1
|
Wagner TM, Torres-Puig S, Yimthin T, Irobalieva RN, Heller M, Kaessmeyer S, Démoulins T, Jores J. Extracellular vesicles of minimalistic Mollicutes as mediators of immune modulation and horizontal gene transfer. Commun Biol 2025; 8:674. [PMID: 40301684 DOI: 10.1038/s42003-025-08099-4] [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: 12/02/2024] [Accepted: 04/16/2025] [Indexed: 05/01/2025] Open
Abstract
Extracellular vesicles (EVs) are central components of bacterial secretomes, including the small, cell wall-less Mollicutes. Although EV release in Mollicutes has been reported, EV proteomic composition and function have not been explored yet. We developed a protocol for isolating EVs of the pathogens Mycoplasma mycoides subsp. capri (Mmc) and Mycoplasma (Mycoplasmopsis) bovis and examined their functionality. Proteomic analysis demonstrated that EVs mirror the proteome of the EV-producing bacteria. EVs exhibited nuclease activity, effectively digesting both circular and linear DNA. Notably, M. bovis EVs elicited immune responses in bovine primary blood cells, like those induced by live M. bovis. Our findings reveal that EVs can carry plasmids and enable their horizontal transfer, known as vesiduction. Specifically, the natural plasmid pKMK1, with an unknown transmission route, was detected in EVs of Mmc 152/93 and the tetM-containing pIVB08 plasmid was associated with EVs released by an Mmc GM12 strain carrying this plasmid. pIVB08 could be transferred via homo- and heterologous vesiduction to Mmc, M. capricolum subsp. capricolum and M. leachii. Vesiduction was impeded by membrane disruption but resisted DNase and Proteinase K treatment, suggesting that EVs protect their cargo. These findings enhance our understanding of Mollicutes EVs, particularly in host interactions and horizontal gene transfer.
Collapse
Affiliation(s)
- Theresa Maria Wagner
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland.
| | - Sergi Torres-Puig
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
| | - Thatcha Yimthin
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Rossitza N Irobalieva
- Division of Veterinary Anatomy, Department of Clinical Research and Veterinary Public Health, University of Bern, Bern, Switzerland
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sabine Kaessmeyer
- Division of Veterinary Anatomy, Department of Clinical Research and Veterinary Public Health, University of Bern, Bern, Switzerland
| | - Thomas Démoulins
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
| | - Jörg Jores
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland.
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, Bern, Switzerland.
| |
Collapse
|
2
|
Yacoub E, Baby V, Sirand-Pugnet P, Arfi Y, Mardassi H, Blanchard A, Chibani S, Ben Abdelmoumen Mardassi B. A sweeping view of avian mycoplasmas biology drawn from comparative genomic analyses. BMC Genomics 2025; 26:24. [PMID: 39789465 PMCID: PMC11720521 DOI: 10.1186/s12864-024-11201-5] [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: 10/02/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Avian mycoplasmas are small bacteria associated with several pathogenic conditions in many wild and poultry bird species. Extensive genomic data are available for many avian mycoplasmas, yet no comparative studies focusing on this group of mycoplasmas have been undertaken so far. RESULTS Here, based on the comparison of forty avian mycoplasma genomes belonging to ten different species, we provide insightful information on the phylogeny, pan/core genome, energetic metabolism, and virulence of these avian pathogens. Analyses disclosed considerable inter- and intra-species genomic variabilities, with genome sizes that can vary by twice as much. Phylogenetic analysis based on concatenated orthologous genes revealed that avian mycoplasmas fell into either Hominis or Pneumoniae groups within the Mollicutes and could split into various clusters. No host co-evolution of avian mycoplasmas can be inferred from the proposed phylogenetic scheme. With 3,237 different gene clusters, the avian mycoplasma group under study proved diverse enough to have an open pan genome. However, a set of 150 gene clusters was found to be shared between all avian mycoplasmas, which is likely encoding essential functions. Comparison of energy metabolism pathways showed that avian mycoplasmas rely on various sources of energy. Superposition between phylogenetic and energy metabolism groups revealed that the glycolytic mycoplasmas belong to two distinct phylogenetic groups (Hominis and Pneumoniae), while all the arginine-utilizing mycoplasmas belong only to Hominis group. This can stand for different evolutionary strategies followed by avian mycoplasmas and further emphasizes the diversity within this group. Virulence determinants survey showed that the involved gene arsenals vary significantly within and between species, and could even be found in species often reported apathogenic. Immunoglobulin-blocking proteins were detected in almost all avian mycoplasmas. Although these systems are not exclusive to this group, they seem to present some particular features making them unique among mycoplasmas. CONCLUSION This comparative genomic study uncovered the significant variable nature of avian mycoplasmas, furthering our knowledge on their biological attributes and evoking new hallmarks.
Collapse
Affiliation(s)
- Elhem Yacoub
- Unit of Mycoplasmas, Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Vincent Baby
- Centre de Diagnostic Vétérinaire de L'Université de Montréal (CDVUM), Faculty of Veterinary Medecine, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | | | - Yonathan Arfi
- Univ. Bordeaux, INRAE, UMR BFP, 33882, Villenave d'Ornon, France
| | - Helmi Mardassi
- Unit of Typing and Genetics of Mycobacteria, Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Alain Blanchard
- Univ. Bordeaux, INRAE, UMR BFP, 33882, Villenave d'Ornon, France
| | - Salim Chibani
- Unit of Mycoplasmas, Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Boutheina Ben Abdelmoumen Mardassi
- Unit of Mycoplasmas, Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
3
|
Khan A, Ammar Zahid M, Farrukh F, Salah Abdelsalam S, Mohammad A, Al-Zoubi RM, Shkoor M, Ait Hssain A, Wei DQ, Agouni A. Integrated structural proteomics and machine learning-guided mapping of a highly protective precision vaccine against mycoplasma pulmonis. Int Immunopharmacol 2024; 141:112833. [PMID: 39153303 DOI: 10.1016/j.intimp.2024.112833] [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: 06/13/2024] [Revised: 07/09/2024] [Accepted: 07/27/2024] [Indexed: 08/19/2024]
Abstract
Mycoplasma pulmonis (M. pulmonis) is an emerging respiratory infection commonly linked to prostate cancer, and it is classified under the group of mycoplasmas. Improved management of mycoplasma infections is essential due to the frequent ineffectiveness of current antibiotic treatments in completely eliminating these pathogens from the host. The objective of this study is to design and construct effective and protective vaccines guided by structural proteomics and machine learning algorithms to provide protection against the M. pulmonis infection. Through a thorough examination of the entire proteome of M. pulmonis, four specific targets Membrane protein P80, Lipoprotein, Uncharacterized protein and GGDEF domain-containing protein have been identified as appropriate for designing a vaccine. The proteins underwent mapping of cytotoxic T lymphocyte (CTL), helper T lymphocyte (HTL) (IFN)-γ ±, and B-cell epitopes using artificial and recurrent neural networks. The design involved the creation of mRNA and peptide-based vaccine, which consisted of 8 CTL epitopes associated by GGS linkers, 7 HTL (IFN-positive) epitopes, and 8 B-cell epitopes joined by GPGPG linkers. The vaccine designed exhibit antigenic behavior, non-allergenic qualities, and exceptional physicochemical attributes. Structural modeling revealed that correct folding is crucial for optimal functioning. The coupling of the MEVC and Toll-like Receptors (TLR)1, TLR2, and TLR6 was examined through molecular docking experiments. This was followed by molecular simulation investigations, which included binding free energy estimations. The results indicated that the dynamics of the interaction were stable, and the binding was strong. In silico cloning and optimization analysis revealed an optimized sequence with a GC content of 49.776 % and a CAI of 0.982. The immunological simulation results showed strong immune responses, with elevated levels of active and plasma B-cells, regulatory T-cells, HTL, and CTL in both IgM+IgG and secondary immune responses. The antigen was completely cleared by the 50th day. This study lays the foundation for creating a potent and secure vaccine candidate to combat the newly identified M. pulmonis infection in people.
Collapse
Affiliation(s)
- Abbas Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Muhammad Ammar Zahid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Farheen Farrukh
- Gujranwala Medical College, 5 KM Alipur Chatha Rd, Gondlanwala Rd, Gujranwala, Pakistan
| | - Shahenda Salah Abdelsalam
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Raed M Al-Zoubi
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar; Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Chemistry, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan.
| | - Mohanad Shkoor
- Department of Chemistry, College of Arts and Science, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | - Dong-Qing Wei
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
4
|
Wynn EL, Browne AS, Clawson ML. Diversity and antigenic potentials of Mycoplasmopsis bovis secreted and outer membrane proteins within a core genome of strains isolated from North American bison and cattle. Genome 2024; 67:204-209. [PMID: 38330385 DOI: 10.1139/gen-2023-0084] [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] [Indexed: 02/10/2024]
Abstract
Mycoplasmopsis bovis is a worldwide economically important pathogen of cattle that can cause or indirectly contribute to bovine respiratory disease. M. bovis is also a primary etiological agent of respiratory disease in bison with high mortality rates. A major challenge in the development of an efficacious M. bovis vaccine is the design of antigens that contain both MHC-1 and MHC-2 T-cell epitopes, and that account for population level diversity within the species. Publicly available genomes and sequence read archive libraries of 381 M. bovis strains isolated from cattle (n = 202) and bison (n = 179) in North America were used to identify a core genome of 575 genes, including 38 that encode either known or predicted secreted or outer membrane proteins. The antigenic potentials of the proteins were characterized by the presence and strength of their T-cell epitopes, and their protein variant diversity at the population-level. The proteins had surprisingly low diversity and varying predictive levels of T-cell antigenicity. These results provide a reference for the selection or design of antigens for vaccine testing against strains infecting North American cattle and bison.
Collapse
Affiliation(s)
- Emily L Wynn
- United States Department of Agriculture (USDA), Agricultural Research Service (ARS) US Meat Animal Research Center, Clay Center, NE, USA
| | - A Springer Browne
- USDA, Animal and Plant Health Inspection Service (APHIS), Center for Epidemiology and Animal Health, Fort Collins, CO, USA
| | - Michael L Clawson
- United States Department of Agriculture (USDA), Agricultural Research Service (ARS) US Meat Animal Research Center, Clay Center, NE, USA
| |
Collapse
|
5
|
Rosales RS, Risco D, García-Nicolás O, Pallarés FJ, Ramírez AS, Poveda JB, Nicholas RAJ, Salguero FJ. Differential Gene Expression in Porcine Lung Compartments after Experimental Infection with Mycoplasma hyopneumoniae. Animals (Basel) 2024; 14:1290. [PMID: 38731294 PMCID: PMC11083927 DOI: 10.3390/ani14091290] [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: 03/10/2024] [Revised: 04/20/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Mycoplasma hyopneumoniae (Mhyo) is the causative agent of porcine enzootic pneumonia (EP), as well as one of the main pathogens involved in the porcine respiratory disease complex. The host-pathogen interaction between Mhyo and infected pigs is complex and not completely understood; however, improving the understanding of these intricacies is essential for the development of effective control strategies of EP. In order to improve our knowledge about this interaction, laser-capture microdissection was used to collect bronchi, bronchi-associated lymphoid tissue, and lung parenchyma from animals infected with different strains of Mhyo, and mRNA expression levels of different molecules involved in Mhyo infection (ICAM1, IL-8, IL-10, IL-23, IFN-α, IFN-γ, TGF-β, and TNF-α) were analyzed by qPCR. In addition, the quantification of Mhyo load in the different lung compartments and the scoring of macroscopic and microscopic lung lesions were also performed. Strain-associated differences in virulence were observed, as well as the presence of significant differences in expression levels of cytokines among lung compartments. IL-8 and IL-10 presented the highest upregulation, with limited differences between strains and lung compartments. IFN-α was strongly downregulated in BALT, implying a relevant role for this cytokine in the immunomodulation associated with Mhyo infections. IL-23 was also upregulated in all lung compartments, suggesting the potential involvement of a Th17-mediated immune response in Mhyo infections. Our findings highlight the relevance of Th1 and Th2 immune response in cases of EP, shedding light on the gene expression levels of key cytokines in the lung of pigs at a microscopic level.
