1
|
Yi X, Huang Y, Li X, Xu H, Liu C, Li C, Zeng Q, Luo H, Ye Z, He J, You X. Decoding Mycoplasma Nucleases: Biological Functions and Pathogenesis. Toxins (Basel) 2025; 17:215. [PMID: 40423298 DOI: 10.3390/toxins17050215] [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: 03/09/2025] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Nucleases are critical metabolic enzymes expressed by mycoplasmas to acquire nucleic acid precursors from the host for their parasitic existence. Certain nucleases, either membrane-bound or secreted, not only contribute to the growth of mycoplasmas but also serve as key virulence factors due to their unique spatial structures and physiological activity. The pathogenesis includes, but is not limited to, degradation of host DNA and RNA, leading to disruptions of nucleic acid metabolism and the induction of host cell apoptosis; degradation of neutrophil extracellular traps (NETs), allowing escape from neutrophil-mediated killing; and upregulation of inflammatory molecules to modulate the immune response of the host. Understanding the biological functions of nucleases is essential for gaining deeper insights into the virulence and immune evasion strategies of mycoplasmas, which can inform the development of novel approaches for the prevention, diagnosis, and treatment of mycoplasma infections.
Collapse
Affiliation(s)
- Xinchao Yi
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421002, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Ying Huang
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421002, China
| | - Xinru Li
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Hao Xu
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421002, China
| | - Chang Liu
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Chao Li
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421002, China
| | - Qianrui Zeng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Haodang Luo
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421002, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Zufeng Ye
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421002, China
| | - Jun He
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421002, China
- Department of Clinical Laboratory, The Second Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Xiaoxing You
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421002, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, China
| |
Collapse
|
2
|
Caldeira JLA, Costa DG, Polveiro RC, Gomes do Rêgo ME, Barbosa WF, de Oliveira LL, Moreira MAS. Short communication: Goat mastitis and the formation of neutrophil extracellular traps (NETs). Vet Immunol Immunopathol 2024; 274:110793. [PMID: 38943998 DOI: 10.1016/j.vetimm.2024.110793] [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: 04/17/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 07/01/2024]
Abstract
Mastitis, an inflammation of the mammary gland affecting milk production and quality in dairy herds, is often associated with Staphylococcus spp. in goats. Neutrophils are crucial in combating infections by migrating into milk and deploying various defense strategies, including the release of neutrophil extracellular traps (NETs) composed of DNA, histones, and bactericidal proteins. This study investigated whether NETs are released by goat neutrophils stimulated in vitro by Staphylococcus aureus and Staphylococcus warneri, two common pathogens of goat mastitis. PMNs were isolated from blood from healthy adult goats. We evaluated goat NET formation by stimulating cells with: phorbol 12-myristate 13-acetate (PMA) as a positive control, cytochalasin for inhibition of actin polymerization, S. aureus, and S. warneri. NET formation was observed in response to chemical stimulation and bacterial presence, effectively trapping pathogens. Variations in NET formation between S. aureus and S. warneri suggest pathogen-specific responses. These findings suggest that the formation of NETs may be an important complementary mechanism in the defense against mastitis in goats. In conclusion, this study unveils a novel defense mechanism in goats, indicating the role of NETs against S. aureus and S. warneri in mastitis.
Collapse
Affiliation(s)
- Jéssica Lobo Albuquerque Caldeira
- Bacterial Diseases Laboratory, Department of Preventive Veterinary Medicine and Public Health, Veterinary Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Daiene Gaione Costa
- Bacterial Diseases Laboratory, Department of Preventive Veterinary Medicine and Public Health, Veterinary Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Richard Costa Polveiro
- Bacterial Diseases Laboratory, Department of Preventive Veterinary Medicine and Public Health, Veterinary Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Maria Eduarda Gomes do Rêgo
- Bacterial Diseases Laboratory, Department of Preventive Veterinary Medicine and Public Health, Veterinary Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Wagner Faria Barbosa
- Department of Statistics, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Leandro Licursi de Oliveira
- Immunochemistry and Glycobiology Laboratory, Department of General Biology, Universidade Federal de Viçosa, University Campus, PH Rolfs Avenue, Viçosa, Minas Gerais 36570-000, Brazil
| | - Maria Aparecida Scatamburlo Moreira
- Bacterial Diseases Laboratory, Department of Preventive Veterinary Medicine and Public Health, Veterinary Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil.
| |
Collapse
|
3
|
Bittencourt DDC, Brown DM, Assad-Garcia N, Romero MR, Sun L, Palhares de Melo LAM, Freire M, Glass JI. Minimal Bacterial Cell JCVI-syn3B as a Chassis to Investigate Interactions between Bacteria and Mammalian Cells. ACS Synth Biol 2024; 13:1128-1141. [PMID: 38507598 PMCID: PMC11036491 DOI: 10.1021/acssynbio.3c00513] [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: 08/21/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Abstract
Mycoplasmas are atypical bacteria with small genomes that necessitate colonization of their respective animal or plant hosts as obligate parasites, whether as pathogens, or commensals. Some can grow axenically in specialized complex media yet show only host-cell-dependent growth in cell culture, where they can survive chronically and often through interactions involving surface colonization or internalization. To develop a mycoplasma-based system to identify genes mediating such interactions, we exploited genetically tractable strains of the goat pathogen Mycoplasma mycoides (Mmc) with synthetic designer genomes representing the complete natural organism (minus virulence factors; JCVI-syn1.0) or its reduced counterpart (JCVI-syn3B) containing only those genes supporting axenic growth. By measuring growth of surviving organisms, physical association with cultured human cells (HEK-293T, HeLa), and induction of phagocytosis by human myeloid cells (dHL-60), we determined that JCVI-syn1.0 contained a set of eight genes (MMSYN1-0179 to MMSYN1-0186, dispensable for axenic growth) conferring survival, attachment, and phagocytosis phenotypes. JCVI-syn3B lacked these phenotypes, but insertion of these genes restored cell attachment and phagocytosis, although not survival. These results indicate that JCVI-syn3B may be a powerful living platform to analyze the role of specific gene sets, from any organism, on the interaction with diverse mammalian cells in culture.
