foods10081906. [PMID: 34441683 PMCID: PMC8394110 DOI: 10.3390/foods10081906] [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: 06/22/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 01/22/2023] Open
Abstract
Bacillus siamensis strain B28 was previously isolated from traditional Korean fermented kimchi and inhibited expression of the microphthalmia-associated transcription factor and β-catenin in human embryonic kidney 293 cells. Here, we determined the complete genome sequence of strain B28 and compared it with other strains to elucidate its potential probiotic properties. Strain B28 does not contain antibiotic resistance-, hemolysin- or enterotoxin-encoding genes. The genome includes genes related to survival in extreme conditions, adhesion in the gut, and synthesis of the bacteriocin. Considering the potential for enhancement of human health, the strain B28 genome encodes genes related to production of eight essential amino acids, γ-aminobutyric acid, branched-chain fatty acids, γ-glutamyltransferase, and subtilisin. There are genes for the synthesis of uracil, lipoteichoic acid, glutathione, and several reactive oxygen species-scavenging enzymes. Experimentally, strain B28 exhibited sensitivity to eight antibiotics and antibacterial activity against seven foodborne pathogens. B. siamensis B28 is a safe strain with potential for development as a probiotic.
Collapse
|
48
|
Matsunaga N, Narukawa N, Yamasaki T, Katayama S, Hitsumoto Y. Inhibition of the interaction between fibronectin and dermatopontin by Clostridium perfringens fibronectin-binding proteins. Microbiol Immunol 2021; 65:333-341. [PMID: 33991001 DOI: 10.1111/1348-0421.12917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 11/26/2022]
Abstract
Fibronectin (Fn) is an approximately 450 kDa glycoprotein that consists of 12 type I, 2 type II, and 15-17 type III modules. Fibrillation of Fn is important for tissue reconstitution and wound healing. We previously reported that Clostridium perfringens produces several Fn-binding proteins (Fbps), two of which, FbpA and FbpB, bind to III1 -C (a fragment of Fn derived from the carboxyl-terminal two-thirds of the first-type III module). Dermatopontin (DPT), a 22 kDa noncollagenous extracellular matrix protein, accelerates normal collagen fibrillation and induces Fn fibrillation. DPT interacts with Fn-type III12-14 (III12-14 ), leading to a change in Fn conformation and promoting Fn fibrillation. Here, we investigated the effects of FbpA and FbpB on the binding of Fn and the III12-14 fragment to DPT and on the DPT-induced Fn fibrillation. Both recombinant FbpA (rFbpA) and recombinant FbpB (rFbpB) significantly inhibited Fn binding to DPT and recombinant III12-14 (rIII12-14 ) binding, and inhibited DPT-induced Fn fibrillation. Furthermore, it was found that both rFbpA and rFbpB significantly bound to coated DPT in an enzyme-linked avidin-biotin complex system, whereas rIII12-14 did not bind to either coated rFbpA or rFbpB. In conclusion, both FbpA and FbpB inhibited DPT-induced Fn fibrillation via their interaction with DPT. Both FbpA and FbpB released from lysed C. perfringens cells in wounds and/or infected tissue may prevent Fn fibrillation and delay the wound healing process, subsequently exacerbating infection.
Collapse
Affiliation(s)
- Nozomu Matsunaga
- Department of Life Science, Faculty of Science, Okayama University of Science, Okayama, Japan
| | - Nodoka Narukawa
- Department of Life Science, Faculty of Science, Okayama University of Science, Okayama, Japan
| | | | - Seiichi Katayama
- Department of Life Science, Faculty of Science, Okayama University of Science, Okayama, Japan
| | - Yasuo Hitsumoto
- Department of Life Science, Faculty of Science, Okayama University of Science, Okayama, Japan
| |
Collapse
|
49
|
Identification and Characterization of Immunodominant Proteins from Tick Tissue Extracts Inducing a Protective Immune Response against Ixodes ricinus in Cattle. Vaccines (Basel) 2021; 9:vaccines9060636. [PMID: 34200738 PMCID: PMC8229163 DOI: 10.3390/vaccines9060636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 11/16/2022] Open
Abstract
Ixodes ricinus is the main vector of tick-borne diseases in Europe. An immunization trial of calves with soluble extracts of I. ricinus salivary glands (SGE) or midgut (ME) previously showed a strong response against subsequent tick challenge, resulting in diminished tick feeding success. Immune sera from these trials were used for the co-immunoprecipitation of tick tissue extracts, followed by LC-MS/MS analyses. This resulted in the identification of 46 immunodominant proteins that were differentially recognized by the serum of immunized calves. Some of these proteins had previously also drawn attention as potential anti-tick vaccine candidates using other approaches. Selected proteins were studied in more detail by measuring their relative expression in tick tissues and RNA interference (RNAi) studies. The strongest RNAi phenotypes were observed for MG6 (A0A147BXB7), a protein containing eight fibronectin type III domains predominantly expressed in tick midgut and ovaries of feeding females, and SG2 (A0A0K8RKT7), a glutathione-S-transferase that was found to be upregulated in all investigated tissues upon feeding. The results demonstrated that co-immunoprecipitation of tick proteins with host immune sera followed by protein identification using LC-MS/MS is a valid approach to identify antigen–antibody interactions, and could be integrated into anti-tick vaccine discovery pipelines.
Collapse
|
50
|
Potential factors involved in the early pathogenesis of Streptococcus uberis mastitis: a review. Folia Microbiol (Praha) 2021; 66:509-523. [PMID: 34085166 DOI: 10.1007/s12223-021-00879-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
Bovine mastitis is an inflammation of the mammary gland, which could be the result of allergy, physical trauma, or invasion by pathogens as Streptococcus uberis. This pathogen is an environmental pathogen associated with subclinical and clinical intramammary infection (IMI) in both lactating and non-lactating cows, which can persist in the udder and cause a chronic infection in the mammary gland. In spite of the important economic losses and increased prevalence caused by S. uberis mastitis, virulence factors involved in bacterial colonization of mammary glands and the pathogenic mechanisms are not yet clear. In the last 30 years, several studies have defined adherence and internalization of S. uberis as the early stages in IMI. S. uberis adheres to and invades into mammary gland cells, and this ability has been observed in in vitro assays. Until now, these abilities have not been determined in vivo challenges since they have been difficult to study. Bacterial surface proteins are able to bind to extracellular matrix protein components such as fibronectin, collagen and laminin, as well as proteins in milk. These proteins play a role in adhesion to host cells and have been denominated microbial surface components recognizing adhesive matrix molecules (MSCRAMMs). This article aims to summarize our current knowledge on the most relevant properties of the potential factors involved in the early pathogenesis of S. uberis mastitis.
Collapse
|