Collapse
Affiliation(s)
- Rubén S. Rosales
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Veterinary Faculty, University of Las Palmas de Gran Canaria, Trasmontaña s/n, 35416 Arucas, Spain; (R.S.R.); (A.S.R.); (J.B.P.)
| | - David Risco
- Unidad de Histología y Anatomía Patológica, Departamento de Medicina Animal, Veterinary Faculty, University of Extremadura, Avenida de la Universidad, s/n, 10003 Cáceres, Spain
| | - Obdulio García-Nicolás
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland;
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Francisco J. Pallarés
- Pathology and Immunology Group (UCO-PIG), Department of Anatomy and Comparative Pathology and Toxicology, UIC Zoonosis y Enfermedades Emergentes ENZOEM, University of Córdoba, International Excellence Agrifood Campus “CeiA3”, 14001 Córdoba, Spain;
| | - Ana S. Ramírez
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Veterinary Faculty, University of Las Palmas de Gran Canaria, Trasmontaña s/n, 35416 Arucas, Spain; (R.S.R.); (A.S.R.); (J.B.P.)
| | - José B. Poveda
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Veterinary Faculty, University of Las Palmas de Gran Canaria, Trasmontaña s/n, 35416 Arucas, Spain; (R.S.R.); (A.S.R.); (J.B.P.)
| | | | - Francisco J. Salguero
- School of Veterinary Medicine, University of Surrey, Daphne Jackson Rd, Guildford GU2 7AL, UK;
| |
Collapse
|
6
|
Santos MR, Toledo LT, Bassi ÊJ, Porto WJN, Bressan GC, Moreira MAS, Chang YF, Silva-Júnior A. Chimeric proteins of Mycoplasma hyopneumoniae as vaccine and preclinical model for immunological evaluation. Braz J Microbiol 2024; 55:943-953. [PMID: 38217795 PMCID: PMC10920614 DOI: 10.1007/s42770-023-01240-7] [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: 08/29/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024] Open
Abstract
Mycoplasma hyopneumoniae (M. hyopneumoniae) is a primary agent of porcine enzootic pneumonia, a disease that causes significant economic losses to pig farming worldwide. Commercial vaccines induce partial protection, evidencing the need for a new vaccine against M. hyopneumoniae. In our work, three chimeric proteins were constructed, composed of potentially immunogenic domains from M. hyopneumoniae proteins. We designed three chimeric proteins (Q1, Q2, and Q3) based on bioinformatics analysis that identified five potential proteins with immunogenic potential (MHP418, MHP372, MHP199, P97, and MHP0461). The chimeric proteins were inoculated in the murine model to evaluate the immune response. The mice vaccinated with the chimeras presented IgG and IgG1 against proteins of M. hyopneumoniae. There was induction of IgG in mice immunized with Q3 starting from 30 days post-vaccination, and groups Q1 and Q2 showed induction at 45 days. Mice of the group immunized with Q3 showed the production of IgA. In addition, the mice inoculated with chimeric proteins showed a proinflammatory cytokine response; Q1 demonstrated higher levels of TNF, IL-6, IL2, and IL-17. In contrast, animals immunized with Q2 showed an increase in the concentrations of TNF, IL-6, and IL-4, whereas those immunized with Q3 exhibited an increase in the concentrations of TNF, IL-6, IL-10, and IL-4. The results of the present study indicate that these three chimeric proteins can be used in future vaccine trials with swine because of the promising antigenicity.
Collapse
Affiliation(s)
- Marcus Rebouças Santos
- Department of Veterinary Medicine, Universidade Federal de Vicosa, Vicosa, Minas Gerais, Brazil
| | - Leonardo Teófilo Toledo
- Department of Veterinary Medicine, Universidade Federal de Vicosa, Vicosa, Minas Gerais, Brazil
| | - Ênio José Bassi
- Institute of Biological Sciences and Health, Universidade Federal de Alagoas, Maceió, Brazil
| | | | - Gustavo Costa Bressan
- Department of Veterinary Medicine, Universidade Federal de Vicosa, Vicosa, Minas Gerais, Brazil
| | | | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Abelardo Silva-Júnior
- Institute of Biological Sciences and Health, Universidade Federal de Alagoas, Maceió, Brazil.
| |
Collapse
|
7
|
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
|
8
|
Si D, Sun J, Guo L, Yang F, Li J, He S. Mycoplasma synoviae lipid-associated membrane proteins identification and expression changes when exposed to chicken cells. Front Vet Sci 2023; 10:1249499. [PMID: 38026678 PMCID: PMC10652285 DOI: 10.3389/fvets.2023.1249499] [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] [Received: 06/28/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Mycoplasma synoviae is a significant cause of respiratory disease and synovitis among chickens, and has an adverse economic impact on broiler breeding efforts. The present study was designed to develop a systematic understanding of the role that M. synoviae lipid-associated membrane proteins (LAMPs) may play in the virulence of this pathogen. Bioinformatics tools were used to identify 146 predicted membrane proteins and lipoproteins in the M. synoviae proteome. Then, Triton X-114 was used to extract LAMPs that were subsequently identified via LC-MS/MS. This approach enabled the detection of potential LAMPs, and the top 200 most abundant proteins detected using this strategy were subject to further analysis. M. synoviae cells (100 MOI) were exposed to chicken fibroblasts (DF-1) and macrophages (HD-11) in a 1:1 mixed culture. Analysis of LAMP transcripts identified 72 up-regulated LAMP genes which were analyzed in depth by bioinformatics. GO analysis revealed these genes to be enriched in the nucleotide binding, sulfur amino acid transmembrane transporter activity, tRNA binding, rRNA modification, and transition metal ion transport pathways. Moreover, KEGG enrichment analysis suggested that these genes were enriched in the biosynthesis of secondary metabolites, carbon metabolism, glycolysis/gluconeogenesis, and nitrogen metabolism pathways.
Collapse
Affiliation(s)
- Duoduo Si
- College of Animal Science and Technology, Clinical Veterinary Laboratory, Ningxia University, Yinchuan, China
| | - Jialin Sun
- College of Animal Science and Technology, Clinical Veterinary Laboratory, Ningxia University, Yinchuan, China
| | - Lei Guo
- Ningxia Xiaoming Agriculture and Animal Husbandry Co., Ltd., Yinchuan, China
| | - Fei Yang
- College of Animal Science and Technology, Clinical Veterinary Laboratory, Ningxia University, Yinchuan, China
| | - Jidong Li
- College of Animal Science and Technology, Clinical Veterinary Laboratory, Ningxia University, Yinchuan, China
| | - Shenghu He
- College of Animal Science and Technology, Clinical Veterinary Laboratory, Ningxia University, Yinchuan, China
| |
Collapse
|
9
|
Lan S, Li Z, Hao H, Liu S, Huang Z, Bai Y, Li Y, Yan X, Gao P, Chen S, Chu Y. A genome-wide transposon mutagenesis screening identifies LppB as a key factor associated with Mycoplasma bovis colonization and invasion into host cells. FASEB J 2023; 37:e23176. [PMID: 37665592 DOI: 10.1096/fj.202300678r] [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: 04/17/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
Mycoplasma spp., the smallest self-replicating and genome-reduced organisms, have raised a great concern in both the medical and veterinary fields due to their pathogenicity. The molecular determinants of these wall-less bacterium efficiently use their limited genes to ensure successful infection of the host remain unclear. In the present study, we used the ruminant pathogen Mycoplasma bovis as a model to identify the key factors for colonization and invasion into host cells. We constructed a nonredundant fluorescent transposon mutant library of M. bovis using a modified transposon plasmid, and identified 34 novel adhesion-related genes based on a high-throughput screening approach. Among them, the ΔLppB mutant exhibited the most apparent decrease in adhesion to embryonic bovine lung (EBL) cells. The surface-localized lipoprotein LppB, which is highly conserved in Mycoplasma species, was then confirmed as a key factor for M. bovis adhesion with great immunogenicity. LppB interacted with various components (fibronectin, vitronectin, collagen IV, and laminin) of host extracellular matrix (ECM) and promoted plasminogen activation through tPA to degrade ECM. The 439-502 amino acid region of LppB is a critical domain, and F465 and Y493 are important residues for the plasminogen activation activity. We further revealed LppB as a key factor facilitating internalization through clathrin- and lipid raft-mediated endocytosis, which helps the Mycoplasma invade the host cells. Our study indicates that LppB plays a key role in Mycoplasma infection and is a potential new therapeutic and vaccine target for Mycoplasma species.
Collapse
Affiliation(s)
- 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Zhicheng Huang
- 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Yutong Bai
- 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Yanzhao 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, 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, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Animal Biosafety Risk Warning and Control (North), Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| |
Collapse
|
10
|
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
|
11
|
Wenger M, Grosse-Kathoefer S, Kraiem A, Pelamatti E, Nunes N, Pointner L, Aglas L. When the allergy alarm bells toll: The role of Toll-like receptors in allergic diseases and treatment. Front Mol Biosci 2023; 10:1204025. [PMID: 37426425 PMCID: PMC10325731 DOI: 10.3389/fmolb.2023.1204025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
Toll-like receptors of the human immune system are specialized pathogen detectors able to link innate and adaptive immune responses. TLR ligands include among others bacteria-, mycoplasma- or virus-derived compounds such as lipids, lipo- and glycoproteins and nucleic acids. Not only are genetic variations in TLR-related genes associated with the pathogenesis of allergic diseases, including asthma and allergic rhinitis, their expression also differs between allergic and non-allergic individuals. Due to a complex interplay of genes, environmental factors, and allergen sources the interpretation of TLRs involved in immunoglobulin E-mediated diseases remains challenging. Therefore, it is imperative to dissect the role of TLRs in allergies. In this review, we discuss i) the expression of TLRs in organs and cell types involved in the allergic immune response, ii) their involvement in modulating allergy-associated or -protective immune responses, and iii) how differential activation of TLRs by environmental factors, such as microbial, viral or air pollutant exposure, results in allergy development. However, we focus on iv) allergen sources interacting with TLRs, and v) how targeting TLRs could be employed in novel therapeutic strategies. Understanding the contributions of TLRs to allergy development allow the identification of knowledge gaps, provide guidance for ongoing research efforts, and built the foundation for future exploitation of TLRs in vaccine design.