Collapse
Affiliation(s)
- Daniela
Matias de C. Bittencourt
- The
J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, California 92037, United States
- Embrapa
Genetic Resources and Biotechnology/National Institute of Science
and Technology − Synthetic Biology, Parque Estação
Biológica, PqEB, Av. W5 Norte (final), Brasília, DF 70770-917, Brazil
| | - David M. Brown
- The
J. Craig Venter Institute, 9605 Medical Center Drive, Suite 150, Rockville, Maryland 20850, United States
| | - Nacyra Assad-Garcia
- The
J. Craig Venter Institute, 9605 Medical Center Drive, Suite 150, Rockville, Maryland 20850, United States
| | - Michaela R. Romero
- The
J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, California 92037, United States
| | - Lijie Sun
- The
J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, California 92037, United States
| | - Luis Alberto M. Palhares de Melo
- Embrapa
Genetic Resources and Biotechnology/National Institute of Science
and Technology − Synthetic Biology, Parque Estação
Biológica, PqEB, Av. W5 Norte (final), Brasília, DF 70770-917, Brazil
| | - Marcelo Freire
- The
J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, California 92037, United States
| | - John I. Glass
- The
J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, California 92037, United States
| |
Collapse
|
4
|
Wang S, Wan Y, Zhang W. The Clinical Value of Systemic Immune Inflammation Index (SII) in Predicting the Severity of Hospitalized Children with Mycoplasma Pneumoniae Pneumonia: A Retrospective Study. Int J Gen Med 2024; 17:935-942. [PMID: 38495920 PMCID: PMC10944171 DOI: 10.2147/ijgm.s451466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Objective The Systemic Immune Inflammation Index (SII), as a novel inflammation biomarker that comprehensively reflects the inflammatory and immune status of the body, has not been reported in studies on Mycoplasma pneumoniae pneumonia (MPP) in children. This study aims to investigate whether SII can serve as an effective indicator for evaluating the condition of MPP. Methods This study recruited a total of 304 hospitalized patients with mycoplasma pneumoniae pneumonia (MPP), including 78 patients with severe MPP (SMPP) and 226 patients with non-SMPP. Univariate analysis using chi-square test, t-test, and Mann-Whitney U-test was conducted to analyze the clinical data of the patients. Logistic regression analysis was employed to identify the main risk factors for SMPP. Receiver operating characteristic curves were plotted to evaluate the potential of using neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), systemic immune-inflammation index (SII), and systemic immune response index (SIRI) to predict the severity of MPP. Results The ROC curve results show that patients with SII values ≥ 699.00 are more likely to develop severe MPP (sensitivity=0.876, specificity=0.987, AUC=0.940), and the predictive value of SII is significantly better than that of NLR, PLR, and SIRI. The results of multivariate logistic regression analysis indicate that SII can serve as a major risk factor for distinguishing non-SMPP from SMPP. Conclusion This study suggests that SII may be an effective indicator for predicting the severity of MPP in children. SII is more sensitive and specific than NLR, PLR, and SIRI in evaluating the condition of MPP.
Collapse
Affiliation(s)
- Shuye Wang
- Bengbu Medical University, Bengbu, 233000, People’s Republic of China
- Department of Pediatrics, Changzhou No.2 People’s Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Yu Wan
- Department of Pediatrics, Changzhou No.2 People’s Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Wenbo Zhang
- Department of Pediatrics, Changzhou No.2 People’s Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| |
Collapse
|
5
|
Baz AA, Hao H, Lan S, Li Z, Liu S, Chen S, Chu Y. Neutrophil extracellular traps in bacterial infections and evasion strategies. Front Immunol 2024; 15:1357967. [PMID: 38433838 PMCID: PMC10906519 DOI: 10.3389/fimmu.2024.1357967] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/26/2024] [Indexed: 03/05/2024] Open
Abstract
Neutrophils are innate immune cells that have a vital role in host defense systems. Neutrophil extracellular traps (NETs) are one of neutrophils' defense mechanisms against pathogens. NETs comprise an ejected lattice of chromatin associated with histones, granular proteins, and cytosolic proteins. They are thought to be an efficient strategy to capture and/or kill bacteria and received intensive research interest in the recent years. However, soon after NETs were identified, it was observed that certain bacteria were able to evade NET entrapment through many different mechanisms. Here, we outline the recent progress of NETs in bacterial infections and the strategies employed by bacteria to evade or withstand NETs. Identifying the molecules and mechanisms that modulate NET release will improve our understanding of the functions of NETs in infections and provide new avenues for the prevention and treatment of bacterial diseases.