Collapse
|
12
|
Souza dos Santos P, Paes JA, Del Prá Netto Machado L, Paludo GP, Zaha A, Ferreira HB. Differential domains and endoproteolytic processing in dominant surface proteins of unknown function from Mycoplasma hyopneumoniae and Mycoplasma flocculare. Heliyon 2023; 9:e16141. [PMID: 37251846 PMCID: PMC10213202 DOI: 10.1016/j.heliyon.2023.e16141] [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: 09/19/2022] [Revised: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Mycoplasma hyopneumoniae causes porcine enzootic pneumonia (PEP), a chronic respiratory disease that leads to severe economic losses in the pig industry. Swine infection and PEP development depend on the adhesion of the pathogen to the swine respiratory tract and the host immune response, but these and other disease determinants are not fully understood. For instance, M. hyopneumoniae has a large repertoire of proteins of unknown function (PUFs) and some of them are abundant in the cell surface, where they likely mediate so far unknown pathogen-host interactions. Moreover, these surface PUFs may undergo endoproteolytic processing to generate larger repertoires of proteoforms to further complicate this scenario. Here, we investigated the five PUFs more represented on the surface of M. hyopneumoniae pathogenic strain 7448 in comparison with their orthologs from the nonpathogenic M. hyopneumoniae J strain and the closely related commensal species Mycoplasma flocculare. Comparative in silico analyses of deduced amino acid sequences and proteomic data identified differential domains, disordered regions and repeated motifs. We also provide evidence of differential endoproteolytic processing and antigenicity. Phylogenetic analyses were also performed with ortholog sequences, showing higher conservation of three of the assessed PUFs among Mycoplasma species related to respiratory diseases. Overall, our data point out to M. hyopneumoniae surface-dominant PUFs likely associated with pathogenicity.
Collapse
Affiliation(s)
- Priscila Souza dos Santos
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, UFRGS, Porto Alegre, Brazil
| | - Jéssica Andrade Paes
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, UFRGS, Porto Alegre, Brazil
| | - Lais Del Prá Netto Machado
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, UFRGS, Porto Alegre, Brazil
| | - Gabriela Prado Paludo
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, UFRGS, Porto Alegre, Brazil
| | - Arnaldo Zaha
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, UFRGS, Porto Alegre, Brazil
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, UFRGS, Porto Alegre, Brazil
- Departamento de Biologia Molecular e Biotecnologia, Instituto de Biociências, UFRGS, Porto Alegre, Brazil
| | - Henrique Bunselmeyer Ferreira
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, UFRGS, Porto Alegre, Brazil
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, UFRGS, Porto Alegre, Brazil
- Departamento de Biologia Molecular e Biotecnologia, Instituto de Biociências, UFRGS, Porto Alegre, Brazil
| |
Collapse
|
13
|
Yueyue W, Feichen X, Yixuan X, Lu L, Yiwen C, Xiaoxing Y. Pathogenicity and virulence of Mycoplasma genitalium: Unraveling Ariadne's Thread. Virulence 2022; 13:1161-1183. [PMID: 35791283 PMCID: PMC9262362 DOI: 10.1080/21505594.2022.2095741] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mycoplasma genitalium, a pathogen from class Mollicutes, has been linked to sexually transmitted diseases and sparked widespread concern. To adapt to its environment, M. genitalium has evolved specific adhesins and motility mechanisms that allow it to adhere to and invade various eukaryotic cells, thereby causing severe damage to the cells. Even though traditional exotoxins have not been identified, secreted nucleases or membrane lipoproteins have been shown to cause cell death and inflammatory injury in M. genitalium infection. However, as both innate and adaptive immune responses are important for controlling infection, the immune responses that develop upon infection do not necessarily eliminate the organism completely. Antigenic variation, detoxifying enzymes, immunoglobulins, neutrophil extracellular trap-degrading enzymes, cell invasion, and biofilm formation are important factors that help the pathogen overcome the host defence and cause chronic infections in susceptible individuals. Furthermore, M. genitalium can increase the susceptibility to several sexually transmitted pathogens, which significantly complicates the persistence and chronicity of M. genitalium infection. This review aimed to discuss the virulence factors of M. genitalium to shed light on its complex pathogenicity and pathogenesis of the infection.
Collapse
Affiliation(s)
- Wu Yueyue
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xiu Feichen
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xi Yixuan
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Liu Lu
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Chen Yiwen
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - You Xiaoxing
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
14
|
Eating the Enemy: Mycoplasma Strategies to Evade Neutrophil Extracellular Traps (NETs) Promoting Bacterial Nucleotides Uptake and Inflammatory Damage. Int J Mol Sci 2022; 23:ijms232315030. [PMID: 36499356 PMCID: PMC9740415 DOI: 10.3390/ijms232315030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Neutrophils are effector cells involved in the innate immune response against infection; they kill infectious agents in the intracellular compartment (phagocytosis) or in the extracellular milieu (degranulation). Moreover, neutrophils release neutrophil extracellular traps (NETs), complex structures composed of a scaffold of decondensed DNA associated with histones and antimicrobial compounds; NETs entrap infectious agents, preventing their spread and promoting their clearance. NET formation is triggered by microbial compounds, but many microorganisms have evolved several strategies for NET evasion. In addition, the dysregulated production of NETs is associated with chronic inflammatory diseases. Mycoplasmas are reduced genome bacteria, able to induce chronic infections with recurrent inflammatory symptoms. Mycoplasmas' parasitic lifestyle relies on metabolite uptake from the host. Mycoplasmas induce NET release, but their surface or secreted nucleases digest the NETs' DNA scaffold, allowing them to escape from entrapment and providing essential nucleotide precursors, thus promoting the infection. The presence of Mycoplasma species has been associated with chronic inflammatory disorders, such as systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Crohn's disease, and cancer. The persistence of mycoplasma infection and prolonged NET release may contribute to the onset of chronic inflammatory diseases and needs further investigation and insights.
Collapse
|
15
|
Mugunthan SP, Harish MC. In silico structural homology modeling and functional characterization of Mycoplasma gallisepticum variable lipoprotein hemagglutin proteins. Front Vet Sci 2022; 9:943831. [PMID: 35990271 PMCID: PMC9386052 DOI: 10.3389/fvets.2022.943831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Mycoplasma gallisepticum variable lipoprotein hemagglutin (vlhA) proteins are crucial for immune evasion from the host cells, permitting the persistence and survival of the pathogen. However, the exact molecular mechanism behind the immune evasion function is still not clear. In silico physiochemical analysis, domain analysis, subcellular localization, and homology modeling studies have been carried out to predict the structural and functional properties of these proteins. The outcomes of this study provide significant preliminary data for understanding the immune evasion by vlhA proteins. In this study, we have reported the primary, secondary, and tertiary structural characteristics and subcellular localization, presence of the transmembrane helix and signal peptide, and functional characteristics of vlhA proteins from M. gallisepticum strain R low. The results show variation between the structural and functional components of the proteins, signifying the role and diverse molecular mechanisms in functioning of vlhA proteins in host immune evasion. Moreover the 3D structure predicted in this study will pave a way for understanding vlhA protein function and its interaction with other molecules to undergo immune evasion. This study forms the basis for future experimental studies improving our understanding in the molecular mechanisms used by vlhA proteins.
Collapse
|
16
|
Gaeta NC, de Sá Guimarães AM, Timenetsky J, Clouser S, Gregory L, Ganda E. The first Mycoplasma ovipneumoniae recovered from a sheep with respiratory disease in Brazil - draft genome and genomic analysis. Vet Res Commun 2022; 46:1311-1318. [PMID: 35804255 DOI: 10.1007/s11259-022-09972-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/02/2022] [Indexed: 11/28/2022]
Abstract
Mycoplasma ovipneumoniae is an important etiological agent of sheep respiratory disease worldwide. Here, we describe the first isolation and draft genome sequence of M. ovipneumoniae strain USP-BR2017 retrieved from tracheobronchial lavage of a sheep showing clinical signs of respiratory disease in the Rio de Janeiro State, Brazil. The culture of tracheobronchial lavage resulted in glucose-fermenting fried egg colonies, which were identified as M. ovipneumoniae by polymerase chain reaction. The genome was sequenced using the Illumina NextSeq 2000 and de novo assembled using SPAdes. The genome of the sequenced organism presented an approximate size of 1,122,253 bp. The annotation revealed 773 coding DNA sequences (CDSs), 806 genes, three rRNAs, and 30 tRNAs. Data analysis revealed M. ovipneumoniae strain USP-BR2017 contains a few virulence genes, including the hemolysing C gene (hlyC). In addition, strain USP-BR2017 showed high identity over the 16S rRNA gene with other sheep isolates from China and United States. This first description of M. ovipneumoniae in diseased Brazilian sheep demonstrates the importance of continuous surveillance and diagnostics of pathogens causing respiratory disease in sheep in Brazil.
Collapse
Affiliation(s)
- Natália C Gaeta
- Department of Internal Medicine, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil. .,Laboratory of Bacterial Zoonosis, Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.
| | - Ana Marcia de Sá Guimarães
- Laboratory of Applied Research to Mycobacteria, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jorge Timenetsky
- Laboratory of Mycoplasmas, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Stephanie Clouser
- Department of Animal Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Lilian Gregory
- Department of Internal Medicine, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Erika Ganda
- Department of Animal Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
17
|
Ranjitkar S, Duan JE, Srirattana K, Alqahtani F, Tulman ER, Mandoiu I, Venkitanarayanan K, Tian X. Transcriptomic Responses of Mycoplasma bovis Upon Treatments of trans-Cinnamaldehyde, Carvacrol, and Eugenol. Front Microbiol 2022; 13:888433. [PMID: 35733968 PMCID: PMC9207385 DOI: 10.3389/fmicb.2022.888433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Mycoplasma bovis (M. bovis) is an insidious, wall-less primary bacterial pathogen that causes bovine pneumonia, mid-ear infection, mastitis, and arthritis. The economic losses caused by M. bovis due to culling, diminished milk production, and feed conversion are underestimated because of poor diagnosis/recognition. Treatment with common antibiotics targeting the cell wall is ineffective. Plant-derived antimicrobials (PDAs) such as food-grade trans-cinnamaldehyde (TC), eugenol (EU), and carvacrol (CAR) are inexpensive and generally regarded as safe for humans and animals yet possess strong anti-bacterial properties. In preliminary studies, we found that all three PDAs inhibited the growth of M. bovis in vitro. Through RNA sequencing, we report here that CAR affected the expression of 153 genes which included the downregulation of energy generation-related proteins, pentose phosphate pathway, and upregulation of ribosomes and translation-related proteins. Few differentially expressed genes were found when M. bovis was treated with TC, EU, or when the three PDAs were double or triple combined. Our results suggest that, as opposed to the effect of CAR, the growth-inhibitory effects of TC and EU at levels tested may be exerted through mechanisms other than gene expression regulations.
Collapse
Affiliation(s)
- Saurav Ranjitkar
- Department of Animal Science, University of Connecticut, Storrs, CT, United States
| | - Jingyue Ellie Duan
- Department of Animal Science, University of Connecticut, Storrs, CT, United States
| | - Kanokwan Srirattana
- Department of Animal Science, University of Connecticut, Storrs, CT, United States
| | - Fahad Alqahtani
- National Center for Bioinformatics, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Edan R. Tulman
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT, United States
| | - Ion Mandoiu
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, United States
| | | | - Xiuchun Tian
- Department of Animal Science, University of Connecticut, Storrs, CT, United States
- *Correspondence: Xiuchun Tian,
| |
Collapse
|
18
|
Santos-Junior MN, Neves WS, Santos RS, Almeida PP, Fernandes JM, Guimarães BCDB, Barbosa MS, da Silva LSC, Gomes CP, Sampaio BA, Rezende IDS, Correia TML, Neres NSDM, Campos GB, Bastos BL, Timenetsky J, Marques LM. Heterologous Expression, Purification, and Immunomodulatory Effects of Recombinant Lipoprotein GUDIV-103 Isolated from Ureaplasma diversum. Microorganisms 2022; 10:microorganisms10051032. [PMID: 35630474 PMCID: PMC9147684 DOI: 10.3390/microorganisms10051032] [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: 04/14/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 02/06/2023] Open
Abstract
Ureaplasma diversum is a bacterial pathogen that infects cattle and can cause severe inflammation of the genital and reproductive systems. Lipid-associated membrane proteins (LAMPs), including GUDIV-103, are the main virulence factors in this bacterium. In this study, we heterologously expressed recombinant GUDIV-103 (rGUDIV-103) in Escherichia coli, purified it, and evaluated its immunological reactivity and immunomodulatory effects in bovine peripheral blood mononuclear cells (PBMCs). Samples from rabbits inoculated with purified rGUDIV-103 were analysed using indirect enzyme-linked immunosorbent assay and dot blotting to confirm polyclonal antibody production and assess kinetics, respectively. The expression of this lipoprotein in field isolates was confirmed via Western blotting with anti-rGUDIV-103 serum and hydrophobic or hydrophilic proteins from 42 U. diversum strains. Moreover, the antibodies produced against the U. diversum ATCC 49783 strain recognised rGUDIV-103. The mitogenic potential of rGUDIV-103 was evaluated using a lymphoproliferation assay in 5(6)-carboxyfluorescein diacetate succinimidyl ester−labelled bovine PBMCs, where it induced lymphocyte proliferation. Quantitative polymerase chain reaction analysis revealed that the expression of interleukin-1β, toll-like receptor (TLR)-α, TLR2, TLR4, inducible nitric oxide synthase, and caspase-3−encoding genes increased more in rGUDIV-103−treated PBMCs than in untreated cells (p < 0.05). Treating PBMCs with rGUDIV-103 increased nitric oxide and hydrogen peroxide levels. The antigenic and immunogenic properties of rGUDIV-103 suggested its suitability for immunobiological application.
Collapse
Affiliation(s)
- Manoel Neres Santos-Junior
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista 40170-110, Brazil; (M.N.S.-J.); (W.S.N.); (R.S.S.); (J.M.F.); (T.M.L.C.); (N.S.d.M.N.)
- Department of Biology, and Biotechnology of Microorganisms, State University of Santa Cruz (UESC), Ilhéus 45662-900, Brazil; (B.C.d.B.G.); (L.S.C.d.S.); (C.P.G.); (B.A.S.); (G.B.C.); (B.L.B.)
| | - Wanderson Souza Neves
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista 40170-110, Brazil; (M.N.S.-J.); (W.S.N.); (R.S.S.); (J.M.F.); (T.M.L.C.); (N.S.d.M.N.)
| | - Ronaldo Silva Santos
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista 40170-110, Brazil; (M.N.S.-J.); (W.S.N.); (R.S.S.); (J.M.F.); (T.M.L.C.); (N.S.d.M.N.)
| | - Palloma Porto Almeida
- Bioinformatics and Computational Biology Lab, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil;
| | - Janaina Marinho Fernandes
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista 40170-110, Brazil; (M.N.S.-J.); (W.S.N.); (R.S.S.); (J.M.F.); (T.M.L.C.); (N.S.d.M.N.)
| | - Bruna Carolina de Brito Guimarães
- Department of Biology, and Biotechnology of Microorganisms, State University of Santa Cruz (UESC), Ilhéus 45662-900, Brazil; (B.C.d.B.G.); (L.S.C.d.S.); (C.P.G.); (B.A.S.); (G.B.C.); (B.L.B.)
| | - Maysa Santos Barbosa
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil; (M.S.B.); (I.d.S.R.); (J.T.)
| | - Lucas Santana Coelho da Silva
- Department of Biology, and Biotechnology of Microorganisms, State University of Santa Cruz (UESC), Ilhéus 45662-900, Brazil; (B.C.d.B.G.); (L.S.C.d.S.); (C.P.G.); (B.A.S.); (G.B.C.); (B.L.B.)
| | - Camila Pacheco Gomes
- Department of Biology, and Biotechnology of Microorganisms, State University of Santa Cruz (UESC), Ilhéus 45662-900, Brazil; (B.C.d.B.G.); (L.S.C.d.S.); (C.P.G.); (B.A.S.); (G.B.C.); (B.L.B.)
| | - Beatriz Almeida Sampaio
- Department of Biology, and Biotechnology of Microorganisms, State University of Santa Cruz (UESC), Ilhéus 45662-900, Brazil; (B.C.d.B.G.); (L.S.C.d.S.); (C.P.G.); (B.A.S.); (G.B.C.); (B.L.B.)
| | - Izadora de Souza Rezende
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil; (M.S.B.); (I.d.S.R.); (J.T.)
| | - Thiago Macedo Lopes Correia
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista 40170-110, Brazil; (M.N.S.-J.); (W.S.N.); (R.S.S.); (J.M.F.); (T.M.L.C.); (N.S.d.M.N.)
| | - Nayara Silva de Macedo Neres
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista 40170-110, Brazil; (M.N.S.-J.); (W.S.N.); (R.S.S.); (J.M.F.); (T.M.L.C.); (N.S.d.M.N.)
| | - Guilherme Barreto Campos
- Department of Biology, and Biotechnology of Microorganisms, State University of Santa Cruz (UESC), Ilhéus 45662-900, Brazil; (B.C.d.B.G.); (L.S.C.d.S.); (C.P.G.); (B.A.S.); (G.B.C.); (B.L.B.)
| | - Bruno Lopes Bastos
- Department of Biology, and Biotechnology of Microorganisms, State University of Santa Cruz (UESC), Ilhéus 45662-900, Brazil; (B.C.d.B.G.); (L.S.C.d.S.); (C.P.G.); (B.A.S.); (G.B.C.); (B.L.B.)
| | - Jorge Timenetsky
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil; (M.S.B.); (I.d.S.R.); (J.T.)
| | - Lucas Miranda Marques
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista 40170-110, Brazil; (M.N.S.-J.); (W.S.N.); (R.S.S.); (J.M.F.); (T.M.L.C.); (N.S.d.M.N.)
- Department of Biology, and Biotechnology of Microorganisms, State University of Santa Cruz (UESC), Ilhéus 45662-900, Brazil; (B.C.d.B.G.); (L.S.C.d.S.); (C.P.G.); (B.A.S.); (G.B.C.); (B.L.B.)
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil; (M.S.B.); (I.d.S.R.); (J.T.)
- Correspondence:
| |
Collapse
|
19
|
Dawood A, Algharib SA, Zhao G, Zhu T, Qi M, Delai K, Hao Z, Marawan MA, Shirani I, Guo A. Mycoplasmas as Host Pantropic and Specific Pathogens: Clinical Implications, Gene Transfer, Virulence Factors, and Future Perspectives. Front Cell Infect Microbiol 2022; 12:855731. [PMID: 35646746 PMCID: PMC9137434 DOI: 10.3389/fcimb.2022.855731] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022] Open
Abstract
Mycoplasmas as economically important and pantropic pathogens can cause similar clinical diseases in different hosts by eluding host defense and establishing their niches despite their limited metabolic capacities. Besides, enormous undiscovered virulence has a fundamental role in the pathogenesis of pathogenic mycoplasmas. On the other hand, they are host-specific pathogens with some highly pathogenic members that can colonize a vast number of habitats. Reshuffling mycoplasmas genetic information and evolving rapidly is a way to avoid their host's immune system. However, currently, only a few control measures exist against some mycoplasmosis which are far from satisfaction. This review aimed to provide an updated insight into the state of mycoplasmas as pathogens by summarizing and analyzing the comprehensive progress, current challenge, and future perspectives of mycoplasmas. It covers clinical implications of mycoplasmas in humans and domestic and wild animals, virulence-related factors, the process of gene transfer and its crucial prospects, the current application and future perspectives of nanotechnology for diagnosing and curing mycoplasmosis, Mycoplasma vaccination, and protective immunity. Several questions remain unanswered and are recommended to pay close attention to. The findings would be helpful to develop new strategies for basic and applied research on mycoplasmas and facilitate the control of mycoplasmosis for humans and various species of animals.
Collapse
Affiliation(s)
- Ali Dawood
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
- Hubei Hongshan Laboratory, Wuhan, China
| | - Samah Attia Algharib
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, HZAU, Wuhan, China
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Gang Zhao
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan, China
| | - Tingting Zhu
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan, China
| | - Mingpu Qi
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan, China
| | - Kong Delai
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhiyu Hao
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan, China
| | - Marawan A. Marawan
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- Infectious Diseases, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Ihsanullah Shirani
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- Para-Clinic Department, Faculty of Veterinary Medicine, Jalalabad, Afghanistan
| | - Aizhen Guo
- The State Key Laboratory of Agricultural Microbiology, (HZAU), Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
20
|
Sheep Infection Trials with 'Phase-Locked' Vpma Expression Variants of Mycoplasma agalactiae-Towards Elucidating the Role of a Multigene Family Encoding Variable Surface Lipoproteins in Infection and Disease. Microorganisms 2022; 10:microorganisms10040815. [PMID: 35456865 PMCID: PMC9025108 DOI: 10.3390/microorganisms10040815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/27/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
The significance of large multigene families causing high-frequency surface variations in mycoplasmas is not well-understood. Previously, VpmaY and VpmaU clonal variants of the Vpma family of lipoproteins of M. agalactiae were compared via experimental sheep infections using the two corresponding ‘Phase-Locked Mutants’. However, nothing is known about the infectivity of the remaining four Vpma expression variants VpmaX, VpmaW, VpmaZ and VpmaV as they were never evaluated in vivo. Here, in vivo infection and disease progression of all six Vpma expressers constituting the Vpma family of type strain PG2 were compared using the corresponding xer1-disrupted PLMs expressing single well-characterized Vpmas. Each of the six PLMs were separately evaluated using the intramammary sheep infection model along with the control phase-variable wildtype strain PG2. Thorough bacteriological, pathological and clinical examinations were performed, including assessment of milk quality, quantity and somatic cell counts. Altogether, the results indicated that the inability to vary the Vpma expression phase does not hamper the initiation of infection leading to mastitis for all six PLMs, except for PLMU, which showed a defect in host colonization and multiplication for the first 24 h p.i. and pathological/bacteriological analysis indicated a higher potential for systemic spread for PLMV and PLMX. This is the first study in which all isogenic expression variants of a large mycoplasma multigene family are tested in the natural host.