Collapse
Affiliation(s)
- Ahmed Adel Baz
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - 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 Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Shimei Lan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, 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 Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, 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 Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, 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 Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, 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 Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Ministry of Agricultural and Rural Affairs, Lanzhou, China
| |
Collapse
|
6
|
Barbosa MS, Sampaio BA, Spergser J, Rosengarten R, Marques LM, Chopra-Dewasthaly R. Mycoplasma agalactiae Vaccines: Current Status, Hurdles, and Opportunities Due to Advances in Pathogenicity Studies. Vaccines (Basel) 2024; 12:156. [PMID: 38400139 PMCID: PMC10892753 DOI: 10.3390/vaccines12020156] [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: 12/20/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Contagious agalactia (CA) is a serious multietiological disease whose classic etiological agent is Mycoplasma agalactiae and which causes high morbidity and mortality rates in infected herds. CA is classified as a notifiable disease by the World Organization for Animal Health due to its significant worldwide economic impact on livestock, primarily involving goat and sheep farms. The emergence of atypical symptoms and strains of M. agalactiae in wildlife ungulates reestablishes its highly plastic genome and is also of great epidemiological significance. Antimicrobial therapy is the main form of control, although several factors, such as intrinsic antibiotic resistance and the selection of resistant strains, must be considered. Available vaccines are few and mostly inefficient. The virulence and pathogenicity mechanisms of M. agalactiae mainly rely on surface molecules that have direct contact with the host. Because of this, they are essential for the development of vaccines. This review highlights the currently available vaccines and their limitations and the development of new vaccine possibilities, especially considering the challenge of antigenic variation and dynamic genome in this microorganism.
Collapse
Affiliation(s)
- Maysa Santos Barbosa
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitoria da Conquista 45029-094, Brazil; (M.S.B.)
| | | | - Joachim Spergser
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, 1210 Wien, Austria
| | - Renate Rosengarten
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, 1210 Wien, Austria
| | - Lucas Miranda Marques
- Department of Biointeraction, Multidisciplinary Institute of Health, Federal University of Bahia, Vitoria da Conquista 45029-094, Brazil; (M.S.B.)
- Department of Microbiology, State University of Santa Cruz (UESC), Ilheus 45662-900, Brazil
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, Sao Paulo 05508-000, Brazil
| | - Rohini Chopra-Dewasthaly
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, 1210 Wien, Austria
| |
Collapse
|
7
|
Jin Z, Jin Q, Chen M, Liu W, Hong H, Jiang Y, Gao X, Qian Y, Wang Z, Liu Q, Wei Z. Toxoplasma gondii-induced neutrophil extracellular traps are relevant to glycolysis, TLR2, and TLR4 MAPK signaling pathway in goats. Parasitol Res 2023; 123:34. [PMID: 38087003 DOI: 10.1007/s00436-023-08041-9] [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: 04/21/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023]
Abstract
Toxoplasma gondii (T. gondii) exhibits a significantly high prevalence of infection in goats, leading to adverse consequences such as abortion and stillbirth in ewes, thereby posing a substantial challenge to the goat farming industry. Neutrophil extracellular traps (NETs) have been shown to capture T. gondii in goats; however, the precise mechanisms underlying NET release in goats remain poorly understood. Therefore, the aim of our research was to elucidate the involved mechanism. We assessed the cytotoxicity of T. gondii on neutrophils using CCK-8 assay, visualized the structure of T. gondii-induced goat NETs through immunofluorescence, quantified ROS release during T. gondii-induced NET formation using fluorescence microplate analysis, and employed inhibitors targeting TLR 2, TLR4, NADPH oxidase, ERK1/2, and P38 MAPK signaling pathways as well as glycolysis to dissect the mechanisms underlying T. gondii-induced NET release. Within 1 h, T. gondii did not exhibit significant cytotoxicity towards neutrophils in our findings. The formation of typical NET structures induced by T. gondii involved DNA, citrullinated histone 3 (citH3), and neutrophil elastase (NE). Additionally, T. gondii significantly stimulated the release of NETs in goats. The process was accompanied by the production of reactive oxygen species (ROS) mediated through NADPH oxidase, p38, and ERK1/2 signaling pathways. Inhibition of these pathways resulted in a decrease in NET release. Moreover, inhibition of TLR 2, TLR4, and glycolysis also led to a reduction in T. gondii-induced NET release. Overall, our study demonstrates that T. gondii can induce characteristic NET structures and elucidates the involvement of various mechanisms including TLR2/TLR4 signaling pathway activation, NADPH oxidase activity modulation via ROS production regulation through p38 MAPK and ERK1/2 signaling pathways, and glycolysis regulation during the innate immune response against T. gondii infection in goats.
Collapse
Affiliation(s)
- Zha Jin
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Qinqin Jin
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Meiyi Chen
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Wei Liu
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Hongrong Hong
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Yuqian Jiang
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Xinxin Gao
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Yuxiao Qian
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Zedong Wang
- Center for Pathogen Biology and Infectious Diseases, International Center of Future Science, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital, Jilin University, Changchun, 130122, Jilin, People's Republic of China
| | - Quan Liu
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Zhengkai Wei
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China.