Collapse
|
21
|
Barbosa MS, Marques LM, Timenetsky J, Rosengarten R, Spergser J, Chopra-Dewasthaly R. Host cell interactions of novel antigenic membrane proteins of Mycoplasma agalactiae. BMC Microbiol 2022; 22:93. [PMID: 35395771 PMCID: PMC8991494 DOI: 10.1186/s12866-022-02512-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/30/2022] [Indexed: 11/30/2022] Open
Abstract
Background Mycoplasma agalactiae is the main etiological agent of Contagious Agalactia syndrome of small ruminants notifiable to the World Organization for Animal Health. Despite serious economic losses, successful vaccines are unavailable, largely because its colonization and invasion factors are not well understood. This study evaluates the role of two recently identified antigenic proteins (MAG_1560, MAG_6130) and the cytadhesin P40 in pathogenicity related phenotypes. Results Adhesion to HeLa and sheep primary mammary stromal cells (MSC) was evaluated using ELISA, as well as in vitro adhesion assays on monolayer cell cultures. The results demonstrated MAG_6130 as a novel adhesin of M. agalactiae whose capacity to adhere to eukaryotic cells was significantly reduced by specific antiserum. Additionally, these proteins exhibited significant binding to plasminogen and extracellular matrix (ECM) proteins like lactoferrin, fibrinogen and fibronectin, a feature that could potentially support the pathogen in host colonization, tissue migration and immune evasion. Furthermore, these proteins played a detrimental role on the host cell proliferation and viability and were observed to activate pro-apoptotic genes indicating their involvement in cell death when eukaryotic cells were infected with M. agalactiae. Conclusions To summarize, the hypothetical protein corresponding to MAG_6130 has not only been assigned novel adhesion functions but together with P40 it is demonstrated for the first time to bind to lactoferrin and ECM proteins thereby playing important roles in host colonization and pathogenicity. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-022-02512-2.
Collapse
Affiliation(s)
- Maysa Santos Barbosa
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, A-1210, Austria.,Present Address: Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Lucas Miranda Marques
- Present Address: Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil. .,Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista, Brazil.
| | - Jorge Timenetsky
- Present Address: Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Renate Rosengarten
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, A-1210, Austria
| | - Joachim Spergser
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, A-1210, Austria
| | - Rohini Chopra-Dewasthaly
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, A-1210, Austria.
| |
Collapse
|
22
|
Maya-Rodríguez LM, Carrillo-Casas EM, Rojas-Trejo V, Trigo-Tavera F, Miranda-Morales RE. Prevalence of three Mycoplasma sp. by multiplex PCR in cattle with and without respiratory disease in central Mexico. Trop Anim Health Prod 2022; 54:394. [PMID: 36417039 PMCID: PMC9685072 DOI: 10.1007/s11250-022-03398-y] [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: 05/18/2021] [Accepted: 11/09/2022] [Indexed: 11/24/2022]
Abstract
This study aimed to identify Mycoplasma bovis, Myc. dispar, and Myc. bovirhinis, which are involved in bovine respiratory disease through a multiplex PCR as an alternative to culture's features that hamper Mycoplasma isolation. Nasal swabs were taken from 335 cattle with and without respiratory disease background (RDB) from dairy herds in the central region of Mexico. Each sample was divided in two; the first part was processed for the direct DNA extraction of the nasal swab and the second for Mycoplasma isolation, culture, and then the multiplex PCR was performed. In the nasal swabs, Myc. bovis was identified in 21.1%; Myc. dispar, in 11.8%; and Myc. bovirhinis, in 10.8% in cattle with RDB. Isolates were identified as Myc. bovis, 20.1%; Myc. dispar, 11.8%; and Myc. bovirhinis, 6.1%. There is a strong correlation between the presence of Mycoplasma identified by PCR and the clinical history of the disease (ρ < 0.0000). In animals without RDB, Myc. bovirhinis was the only species detected in 6.1% of the samples processed directly for multiplex PCR, and in 2% of the isolates. There is an excellent correlation (kappa 0.803) between the isolation and the 16S PCR and a high correlation (kappa 0.75) between the isolation and the multiplex PCR. Therefore, we conclude that the PCR multiplex test is highly sensitive and may be used for the diagnosis and surveillance of the three species in biological samples and mycoplasma isolates.
Collapse
Affiliation(s)
- L. M. Maya-Rodríguez
- grid.9486.30000 0001 2159 0001Laboratorio de Mycoplasmas, Facultad de Medicina Veterinaria y Zootecnia UNAM, Departamento de Microbiología e Inmunología, Ciudad Universitaria, 04519 CDMX, CP Mexico
| | - E. M. Carrillo-Casas
- grid.414754.70000 0004 6020 7521Hospital General “Dr. Manuel Gea González”, Depto. de Biología Molecular e Histocompatibilidad, Dirección de Investigación, Calz. de Tlalpan 4800, Secc XVI, 14080 Tlalpan CDMX, CP Mexico
| | - V. Rojas-Trejo
- grid.9486.30000 0001 2159 0001Laboratorio de Mycoplasmas, Facultad de Medicina Veterinaria y Zootecnia UNAM, Departamento de Microbiología e Inmunología, Ciudad Universitaria, 04519 CDMX, CP Mexico
| | - F. Trigo-Tavera
- grid.9486.30000 0001 2159 0001Facultad de Medicina Veterinaria y Zootecnia UNAM, Departamento de Patología, Ciudad Universitaria, 04519 CDMX, CP Mexico
| | - R. E. Miranda-Morales
- grid.9486.30000 0001 2159 0001Laboratorio de Mycoplasmas, Facultad de Medicina Veterinaria y Zootecnia UNAM, Departamento de Microbiología e Inmunología, Ciudad Universitaria, 04519 CDMX, CP Mexico
| |
Collapse
|
23
|
Abstract
Mycoplasmas are small, genome-reduced bacteria. They are obligate parasites that can be found in a wide range of host species, including the majority of livestock animals and humans. Colonization of the host can result in a wide spectrum of outcomes. In many cases, these successful parasites are considered commensal, as they are found in the microbiota of asymptomatic carriers. Conversely, mycoplasmas can also be pathogenic, as they are associated with a range of both acute and chronic inflammatory diseases which are problematic in veterinary and human medicine. The chronicity of mycoplasma infections and the ability of these bacteria to infect even recently vaccinated individuals clearly indicate that they are able to successfully evade their host’s humoral immune response. Over the years, multiple strategies of immune evasion have been identified in mycoplasmas, with a number of them aimed at generating important antigenic diversity. More recently, mycoplasma-specific anti-immunoglobulin strategies have also been characterized. Through the expression of the immunoglobulin-binding proteins protein M or mycoplasma immunoglobulin binding (MIB), mycoplasmas have the ability to target the host’s antibodies and to prevent them from interacting with their cognate antigens. In this review, we discuss how these discoveries shed new light on the relationship between mycoplasmas and their host’s immune system. We also propose that these strategies should be taken into consideration for future studies, as they are key to our understanding of mycoplasma diseases' chronic and inflammatory nature and are probably a contributing factor to reduce vaccine efficacy.
Collapse
|
24
|
Pereira JDJ, Ikegami RN, Kawakami JT, Garavelo SM, Reis MM, Palomino SAP, Mangini S, Moreno CR, de Barros SF, Souza AR, Higuchi MDL. Distinct Microbial Communities in Dilated Cardiomyopathy Explanted Hearts Are Associated With Different Myocardial Rejection Outcomes. Front Cell Infect Microbiol 2021; 11:732276. [PMID: 34912727 PMCID: PMC8668412 DOI: 10.3389/fcimb.2021.732276] [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: 06/28/2021] [Accepted: 11/05/2021] [Indexed: 11/27/2022] Open
Abstract
Background Idiopathic dilated cardiomyopathy (IDCM) myocardial inflammation may be associated with external triggering factors such as infectious agents. Here, we searched if moderate/severe heart transplantation rejection is related to the presence of myocardial inflammation in IDCM explanted hearts, associated with microbial communities. Method Receptor myocardial samples from 18 explanted hearts were separated into groups according to post-transplant outcome: persistent moderate rejection (PMR; n = 6), moderate rejection (MR; n = 7) that regressed after pulse therapy, and no rejection (NR; n = 5)/light intensity rejection. Inflammation was quantified through immunohistochemistry (IHC), and infectious agents were evaluated by IHC, molecular biology, in situ hybridization technique, and transmission electron microscopy (TEM). Results NR presented lower numbers of macrophages, as well as B cells (p = 0.0001), and higher HLA class II expression (p ≤ 0.0001). PMR and MR showed higher levels of Mycoplasma pneumoniae (p = 0.003) and hepatitis B core (p = 0.0009) antigens. NR presented higher levels of parvovirus B19 (PVB19) and human herpes virus 6 (HHV6) and a positive correlation between Borrelia burgdorferi (Bb) and enterovirus genes. Molecular biology demonstrated the presence of M. pneumoniae, Bb, HHV6, and PVB19 genes in all studied groups. TEM revealed structures compatible with the cited microorganisms. Conclusions This initial study investigating on infectious agents and inflammation in the IDCM explanted hearts showed that the association between M. pneumoniae and hepatitis B core was associated with a worse outcome after HT, represented by MR and PMR, suggesting that different IDCM microbial communities may be contributing to post-transplant myocardial rejection.
Collapse
Affiliation(s)
- Jaqueline de Jesus Pereira
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Renata Nishiyama Ikegami
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Joyce Tiyeko Kawakami
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Shérrira Menezes Garavelo
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Marcia Martins Reis
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Suely Aparecida Pinheiro Palomino
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Sandrigo Mangini
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Camila Rodrigues Moreno
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Samar Freschi de Barros
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Aline Rodrigues Souza
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Maria de Lourdes Higuchi
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
Greer M, Elnaggar J, Taylor CM, Shen L. Mycoplasma decontamination in Chlamydia trachomatis culture: a curative approach. Pathog Dis 2021; 79:6464140. [PMID: 34918079 DOI: 10.1093/femspd/ftab056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/14/2021] [Indexed: 11/14/2022] Open
Abstract
Mycoplasma contamination of cell culture represents a serious problem in research and decontamination from cell-propagated obligate intracellular bacteria has proven challenging. Here, we presented an optimized protocol to remove Mycoplasma from contaminated Chlamydia trachomatis culture. A stepwise procedure of Mycoplasma removal entails (i) incubation in nonionic detergent containing solution, and (ii) separation of viable chlamydial organisms by fluorescence-activated cell sorting (FACS), followed by subcloning using a focus-forming assay. We also adapted a polymerase chain reaction (PCR) assay using paired universal and Mycoplasma-specific primers, which are distinguishable from the C. trachomatis counterparts, in combination with Sanger sequencing to determine the presence of mycoplasmas' 16S rRNA genes. These integrated approaches allow for full removal of Mycoplasma, as verified by the improved PCR assay, without compromising the capacity of viable C. trachomatis to adapt to new infection in epithelial cells. Some pitfalls during the Mycoplasma decontamination process are discussed.
Collapse
Affiliation(s)
- Madison Greer
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Jacob Elnaggar
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Christopher M Taylor
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Li Shen
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
26
|
Ferreira G, Santander A, Savio F, Guirado M, Sobrevia L, Nicolson GL. SARS-CoV-2, Zika viruses and mycoplasma: Structure, pathogenesis and some treatment options in these emerging viral and bacterial infectious diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166264. [PMID: 34481867 PMCID: PMC8413106 DOI: 10.1016/j.bbadis.2021.166264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/22/2021] [Accepted: 08/30/2021] [Indexed: 01/28/2023]
Abstract
The molecular evolution of life on earth along with changing environmental, conditions has rendered mankind susceptible to endemic and pandemic emerging infectious diseases. The effects of certain systemic viral and bacterial infections on morbidity and mortality are considered as examples of recent emerging infections. Here we will focus on three examples of infections that are important in pregnancy and early childhood: SARS-CoV-2 virus, Zika virus, and Mycoplasma species. The basic structural characteristics of these infectious agents will be examined, along with their general pathogenic mechanisms. Coronavirus infections, such as caused by the SARS-CoV-2 virus, likely evolved from zoonotic bat viruses to infect humans and cause a pandemic that has been the biggest challenge for humanity since the Spanish Flu pandemic of the early 20th century. In contrast, Zika Virus infections represent an expanding infectious threat in the context of global climate change. The relationship of these infections to pregnancy, the vertical transmission and neurological sequels make these viruses highly relevant to the topics of this special issue. Finally, mycoplasmal infections have been present before mankind evolved, but they were rarely identified as human pathogens until recently, and they are now recognized as important coinfections that are able to modify the course and prognosis of various infectious diseases and other chronic illnesses. The infectious processes caused by these intracellular microorganisms are examined as well as some general aspects of their pathogeneses, clinical presentations, and diagnoses. We will finally consider examples of treatments that have been used to reduce morbidity and mortality of these infections and discuss briefly the current status of vaccines, in particular, against the SARS-CoV-2 virus. It is important to understand some of the basic features of these emerging infectious diseases and the pathogens involved in order to better appreciate the contributions of this special issue on how infectious diseases can affect human pregnancy, fetuses and neonates.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay.
| | - Axel Santander
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Florencia Savio
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Mariana Guirado
- Department of Infectious Diseases, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaeology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; Medical School (Faculty of Medicine), São Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston QLD 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (UMCG), 9713GZ Groningen, the Netherlands
| | - Garth L Nicolson
- Department of Molecular Pathology, The Institute for Molecular Medicine, Huntington Beach, CA, USA
| |
Collapse
|
27
|
Rüger N, Sid H, Meens J, Szostak MP, Baumgärtner W, Bexter F, Rautenschlein S. New Insights into the Host-Pathogen Interaction of Mycoplasma gallisepticum and Avian Metapneumovirus in Tracheal Organ Cultures of Chicken. Microorganisms 2021; 9:microorganisms9112407. [PMID: 34835532 PMCID: PMC8618481 DOI: 10.3390/microorganisms9112407] [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: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 01/04/2023] Open
Abstract
Respiratory pathogens are a health threat for poultry. Co-infections lead to the exacerbation of clinical symptoms and lesions. Mycoplasma gallisepticum (M. gallispeticum) and Avian Metapneumovirus (AMPV) are two avian respiratory pathogens that co-circulate worldwide. The knowledge about the host-pathogen interaction of M. gallispeticum and AMPV in the chicken respiratory tract is limited. We aimed to investigate how co-infections affect the pathogenesis of the respiratory disease and whether the order of invading pathogens leads to changes in host-pathogen interaction. We used chicken tracheal organ cultures (TOC) to investigate pathogen invasion and replication, lesion development, and selected innate immune responses, such as interferon (IFN) α, inducible nitric oxide synthase (iNOS) and IFNλ mRNA expression levels. We performed mono-inoculations (AMPV or M. gallispeticum) or dual-inoculations in two orders with a 24-h interval between the first and second pathogen. Dual-inoculations compared to mono-inoculations resulted in more severe host reactions. Pre-infection with AMPV followed by M. gallispeticum resulted in prolonged viral replication, more significant innate immune responses, and lesions (p < 0.05). AMPV as the secondary pathogen impaired the bacterial attachment process. Consequently, the M. gallispeticum replication was delayed, the innate immune response was less pronounced, and lesions appeared later. Our results suggest a competing process in co-infections and offer new insights in disease processes.
Collapse
Affiliation(s)
- Nancy Rüger
- Clinic for Poultry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (N.R.); (F.B.)
| | - Hicham Sid
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Munich, Germany;
| | - Jochen Meens
- Institute for Microbiology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Michael P. Szostak
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Frederik Bexter
- Clinic for Poultry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (N.R.); (F.B.)
| | - Silke Rautenschlein
- Clinic for Poultry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (N.R.); (F.B.)
- Correspondence: ; Tel.: +49-511-953-8779
| |
Collapse
|
28
|
Shedko ED, Goloveshkina EN, Akimkin VG. Molecular epidemiology and antimicrobials resistance mechanism of Mycoplasma genitlaium. VESTNIK DERMATOLOGII I VENEROLOGII 2021. [DOI: 10.25208/vdv1192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Currently, infections caused by Mycoplasma genitalium are ones the most common sexually transmitted infections. Their prevalence is varied from 1.3% to 15.9%. Infections caused by M.genitalium may lead to urethritis in men and a wide spectrum of diseases in women. Antibiotic resistance now is one of the most emerging problems both in the scientific and in the healthcare fields. The usage of antimicrobials inhibiting cell wall synthesis for the treatment of M.genitalium is ineffective, and resistance to macrolides and fluoroquinolones is increasing rapidly. M.genitalium infections diagnostics is complicated due to specific conditions and duration of culture methods. The usage of nucleic acid amplification techniques is the most relevant for laboratory diagnostics, and is used in existing assays. This review compiles current data on the prevalence, molecular mechanisms of pathogenesis and antibiotic resistance, as well as diagnostics methods of M.genitalium.
Collapse
|
29
|
Protein cleavage influences surface protein presentation in Mycoplasma pneumoniae. Sci Rep 2021; 11:6743. [PMID: 33762641 PMCID: PMC7990945 DOI: 10.1038/s41598-021-86217-y] [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: 07/01/2020] [Accepted: 02/23/2021] [Indexed: 01/31/2023] Open
Abstract
Mycoplasma pneumoniae is a significant cause of pneumonia and post infection sequelae affecting organ sites distant to the respiratory tract are common. It is also a model organism where extensive 'omics' studies have been conducted to gain insight into how minimal genome self-replicating organisms function. An N-terminome study undertaken here identified 4898 unique N-terminal peptides that mapped to 391 (56%) predicted M. pneumoniae proteins. True N-terminal sequences beginning with the initiating methionine (iMet) residue from the predicted Open Reading Frame (ORF) were identified for 163 proteins. Notably, almost half (317; 46%) of the ORFS derived from M. pneumoniae strain M129 are post-translationally modified, presumably by proteolytic processing, because dimethyl labelled neo-N-termini were characterised that mapped beyond the predicted N-terminus. An analysis of the N-terminome describes endoproteolytic processing events predominately targeting tryptic-like sites, though cleavages at negatively charged residues in P1' (D and E) with lysine or serine/alanine in P2' and P3' positions also occurred frequently. Surfaceome studies identified 160 proteins (23% of the proteome) to be exposed on the extracellular surface of M. pneumoniae. The two orthogonal methodologies used to characterise the surfaceome each identified the same 116 proteins, a 72% (116/160) overlap. Apart from lipoproteins, transporters, and adhesins, 93/160 (58%) of the surface proteins lack signal peptides and have well characterised, canonical functions in the cell. Of the 160 surface proteins identified, 134 were also targets of endo-proteolytic processing. These processing events are likely to have profound implications for how the host immune system recognises and responds to M. pneumoniae.
Collapse
|
30
|
Yiwen C, Yueyue W, Lianmei Q, Cuiming Z, Xiaoxing Y. Infection strategies of mycoplasmas: Unraveling the panoply of virulence factors. Virulence 2021; 12:788-817. [PMID: 33704021 PMCID: PMC7954426 DOI: 10.1080/21505594.2021.1889813] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Mycoplasmas, the smallest bacteria lacking a cell wall, can cause various diseases in both humans and animals. Mycoplasmas harbor a variety of virulence factors that enable them to overcome numerous barriers of entry into the host; using accessory proteins, mycoplasma adhesins can bind to the receptors or extracellular matrix of the host cell. Although the host immune system can eradicate the invading mycoplasma in most cases, a few sagacious mycoplasmas employ a series of invasion and immune escape strategies to ensure their continued survival within their hosts. For instance, capsular polysaccharides are crucial for anti-phagocytosis and immunomodulation. Invasive enzymes degrade reactive oxygen species, neutrophil extracellular traps, and immunoglobulins. Biofilm formation is important for establishing a persistent infection. During proliferation, successfully surviving mycoplasmas generate numerous metabolites, including hydrogen peroxide, ammonia and hydrogen sulfide; or secrete various exotoxins, such as community-acquired respiratory distress syndrome toxin, and hemolysins; and express various pathogenic enzymes, all of which have potent toxic effects on host cells. Furthermore, some inherent components of mycoplasmas, such as lipids, membrane lipoproteins, and even mycoplasma-generated superantigens, can exert a significant pathogenic impact on the host cells or the immune system. In this review, we describe the proposed virulence factors in the toolkit of notorious mycoplasmas to better understand the pathogenic features of these bacteria, along with their pathogenic mechanisms.