- College of Veterinary Medicine, Southwest University, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
8
|
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
|
9
|
Gioia G, Severgnini M, Cremonesi P, Castiglioni B, Freeman J, Sipka A, Santisteban C, Wieland M, Gallardo VA, Scott JG, Moroni P, Addis MF. Genomic Characterization of Mycoplasma arginini Isolated from a Housefly on a Dairy Farm and Comparison with Isolates from Bovine Milk and Lung Tissue. Microbiol Spectr 2023; 11:e0301022. [PMID: 37199649 PMCID: PMC10269790 DOI: 10.1128/spectrum.03010-22] [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: 08/08/2022] [Accepted: 04/17/2023] [Indexed: 05/19/2023] Open
Abstract
Mycoplasma mastitis can be highly contagious, unresponsive to treatment, and cause severe economic problems in affected herds. Notable routes of Mycoplasma spp. transmissions are contaminated milking equipment and animal contact through respiratory secretions. Only a few studies report the environment as a possible source of infection. Our group studied the presence of pathogens in houseflies (Musca domestica) in a New York State dairy in the United States. Among others, a Mycoplasma spp. was found in the gut of a housefly captured in the sick pen and identified as M. arginini. Here, we characterized its genome and investigated its relatedness with eight isolates from milk, one isolate from lung tissue collected in the same dairy, and five other dairies in New York State. We applied whole-genome sequencing and phylogenetic analysis based on the sequences of the 16S rRNA gene and 76 conserved proteins. We also assessed an in silico virulence profile by considering a panel of 94 putative virulence genes. As a result of the genome analysis, the housefly M. arginini isolate was highly similar to the milk isolates; interestingly, the similarity was highest with M. arginini isolated from milk on the same dairy farm where the housefly was captured. The housefly and milk M. arginini isolates possessed 54 of the 94 pathogenicity genes considered. Our data support the hypothesis that houseflies are carriers of Mycoplasma spp. and can be considered within the possible roots of environmental transmission of infection in dairy cows. Nevertheless, M. arginini pathogenicity will need to be investigated with dedicated studies. IMPORTANCE It is critical to control the spread of bovine mastitis caused by Mycoplasma spp., as this disease can be highly contagious and have a severe economic impact on affected dairies. A better understanding of possible transmission routes is crucial for infection control and prevention. Based on our data, the composite milk isolates are genetically similar to the housefly isolate. This provides evidence that the same Mycoplasma species found in milk and associated with mastitis can also be isolated from houseflies captured in the dairy environment.
Collapse
Affiliation(s)
- G. Gioia
- Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, New York, USA
| | - M. Severgnini
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy
| | - P. Cremonesi
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, Italy
| | - B. Castiglioni
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, Italy
| | - J. Freeman
- Department of Entomology, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA
| | - A. Sipka
- Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, New York, USA
| | - C. Santisteban
- Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, New York, USA
| | - M. Wieland
- Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, New York, USA
| | - V. Alanis Gallardo
- Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, New York, USA
- Departamento de Medicina Preventiva y Salud Pública, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - J. G. Scott
- Department of Entomology, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA
| | - P. Moroni
- Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, New York, USA
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, Lodi, Italy
- Laboratorio di Malattie Infettive degli Animali-MiLab, University of Milan, Lodi, Italy
| | - M. F. Addis
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, Lodi, Italy
- Laboratorio di Malattie Infettive degli Animali-MiLab, University of Milan, Lodi, Italy
| |
Collapse
|
10
|
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
|
11
|
Assessment of the Impact of a Toll-like Receptor 2 Agonist Synthetic Lipopeptide on Macrophage Susceptibility and Responses to African Swine Fever Virus Infection. Viruses 2022; 14:v14102212. [PMID: 36298767 PMCID: PMC9610641 DOI: 10.3390/v14102212] [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: 08/31/2022] [Revised: 09/25/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
Toll-like receptor 2 (TLR2) ligands are attracting attention as prophylactic and immunopotentiator agents against pathogens, including viruses. We previously reported that a synthetic diacylated lipopeptide (Mag-Pam2Cys_P48) polarized porcine macrophages towards a proinflammatory antimicrobial phenotype. Here, we investigated its role in modulating monocyte-derived macrophage (moMΦ) responses against African swine fever virus (ASFV), the etiological agent of one of the greatest threats to the global pig industry. Two ASFV isolates were compared: the attenuated NH/P68 and the virulent 26544/OG10. No effect on virus infection nor the modulation of surface markers’ expression (MHC I, MHC II DR, CD14, CD16, and CD163) were observed when Mag-Pam2Cys_P48 treated moMΦ were infected using a multiplicity of infection (MOI) of 1. Mag-Pam2Cys_P48 treated moMΦ released higher levels of IL-1α, IL-1β, IL-1Ra, and IL-18 in response to infection with NH/P68 ASFV compared to 26544/OG10-infected and mock-infected controls. Surprisingly, when infected using a MOI of 0.01, the virulent ASFV 26544/OG10 isolate replicated even slightly more efficiently in Mag-Pam2Cys_P48 treated moMΦ. These effects also extended to the treatment of moMΦ with two other lipopeptides: Mag-Pam2Cys_P80 and Mag-Pam2Cys_Mag1000. Our data suggested limited applicability of TLR2 agonists as prophylactic or immunopotentiator agents against virulent ASFV but highlighted the ability of the virulent 26544/OG10 to impair macrophage defenses.
Collapse
|
12
|
Huang J, Hong W, Wan M, Zheng L. Molecular mechanisms and therapeutic target of NETosis in diseases. MedComm (Beijing) 2022; 3:e162. [PMID: 36000086 PMCID: PMC9390875 DOI: 10.1002/mco2.162] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
Evidence shows that neutrophils can protect the host against pathogens in multiple ways, including the formation and release of neutrophil extracellular traps (NETs). NETs are web-like structures composed of fibers, DNA, histones, and various neutrophil granule proteins. NETs can capture and kill pathogens, including bacteria, viruses, fungi, and protozoa. The process of NET formation is called NETosis. According to whether they depend on nicotinamide adenine dinucleotide phosphate (NADPH), NETosis can be divided into two categories: "suicidal" NETosis and "vital" NETosis. However, NET components, including neutrophil elastase, myeloperoxidase, and cell-free DNA, cause a proinflammatory response and potentially severe diseases. Compelling evidence indicates a link between NETs and the pathogenesis of a number of diseases, including sepsis, systemic lupus erythematosus, rheumatoid arthritis, small-vessel vasculitis, inflammatory bowel disease, cancer, COVID-19, and others. Therefore, targeting the process and products of NETosis is critical for treating diseases linked with NETosis. Researchers have discovered that several NET inhibitors, such as toll-like receptor inhibitors and reactive oxygen species scavengers, can prevent uncontrolled NET development. This review summarizes the mechanism of NETosis, the receptors associated with NETosis, the pathology of NETosis-induced diseases, and NETosis-targeted therapy.
Collapse
Affiliation(s)
- Jiayu Huang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western MedicineWest China HospitalSichuan UniversityChengduSichuanChina
| | - Limin Zheng
- Guangdong Province Key Laboratory of Pharmaceutical Functional GenesMOE Key Laboratory of Gene Function and RegulationSchool of Life SciencesSun Yat‐Sen UniversityGuangzhouChina
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouChina
| |
Collapse
|
13
|
Vaccination with Mycoplasma pneumoniae membrane lipoproteins induces IL-17A driven neutrophilia that mediates Vaccine-Enhanced Disease. NPJ Vaccines 2022; 7:86. [PMID: 35906257 PMCID: PMC9336141 DOI: 10.1038/s41541-022-00513-w] [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: 02/28/2022] [Accepted: 06/24/2022] [Indexed: 11/24/2022] Open
Abstract
Bacterial lipoproteins are an often-underappreciated class of microbe-associated molecular patterns with potent immunomodulatory activity. We previously reported that vaccination of BALB/c mice with Mycoplasma pneumoniae (Mp) lipid-associated membrane proteins (LAMPs) resulted in lipoprotein-dependent vaccine enhanced disease after challenge with virulent Mp, though the immune responses underpinning this phenomenon remain poorly understood. Herein, we report that lipoprotein-induced VED in a mouse model is associated with elevated inflammatory cytokines TNF-α, IL-1β, IL-6, IL-17A, and KC in lung lavage fluid and with suppurative pneumonia marked by exuberant neutrophilia in the pulmonary parenchyma. Whole-lung-digest flow cytometry and RNAScope analysis identified multiple cellular sources for IL-17A, and the numbers of IL-17A producing cells were increased in LAMPs-vaccinated/Mp-challenged animals compared to controls. Specific IL-17A or neutrophil depletion reduced disease severity in our VED model—indicating that Mp lipoproteins induce VED in an IL-17A-dependent manner and through exuberant neutrophil recruitment. IL-17A neutralization reduced levels of TNF-α, IL-1β, IL-6, and KC, indicating that IL-17A preceded other inflammatory cytokines. Surprisingly, we found that IL-17A neutralization impaired bacterial clearance, while neutrophil depletion improved it—indicating that, while IL-17A appears to confer both maladaptive and protective responses, neutrophils play an entirely maladaptive role in VED. Given that lipoproteins are found in virtually all bacteria, the potential for lipoprotein-mediated maladaptive inflammatory responses should be taken into consideration when developing vaccines against bacterial pathogens.
Collapse
|
14
|
Liao C, Mao F, Qian M, Wang X. Pathogen-Derived Nucleases: An Effective Weapon for Escaping Extracellular Traps. Front Immunol 2022; 13:899890. [PMID: 35865526 PMCID: PMC9294136 DOI: 10.3389/fimmu.2022.899890] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Since the 2004 publication of the first study describing extracellular traps (ETs) from human neutrophils, several reports have shown the presence of ETs in a variety of different animals and plants. ETs perform two important functions of immobilizing and killing invading microbes and are considered a novel part of the phagocytosis-independent, innate immune extracellular defense system. However, several pathogens can release nucleases that degrade the DNA backbone of ETs, reducing their effectiveness and resulting in increased pathogenicity. In this review, we examined the relevant literature and summarized the results on bacterial and fungal pathogens and parasites that produce nucleases to evade the ET-mediated host antimicrobial mechanism.
Collapse
Affiliation(s)
- Chengshui Liao
- College of Animal Science and Technology/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- *Correspondence: Chengshui Liao, ; Xiaoli Wang,
| | - Fuchao Mao
- College of Animal Science and Technology/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- Animal Diseases and Public Health Engineering Research Center of Henan Province, Luoyang Vocational and Technical College, Luoyang, China
| | - Man Qian
- College of Animal Science and Technology/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
| | - Xiaoli Wang
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
- *Correspondence: Chengshui Liao, ; Xiaoli Wang,
| |
Collapse
|
15
|
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
|
16
|
Schultz BM, Acevedo OA, Kalergis AM, Bueno SM. Role of Extracellular Trap Release During Bacterial and Viral Infection. Front Microbiol 2022; 13:798853. [PMID: 35154050 PMCID: PMC8825568 DOI: 10.3389/fmicb.2022.798853] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/04/2022] [Indexed: 12/20/2022] Open
Abstract
Neutrophils are innate immune cells that play an essential role during the clearance of pathogens that can release chromatin structures coated by several cytoplasmatic and granular antibacterial proteins, called neutrophil extracellular traps (NETs). These supra-molecular structures are produced to kill or immobilize several types of microorganisms, including bacteria and viruses. The contribution of the NET release process (or NETosis) to acute inflammation or the prevention of pathogen spreading depends on the specific microorganism involved in triggering this response. Furthermore, studies highlight the role of innate cells different from neutrophils in triggering the release of extracellular traps during bacterial infection. This review summarizes the contribution of NETs during bacterial and viral infections, explaining the molecular mechanisms involved in their formation and the relationship with different components of such pathogens.