Collapse
Affiliation(s)
- Chen Yiwen
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - Wu Yueyue
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - Qin Lianmei
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - Zhu Cuiming
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - You Xiaoxing
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| |
Collapse
|
31
|
Hall JA, Isaiah A, Bobe G, Estill CT, Bishop-Stewart JK, Davis TZ, Suchodolski JS, Pirelli GJ. Feeding selenium-biofortified alfalfa hay during the preconditioning period improves growth, carcass weight, and nasal microbial diversity of beef calves. PLoS One 2020; 15:e0242771. [PMID: 33259499 PMCID: PMC7707589 DOI: 10.1371/journal.pone.0242771] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/09/2020] [Indexed: 01/04/2023] Open
Abstract
We previously reported that feeding Se-biofortified alfalfa hay to weaned beef calves in a preconditioning program decreases morbidity and mortality during the feedlot period. To understand the mode of action by which supranutritional Se supplementation supports calf health, we examined the effect of agronomic Se-biofortification on nasal microbiome and fecal parasites. Recently weaned Angus-cross beef calves (n = 30) were randomly assigned to two groups and fed an alfalfa hay-based diet for 9 weeks in a preconditioning program. Alfalfa hay was harvested from fields fertilized with sodium selenate at a rate of 0 or 90 g Se/ha. Calculated Se intake from dietary sources was 1.09 and 27.45 mg Se/calf per day for calves consuming alfalfa hay with Se concentrations of 0.06 and 3.47 mg Se/kg dry matter, respectively. Feeding Se-biofortified alfalfa hay for 9 weeks was effective at increasing whole-blood Se concentrations (556 ± 11 vs 140 ± 11 ng/mL; P < 0.001) and increasing body weight (PTreatment, = 0.03) in weaned beef calves. Slaughter yield grades were higher for calves that had been fed Se-enriched alfalfa hay during the preconditioning period (PTreatment = 0.008). No significant differences were observed in fecal parasite load, which remained low. The nasal microbiome and microbiota diversity within calves and across calves expanded from weaning (week 0) to the feedlot period (week 12), which was promoted by feeding Se-biofortified alfalfa hay. Especially concerning was the expansion of nasal Mycoplasmataceae in the feedlot, which reached over 50% of the total microbiota in some calves. In conclusion, we identified dietary Se-biofortified alfalfa hay as a potential promoter of nasal microbiome genome and microbiota diversity, which may explain in part high-Se benefits for prevention of bovine respiratory disease complex in beef calves.
Collapse
Affiliation(s)
- Jean A. Hall
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States of America
- * E-mail:
| | - Anitha Isaiah
- Gastrointestinal Laboratory, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Texas A&M University, TX, United States of America
| | - Gerd Bobe
- Department of Animal and Rangeland Sciences, College of Agricultural Sciences, Oregon State University, Corvallis, OR, United States of America
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States of America
| | - Charles T. Estill
- Department of Animal and Rangeland Sciences, College of Agricultural Sciences, Oregon State University, Corvallis, OR, United States of America
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States of America
| | - Janell K. Bishop-Stewart
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States of America
| | - T. Zane Davis
- USDA-ARS-Poisonous Plant Research Lab, Logan, UT, United States of America
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Texas A&M University, TX, United States of America
| | - Gene J. Pirelli
- Department of Animal and Rangeland Sciences, College of Agricultural Sciences, Oregon State University, Corvallis, OR, United States of America
| |
Collapse
|
32
|
Luo H, He J, Qin L, Chen Y, Chen L, Li R, Zeng Y, Zhu C, You X, Wu Y. Mycoplasma pneumoniae lipids license TLR-4 for activation of NLRP3 inflammasome and autophagy to evoke a proinflammatory response. Clin Exp Immunol 2020; 203:66-79. [PMID: 32894580 DOI: 10.1111/cei.13510] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/15/2020] [Accepted: 08/21/2020] [Indexed: 12/23/2022] Open
Abstract
Mycoplasma pneumoniae is an obligate pathogen that causes pneumonia, tracheobronchitis, pharyngitis and asthma in humans. It is well recognized that membrane lipoproteins are immunostimulants exerting as lipopolysaccharides (LPS) and play a crucial role in the pathogenesis of inflammatory responses upon M. pneumoniae infection. Here, we report that the M. pneumoniae-derived lipids are another proinflammatory agents. Using an antibody-neutralizing assay, RNA interference or specific inhibitors, we found that Toll-like receptor 4 (TLR-4) is essential for M. pneumoniae lipid-induced tumour necrosis factor (TNF)-α and interleukin (IL)-1β production. We also demonstrate that NLR family pyrin domain containing 3 inflammasome (NLRP3) inflammasome, autophagy and nuclear factor kappa B (NF-κB)-dependent pathways are critical for the secretion of proinflammatory cytokines, while inhibition of TLR-4 significantly abrogates these events. Further characterization revealed that autophagy-mediated inflammatory responses involved the activation of NF-κB. In addition, the activation of NF-κB promoted lipid-induced autophagosome formation, as revealed by assays using pharmacological inhibitors, 3-methyladenine (3-MA) and Bay 11-7082, or silencing of atg5 and beclin-1. These findings suggest that, unlike the response to lipoprotein stimulation, the inflammation in response to M. pneumoniae lipids is mediated by the TLR-4 pathway, which subsequently initiates the activation of NLRP3 inflammasome and formation of a positive feedback loop between autophagy and NF-κB signalling cascade, ultimately promoting TNF-α and Il-1β production in macrophages.
Collapse
Affiliation(s)
- H Luo
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China.,Department of Clinical Laboratory, The Affiliated Nanhua Hospital of University of South China, Hengyang, China
| | - J He
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital of University of South China, Hengyang, China
| | - L Qin
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - Y Chen
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - L Chen
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - R Li
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - Y Zeng
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - C Zhu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - X You
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| | - Y Wu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, China
| |
Collapse
|
33
|
Cacciotto C, Dessì D, Cubeddu T, Cocco AR, Pisano A, Tore G, Fiori PL, Rappelli P, Pittau M, Alberti A. MHO_0730 as a Surface-Exposed Calcium-Dependent Nuclease of Mycoplasma hominis Promoting Neutrophil Extracellular Trap Formation and Escape. J Infect Dis 2020; 220:1999-2008. [PMID: 31420650 DOI: 10.1093/infdis/jiz406] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/08/2019] [Indexed: 12/22/2022] Open
Abstract
Mycoplasma lipoproteins play a relevant role in pathogenicity and directly interact with the host immune system. Among human mycoplasmas, Mycoplasma hominis is described as a commensal bacterium that can be associated with a number of genital and extragenital conditions. Mechanisms of M. hominis pathogenicity are still largely obscure, and only a limited number of proteins have been associated with virulence. The current study focused on investigating the role of MHO_0730 as a virulence factor and demonstrated that MHO_0730 is a surface lipoprotein, potentially expressed in vivo during natural infection, acting both as a nuclease with its amino acidic portion and as a potent inducer of Neutrophil extracellular trapsosis with its N-terminal lipid moiety. Evidence for M. hominis neutrophil extracellular trap escape is also presented. Results highlight the relevance of MHO_0730 in promoting infection and modulation and evasion of innate immunity and provide additional knowledge on M. hominis virulence and survival in the host.
Collapse
Affiliation(s)
- Carla Cacciotto
- Department of Veterinary Medicine, University of Sassari, Italy
| | - Daniele Dessì
- Department of Biomedical Sciences, University of Sassari, Italy.,Mediterranean Center for Disease Control, University of Sassari, Italy
| | - Tiziana Cubeddu
- Department of Veterinary Medicine, University of Sassari, Italy
| | - Anna Rita Cocco
- Department of Biomedical Sciences, University of Sassari, Italy
| | - Andrea Pisano
- Department of Veterinary Medicine, University of Sassari, Italy
| | - Gessica Tore
- Department of Veterinary Medicine, University of Sassari, Italy
| | - Pier Luigi Fiori
- Department of Biomedical Sciences, University of Sassari, Italy.,Mediterranean Center for Disease Control, University of Sassari, Italy
| | - Paola Rappelli
- Department of Biomedical Sciences, University of Sassari, Italy.,Mediterranean Center for Disease Control, University of Sassari, Italy
| | - Marco Pittau
- Department of Veterinary Medicine, University of Sassari, Italy.,Mediterranean Center for Disease Control, University of Sassari, Italy
| | - Alberto Alberti
- Department of Veterinary Medicine, University of Sassari, Italy.,Mediterranean Center for Disease Control, University of Sassari, Italy
| |
Collapse
|
34
|
Chen L, Chen J, Ramesh T, Seshadri VD, Zhu L. Zinc oxide nanoparticles from Corydalis yanhusuo attenuated the mycoplasmal pneumonia in mice through inhibiting the MAPKs signaling pathway. Microb Pathog 2020; 147:104270. [PMID: 32446872 DOI: 10.1016/j.micpath.2020.104270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND The Mycoplasma pneumoniae (M.pneumoniae) was accounted to 3-10% of total pneumonia incidences. In recent decades, metallic nanoparticles were extensively examined as nano-antibiotics. OBJECTIVE In this investigation, we intended to inspect the therapeutic potential of Zinc oxide nanoparticles (ZnONPs) from (Corydalis yanhusuo) C. yanhusuo against the mycoplasma infected pneumonia in mice. METHODOLOGY The ZnONPs were formulated via green route technique and characterized by UV-vis spectroscopy, transmission electron microscopy, Fourier transform infrared technique, and atomic force microscopy. The antimicrobial activity of formulated ZnONPs was tested by well diffusion method. The total protein, interleukin-1 (IL-1), interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor alpha (TNF-α) and transforming growth factor (TGF) status in the BALF of M. pneumonia infected animals were investigated via kit method. The expressions of ERK1/2, JNK1/2, and NF-κB were examined through the Western blotting. The Histopathological analysis of lung tissues of experimental animals was done. RESULTS The UV-vis spectroscopy and TEM examinations were proved the existence of CY-ZnONPs. The formulated CY-ZnONPs were displayed the potential antimicrobial activity. The supplementation of CY-ZnONPs were noticeably diminished the total protein and IL-6, IL-8, and TNF-α levels in the BALF of pneumonia mice. The ERK1/2, JNK1/2, and NF-κB expressions were appreciably diminished in the CY-ZnONPs supplemented mice. It also reduced the inflammatory cells penetration, and exhibited normal tissue arrangements in the lung tissues of pneumonia mice. CONCLUSION The findings of this investigation were proved that the synthesized CY-ZnONPs has the potential to ameliorate the M. pneumoniae infected pneumonia in investigational mice.
Collapse
Affiliation(s)
- Liang Chen
- Department of Infectious Disease, Beijing Jishuitan Hospital, 4 Th Medical College of Peking University, Beijing, 100096, China.
| | - Jun Chen
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Vidya Devanathadesikan Seshadri
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Limei Zhu
- Department of Chronic Disease and Infectious Disease Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
35
|
Almeida HMS, Mechler-Dreibi ML, Sonálio K, Ferraz MES, Storino GY, Barbosa FO, Maes D, Montassier HJ, de Oliveira LG. Cytokine expression and Mycoplasma hyopneumoniae burden in the development of lung lesions in experimentally inoculated pigs. Vet Microbiol 2020; 244:108647. [PMID: 32402328 DOI: 10.1016/j.vetmic.2020.108647] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/28/2020] [Accepted: 03/17/2020] [Indexed: 12/19/2022]
Abstract
This study aimed to assess immunopathological factors and M. hyopneumoniae (M. hyo) load in macroscopic lesion formation at four timepoints after experimental infection of swine. To do this, 24 M. hyo-free pigs were divided into two groups: non-inoculated control (n = 8) and inoculated (n = 16). At day 0 post-infection (dpi), animals of infected group were intratracheally inoculated with 5 mL of lung inoculum containing 107 CCU (Color Changing Units) ∕mL of M. hyo strain 232, while control group was mock infected with 5 mL of sterilized Friis medium. At 14, 28, 42 and 56 dpi, four animals from the infected group and two from the control group were euthanized and necropsied. The extent of macroscopic lung lobe lesions was visually assessed, scored and lesion samples (qPCR, histopathology and gene expression) were collected. The macroscopic lesion score and estimated M. hyo load (in copies/μL) at the different timepoints were: 14 dpi: 18.5 %-1.55 × 103 copies∕μL; 28dpi: 15.8 %-8.4 × 103 copies∕μL; 42 dpi: 7.0 %-3.2 × 104 copies∕μL and 56 dpi: 6.3 %-1.11 × 105 copies∕μL; Significant and positive correlations between macroscopic lung lesion and the pathogen load were found (coefficient range: 0.77-0.99). The cytokine's IL-6 (0.73) and INF-γ (-0.69) gene expression were significantly (p < 0.05) correlated to macroscopic lung lesion score while IL-8, TNF- α, IL-1α and IL-1β were associated to other pathological effects such as losses in average daily weight gain and microscopic lesion score. The results provide a better understanding about the pathogenicity of M. hyo strain 232 and the host-pathogen interactions, which may be helpful for the development of new treatments or control measures.