Collapse
Affiliation(s)
- Bárbara M Schultz
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Orlando A Acevedo
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
17
|
Addis MF, Pisanu S, Monistero V, Gazzola A, Penati M, Filipe J, Di Mauro S, Cremonesi P, Castiglioni B, Moroni P, Pagnozzi D, Tola S, Piccinini R. Comparative secretome analysis of Staphylococcus aureus strains with different within-herd intramammary infection prevalence. Virulence 2022; 13:174-190. [PMID: 35030987 PMCID: PMC8765078 DOI: 10.1080/21505594.2021.2024014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus is a major pathogen causing intramammary infection and mastitis in dairy cows. S. aureus genotypes (GT) can differ significantly in their ability to diffuse and persist in the herd; while the association of virulence gene carriage with epidemiological behavior remains unclear, a role for secreted proteins has been postulated. We characterized the secretome of six S. aureus strains belonging to two genotypes with opposite within-herd prevalence, GTB (high) and GTS (low), corresponding to sequence types (ST) 8 and 398, by high-resolution tandem mass spectrometry and differential analysis with Proteome Discoverer. Data are available via ProteomeXchange with identifier PXD029571. Out of 720 identified proteins, 98 were unique or more abundant in GTB/ST8 and 68 in GTS/ST398. GTB/ST8 released more immunoglobulin-binding proteins, complement and antimicrobial peptide inhibitors, enterotoxins, and metabolic enzymes, while GTS/ST398 released more leukocidins, hemolysins, lipases, and peptidases. Furthermore, GTB/ST8 released the von Willebrand factor protein, staphylokinase, and clumping factor B, while GTS released the staphylococcal coagulase and clumping factor A. Hence, GTB/ST8 secretomes indicated a higher propensity for immune evasion and chronicity and GTS/ST398 secretomes for cellular damage and inflammation, consistent with their epidemiological characteristics. Accordingly, GTS/ST398 secretions were significantly more cytotoxic against bovine PBMCs in vitro. Our findings confirm the crucial role of extracellular virulence factors in S. aureus pathogenesis and highlight the need to investigate their differential release adding to gene carriage for a better understanding of the relationship of S. aureus genotypes with epidemiological behavior and, possibly, disease severity.
Collapse
Affiliation(s)
- M Filippa Addis
- Dipartimento Di Medicina Veterinaria, Università Degli Studi Di Milano, Lodi, Italy
| | | | - Valentina Monistero
- Dipartimento Di Medicina Veterinaria, Università Degli Studi Di Milano, Lodi, Italy
| | - Alessandra Gazzola
- Dipartimento Di Medicina Veterinaria, Università Degli Studi Di Milano, Lodi, Italy
| | - Martina Penati
- Dipartimento Di Medicina Veterinaria, Università Degli Studi Di Milano, Lodi, Italy
| | - Joel Filipe
- Dipartimento Di Medicina Veterinaria, Università Degli Studi Di Milano, Lodi, Italy
| | - Susanna Di Mauro
- Dipartimento Di Medicina Veterinaria, Università Degli Studi Di Milano, Lodi, Italy
| | - Paola Cremonesi
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, Italy
| | - Bianca Castiglioni
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, Italy
| | - Paolo Moroni
- Dipartimento Di Medicina Veterinaria, Università Degli Studi Di Milano, Lodi, Italy.,Quality Milk Production Services, Animal Health Diagnostic Center, Cornell University, Ithaca, NY, USA
| | | | - Sebastiana Tola
- Istituto Zooprofilattico Sperimentale Della Sardegna "G. Pegreffi", Sassari, Italy
| | - Renata Piccinini
- Dipartimento Di Medicina Veterinaria, Università Degli Studi Di Milano, Lodi, Italy
| |
Collapse
|
18
|
Filioussis G, Bramis G, Petridou E, Giadinis ND, Nouvel LX, Citti C, Frey J. Mycoplasma agalactiae ST35: a new sequence type with a minimal accessory genome primarily affecting goats. BMC Vet Res 2022; 18:29. [PMID: 35016679 PMCID: PMC8751087 DOI: 10.1186/s12917-021-03128-w] [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: 09/21/2021] [Accepted: 12/20/2021] [Indexed: 09/17/2024] Open
Abstract
Background Mycoplasma agalactiae, causing agent of contagious agalactia, infects domestic small ruminants such as sheep and goats but also wild Caprinae. M. agalactiae is highly contagious and transmitted through oral, respiratory, and mammary routes spreading rapidly in an infected herd. Results In an outbreak of contagious agalactia in a mixed herd of sheep and goats, 80% of the goats were affected displaying swollen udders and loss of milk production but no other symptom such as kerato-conjunctivitis, arthritis or pulmonary distress commonly associated to contagious agalactia. Surprisingly, none of the sheep grazing on a common pasture and belonging to the same farm as the goats were affected. Whole genome sequencing and analysis of M. agalactiae strain GrTh01 isolated from the outbreak, revealed a previously unknown sequence type, ST35, and a particularly small, genome size of 841′635 bp when compared to others available in public databases. Overall, GrTh01 displayed a reduced accessory genome, with repertoires of gene families encoding variable surface proteins involved in host-adhesion and variable antigenicity being scaled down. GrTh01 was also deprived of Integrative Conjugative Element or prophage, and had a single IS element, suggesting that GrTh01 has a limited capacity to adapt and evolve. Conclusions The lack of most of the variable antigens and the Integrative Conjugative Element, both major virulence- and host specificity factors of a M. agalactiae strain isolated from an outbreak affecting particularly goats, indicates the implication of these factors in host specificity. Whole genome sequencing and full assembly of bacterial pathogens provides a most valuable tool for epidemiological and virulence studies of M. agalactiae without experimental infections.
Collapse
Affiliation(s)
- George Filioussis
- Laboratory of Microbiology and Infectious Diseases, Faculty of Veterinary Medicine, School of Health Science, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Georgios Bramis
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, School of Health Science, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Evanthia Petridou
- Laboratory of Microbiology and Infectious Diseases, Faculty of Veterinary Medicine, School of Health Science, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Nektarios D Giadinis
- Clinic of Farm Animals, Faculty of Veterinary Medicine, School of Health Science, Aristotle University of Thessaloniki, St. Voutyra 11, 54627, Thessaloniki, Greece
| | | | | | - Joachim Frey
- Vetsuisse Faculty, University of Bern, Laenggasstrasse 120, 3001, Bern, Switzerland.
| |
Collapse
|
19
|
Qiu J, Ge J, Cao L. D-dimer: The Risk Factor of Children's Severe Mycoplasma Pneumoniae Pneumonia. Front Pediatr 2022; 10:828437. [PMID: 35498793 PMCID: PMC9039299 DOI: 10.3389/fped.2022.828437] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Mycoplasma Pneumoniae (MP) is an important cause of community-acquired pneumonia in children, which can cause serious consequences. There has been some research into predicting Severe Mycoplasma Pneumoniae Pneumonia (SMPP) primarily focused on pre-treatment time by macrolide, pre-hospital course, CRP and LDH et.al. while seldom reporting on concoagulation status. We designed this retrospective study to compare the difference between SMPP and Non-severe MPP (NSMPP) with an attempt to find the risk factors, with a special focus on concoagulation status. METHOD We performed a retrospective study of 786 MPP patients who were hospitalized from January 1, 2016 to December 31, 2018, age ranging from 28 days to 18 years old. All patients were divided into SMPP group and NSMPP group. A univariate analysis was conducted between both groups. The factors with statistical differences were included in logistic regression analysis to summarize the predictors of SMPP. Next, the predictive value of each risk factor was calculated from the receiver operating characteristic curve (ROC curve). Patients who had D-dimer records were divided into the elevated D-dimer group (D-dimer > 308ug/L) and the control group (D-dimer ≤ 308ug/L), and the clinical manifestations were compared. RESULTS There was no significant difference in gender, age, pre-treatment time by macrolide, the white blood cell counts (WBC), Fibrinogen (FIB), Activated Partial Prothrombin Time (APTT), Prothrombin Time (PT) and Thrombin Time (TT) between SMPP and NSMPP. Compared with NSMPP, the pre-hospital course of SMPP was longer (P < 0.05), the neutrophil ratio (N%), platelet Count (PLT), C-reactive Protein (CRP), Lactate Dehydrogenase (LDH) and D-dimer were significantly higher (P < 0.01). The binary logistic regression analysis showed that the N%, PLT, CRP, LDH and D-dimer were the key predictors for SMPP, the N% > 67%, OR = 3.233, PLT > 445 × 109 /L, OR = 2.589, LDH > 354U/L, OR = 4.335 and D-dimer level > 403 ug/L, OR = 7.316. The D-dimer possessed the best predictive value. The incidence of complications such as pleural effusion, myocardial and liver damage of MPP was higher in the elevated D-dimer group than that in the control group (P < 0.05). CONCLUSION The N%, PLT, CRP, LDH and D-dimer were risk factors for SMPP. D-dimer was the best predictor among them. MPP patients with D-dimer > 308ug/L had more complications such as pleural effusion, myocardial and liver damage. More attention should be given in the treatment for this group.
Collapse
Affiliation(s)
- Juan Qiu
- Children's Hospital, Shandong University, Shandong, China.,Jinan Children's Hospital, Shandong, China.,Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Jin Ge
- Children's Hospital, Shandong University, Shandong, China
| | - Ling Cao
- Children's Hospital, Shandong University, Shandong, China
| |
Collapse
|
20
|
Vilela Rodrigues TC, Jaiswal AK, Lemes MR, da Silva MV, Sales-Campos H, Alcântara LCJ, Tosta SFDO, Kato RB, Alzahrani KJ, Barh D, Azevedo VADC, Tiwari S, Soares SDC. An immunoinformatics-based designed multi-epitope candidate vaccine (mpme-VAC/STV-1) against Mycoplasma pneumoniae. Comput Biol Med 2021; 142:105194. [PMID: 35007945 DOI: 10.1016/j.compbiomed.2021.105194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 11/18/2022]
Abstract
Pneumonia is a serious global health problem that accounts for over one million deaths annually. Among the main microorganisms causing pneumonia, Mycoplasma pneumoniae is one of the most common ones for which a vaccine is immediately required. In this context, a multi-epitope vaccine against this pathogen could be the best option that can induce effective immune response avoiding any serious adverse reactions. In this study, using an immunoinformatics approach we have designed a multi-epitope vaccine (mpme-VAC/STV-1) against M. pneumoniae. Our designed mpme-VAC/STV-1 is constructed using CTL (cytotoxic T lymphocyte), HTL (Helper T lymphocyte), and B-cell epitopes. These epitopes are selected from the core proteins of 88 M. pneumoniae genomes that were previously identified through reverse vaccinology approaches. The epitopes were filtered according to their immunogenicity, population coverage, and several other criteria. Sixteen CTL/B- and thirteen HTL/B- epitopes that belong to 5 core proteins were combined together through peptide linkers to develop the mpme-VAC/STV-1. The heat-labile enterotoxin from E. coli was used as an adjuvant. The designed mpme-VAC/STV-1 is predicted to be stable, non-toxic, non-allergenic, non-host homologous, and with required antigenic and immunogenic properties. Docking and molecular dynamic simulation of mpme-VAC/STV-1 shows that it can stimulate TLR2 pathway mediated immunogenic reactions. In silico cloning of mpme-VAC/STV-1 in an expression vector also shows positive results. Finally, the mpme-VAC/STV-1 also shows promising efficacy in immune simulation tests. Therefore, our constructed mpme-VAC/STV-1 could be a safe and effective multi-epitope vaccine for immunization against pneumonia. However, it requires further experimental and clinical validations.