Collapse
Affiliation(s)
- Henrique M S Almeida
- São Paulo State University (Unesp), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil
| | - Marina L Mechler-Dreibi
- São Paulo State University (Unesp), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil
| | - Karina Sonálio
- São Paulo State University (Unesp), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil
| | - Maria Eugênia S Ferraz
- São Paulo State University (Unesp), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil
| | - Gabriel Y Storino
- São Paulo State University (Unesp), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil
| | - Fernanda O Barbosa
- São Paulo State University (Unesp), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil
| | - Dominiek Maes
- Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Hélio J Montassier
- São Paulo State University (Unesp), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil
| | - Luis G de Oliveira
- São Paulo State University (Unesp), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, Brazil.
| |
Collapse
|
36
|
Proteases as Secreted Exoproteins in Mycoplasmas from Ruminant Lungs and Their Impact on Surface-Exposed Proteins. Appl Environ Microbiol 2019; 85:AEM.01439-19. [PMID: 31540994 DOI: 10.1128/aem.01439-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/10/2019] [Indexed: 12/29/2022] Open
Abstract
Many mycoplasma species are isolated from the ruminant lungs as either saprophytes or true pathogens. These wall-less bacteria possess a minimal genome and reduced metabolic capabilities. Accordingly, they rely heavily on their hosts for the supply of essential metabolites and, notably, peptides. Seven of 13 ruminant lung-associated Mycoplasma (sub)species were shown to possess caseinolytic activity when grown in rich media and assessed with a quantitative fluorescence test. For some species, this activity was detected in spent medium, an indication that proteases were secreted outside the mycoplasma cells. To identify these proteases, we incubated concentrated washed cell pellets in a defined medium and analyzed the supernatants by tandem mass spectrometry. Secreted-protease activity was detected mostly in the species belonging to the Mycoplasma mycoides cluster (MMC) and, to a lesser extent, in Mycoplasma bovirhinis Analyzing a Mycoplasma mycoides subsp. capri strain, chosen as a model, we identified 35 expressed proteases among 55 predicted coding genes, of which 5 were preferentially found in the supernatant. Serine protease S41, acquired by horizontal gene transfer, was responsible for the caseinolytic activity, as demonstrated by zymography and mutant analysis. In an M. capricolum mutant, inactivation of the S41 protease resulted in marked modification of the expression or secretion of 17 predicted surface-exposed proteins. This is an indication that the S41 protease could have a role in posttranslational cleavage of surface-exposed proteins and ectodomain shedding, whose physiological impacts still need to be explored.IMPORTANCE Few studies pertaining to proteases in ruminant mycoplasmas have been reported. Here, we focus on proteases that are secreted outside the mycoplasma cell using a mass spectrometry approach. The most striking result is the identification, within the Mycoplasma mycoides cluster, of a serine protease that is exclusively detected outside the mycoplasma cells and is responsible for casein digestion. This protease may also be involved in the posttranslational processing of surface proteins, as suggested by analysis of mutants showing a marked reduction in the secretion of extracellular proteins. By analogy, this finding may help increase understanding of the mechanisms underlying this ectodomain shedding in other mycoplasma species. The gene encoding this protease is likely to have been acquired via horizontal gene transfer from Gram-positive bacteria and sortase-associated surface proteases. Whether this protease and the associated ectodomain shedding are related to virulence has yet to be ascertained.
Collapse
|
37
|
Qin L, Chen Y, You X. Subversion of the Immune Response by Human Pathogenic Mycoplasmas. Front Microbiol 2019; 10:1934. [PMID: 31497004 PMCID: PMC6712165 DOI: 10.3389/fmicb.2019.01934] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
Mycoplasmas are a large group of prokaryotes which is believed to be originated from Gram-positive bacteria via degenerative evolution, and mainly capable of causing a wide range of human and animal infections. Although innate immunity and adaptive immunity play crucial roles in preventing mycoplasma infection, immune response that develops after infection fails to completely eliminate this bacterium under certain circumstances. Thus, it is reasonable to speculate that mycoplasmas employ some mechanisms to deal with coercion of host defense system. In this review, we will highlight and provide a comprehensive overview of immune evasion strategies that have emerged in mycoplasma infection, which can be divided into four aspects: (i) Molecular mimicry and antigenic variation on the surface of the bacteria to evade the immune surveillance; (ii) Overcoming the immune effector molecules assaults: Induction of detoxified enzymes to degradation of reactive oxygen species; Expression of nucleases to degrade the neutrophil extracellular traps to avoid killing by Neutrophil; Capture and cleavage of immunoglobulins to evade humoral immune response; (iii) Persistent survival: Invading into the host cell to escape the immune damage; Formation of a biofilm to establish a persistent infection; (iv) Modulation of the immune system to down-regulate the intensity of immune response. All of these features increase the probability of mycoplasma survival in the host and lead to a persistent, chronic infections. A profound understanding on the mycoplasma to subvert the immune system will help us to better understand why mycoplasma is so difficult to eradicate and ultimately provide new insights on the development of therapeutic regimens against this bacterium in future.
Collapse
Affiliation(s)
- Lianmei Qin
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Yiwen Chen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xiaoxing You
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
38
|
Schumacher M, Nicholson P, Stoffel MH, Chandran S, D’Mello A, Ma L, Vashee S, Jores J, Labroussaa F. Evidence for the Cytoplasmic Localization of the L-α-Glycerophosphate Oxidase in Members of the " Mycoplasma mycoides Cluster". Front Microbiol 2019; 10:1344. [PMID: 31275271 PMCID: PMC6593217 DOI: 10.3389/fmicb.2019.01344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022] Open
Abstract
Members of the "Mycoplasma mycoides cluster" are important animal pathogens causing diseases including contagious bovine pleuropneumonia and contagious caprine pleuropneumonia, which are of utmost importance in Africa or Asia. Even if all existing vaccines have shortcomings, vaccination of herds is still considered the best way to fight mycoplasma diseases, especially with the recent and dramatic increase of antimicrobial resistance observed in many mycoplasma species. A new generation of vaccines will benefit from a better understanding of the pathogenesis of mycoplasmas, which is very patchy up to now. In particular, surface-exposed virulence traits are likely to induce a protective immune response when formulated in a vaccine. The candidate virulence factor L-α-glycerophosphate oxidase (GlpO), shared by many mycoplasmas including Mycoplasma pneumoniae, was suggested to be a surface-exposed enzyme in Mycoplasma mycoides subsp. mycoides responsible for the production of hydrogen peroxide directly into the host cells. We produced a glpO isogenic mutant GM12::YCpMmyc1.1-ΔglpO using in-yeast synthetic genomics tools including the tandem-repeat endonuclease cleavage (TREC) technique followed by the back-transplantation of the engineered genome into a mycoplasma recipient cell. GlpO localization in the mutant and its parental strain was assessed using scanning electron microscopy (SEM). We obtained conflicting results and this led us to re-evaluate the localization of GlpO using a combination of in silico and in vitro techniques, such as Triton X-114 fractionation or tryptic shaving followed by immunoblotting. Our in vitro results unambiguously support the finding that GlpO is a cytoplasmic protein throughout the "Mycoplasma mycoides cluster." Thus, the use of GlpO as a candidate vaccine antigen is unlikely to induce a protective immune response.
Collapse
Affiliation(s)
- Melanie Schumacher
- Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland
| | - Pamela Nicholson
- Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland
| | | | | | - Adonis D’Mello
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Li Ma
- J. Craig Venter Institute, Rockville, MD, United States
| | - Sanjay Vashee
- J. Craig Venter Institute, Rockville, MD, United States
| | - Joerg Jores
- Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland
| | - Fabien Labroussaa
- Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland
| |
Collapse
|
39
|
Haodang L, Lianmei Q, Ranhui L, Liesong C, Jun H, Yihua Z, Cuiming Z, Yimou W, Xiaoxing Y. HO-1 mediates the anti-inflammatory actions of Sulforaphane in monocytes stimulated with a mycoplasmal lipopeptide. Chem Biol Interact 2019; 306:10-18. [PMID: 30965051 DOI: 10.1016/j.cbi.2019.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/24/2019] [Accepted: 04/04/2019] [Indexed: 12/14/2022]
Abstract
Exposure to Mycoplasma pneumoniae leads to lung inflammation through a host defense pathway. Increasing evidence has indicated that the mycoplasma-derived membrane lipoprotein, or its analogue macrophage-activating lipopeptide-2 (MALP-2), excretes LPS as an immune system-stimulating substance and plays a crucial role in pathological injury during M. pneumoniae infection. It has been established that Sulforaphane confers anti-inflammatory properties. However, the underlying mechanism responsible for the inhibitory actions of Sulforaphane in the context of mycoplasmal pneumoniae are poorly understood. Here, we report that Sulforaphane is an inducer of heme oxygenase (HO)-1, a cytoprotective enzyme that catalyzes the degradation of heme through signaling pathways in human monocytes. Sulforaphane stimulated NF-E2-related factor 2 (Nrf2) translocation from the cytosol to the nucleus, and small interfering RNA-mediated knock-down of Nrf2 significantly inhibited Sulforaphane-induced HO-1 expression. Additionally, PI3K/Akt and ROS were also involved in Sulforaphane-induced Nrf2 activation and HO-1 expression, as revealed by the pharmacological inhibitors LY294002 and NAC. Moreover, Sulforaphane treatment inhibited MALP-2-induced pro-inflammatory cytokine secretion and pulmonary inflammation in mice, as well as MALP-2-triggered NF-κB activation. Furthermore, SnPP, a selective inhibitor of HO-1, reversed the inhibitory actions of Sulforaphane, while a carbon monoxide-releasing molecule, CORM-2, caused a significant decrease in MALP-2-induced cytokine secretion. Collectively, these results suggest that Sulforaphane functions as a suppressor of the MALP-2-induced inflammatory response, not only by inhibiting the expression of cytokines and the induction of HO-1 but also by diminishing NF-κB activation in cultured monocytes and the lungs of mice.
Collapse
Affiliation(s)
- Luo Haodang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - Qin Lianmei
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - Li Ranhui
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - Chen Liesong
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - He Jun
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital of University of South China, Hengyang, 421001, China
| | - Zeng Yihua
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - Zhu Cuiming
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - Wu Yimou
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - You Xiaoxing
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China.
| |
Collapse
|