Collapse
Affiliation(s)
- Thaís Cristina Vilela Rodrigues
- Programa PG Em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Arun Kumar Jaiswal
- Programa PG Em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcela Rezende Lemes
- Department of Immunology, Microbiology and Parasitology, Institute of Biological Science and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, 38025-180, MG, Brazil
| | - Marcos Vinícius da Silva
- Department of Immunology, Microbiology and Parasitology, Institute of Biological Science and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, 38025-180, MG, Brazil
| | - Helioswilton Sales-Campos
- Institute of Tropical Pathology and Public Health, Federal University of Goias (UFG), Goiânia, 74605-050, GO, Goiás, Brazil
| | | | - Sthephane Fraga de Oliveira Tosta
- Programa PG Em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo Bentes Kato
- Programa PG Em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Debmalya Barh
- Programa PG Em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal, 721172, India
| | - Vasco Ariston de Carvalho Azevedo
- Programa PG Em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sandeep Tiwari
- Programa PG Em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Siomar de Castro Soares
- Department of Immunology, Microbiology and Parasitology, Institute of Biological Science and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, 38025-180, MG, Brazil.
| |
Collapse
|
21
|
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
|
22
|
Chen T, Li Y, Sun R, Hu H, Liu Y, Herrmann M, Zhao Y, Muñoz LE. Receptor-Mediated NETosis on Neutrophils. Front Immunol 2021; 12:775267. [PMID: 34804066 PMCID: PMC8600110 DOI: 10.3389/fimmu.2021.775267] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023] Open
Abstract
Neutrophil extracellular traps (NETs), a web-like structures containing chromatin, have a significant role in assisting the capture and killing of microorganisms by neutrophils during infection. The specific engagement of cell-surface receptors by extracellular signaling molecules activates diverse intracellular signaling cascades and regulates neutrophil effector functions, including phagocytosis, reactive oxygen species release, degranulation, and NET formation. However, overproduction of NETs is closely related to the occurrence of inflammation, autoimmune disorders, non-canonical thrombosis and tumor metastasis. Therefore, it is necessary to understand neutrophil activation signals and the subsequent formation of NETs, as well as the related immune regulation. In this review, we provide an overview of the immunoreceptor-mediated regulation of NETosis. The pathways involved in the release of NETs during infection or stimulation by noninfectious substances are discussed in detail. The mechanisms by which neutrophils undergo NETosis help to refine our views on the roles of NETs in immune protection and autoimmune diseases, providing a theoretical basis for research on the immune regulation of NETs.
Collapse
Affiliation(s)
- Tao Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Sun
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Huifang Hu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
23
|
A Review of the Neutrophil Extracellular Traps (NETs) from Cow, Sheep and Goat Models. Int J Mol Sci 2021; 22:ijms22158046. [PMID: 34360812 PMCID: PMC8347029 DOI: 10.3390/ijms22158046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
This review provides insight into the importance of understanding NETosis in cows, sheep, and goats in light of the importance to their health, welfare and use as animal models. Neutrophils are essential to innate immunity, pathogen infection, and inflammatory diseases. The relevance of NETosis as a conserved innate immune response mechanism and the translational implications for public health are presented. Increased understanding of NETosis in ruminants will contribute to the prediction of pathologies and design of strategic interventions targeting NETs. This will help to control pathogens such as coronaviruses and inflammatory diseases such as mastitis that impact all mammals, including humans. Definition of unique attributes of NETosis in ruminants, in comparison to what has been observed in humans, has significant translational implications for one health and global food security, and thus warrants further study.
Collapse
|
24
|
Franzoni G, Anfossi A, De Ciucis CG, Mecocci S, Carta T, Dei Giudici S, Fruscione F, Zinellu S, Vito G, Graham SP, Oggiano A, Chessa B, Razzuoli E. Targeting Toll-Like Receptor 2: Polarization of Porcine Macrophages by a Mycoplasma-Derived Pam2cys Lipopeptide. Vaccines (Basel) 2021; 9:vaccines9070692. [PMID: 34201691 PMCID: PMC8310132 DOI: 10.3390/vaccines9070692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022] Open
Abstract
Toll-like receptor 2 (TLR2) ligands are attracting increasing attention as prophylactic and immunotherapeutic agents against pathogens and tumors. We previously observed that a synthetic diacylated lipopeptide based on a surface protein of Mycoplasma agalactiae (Mag-Pam2Cys) strongly activated innate immune cells, including porcine monocyte-derived macrophages (moMΦ). In this study, we utilized confocal microscopy, flow cytometry, multiplex cytokine ELISA, and RT-qPCR to conduct a comprehensive analysis of the effects of scalar doses of Mag-Pam2Cys on porcine moMΦ. We observed enhanced expression of activation markers (MHC class I, MHC class II DR, CD25), increased phagocytotic activity, and release of IL-12 and proinflammatory cytokines. Mag-Pam2Cys also upregulated the gene expression of several IFN-α subtypes, p65, NOS2, and molecules with antimicrobial activities (CD14, beta defensin 1). Overall, our data showed that Mag-Pam2Cys polarized porcine macrophages towards a proinflammatory antimicrobial phenotype. However, Mag-Pam2Cys downregulated the expression of IFN-α3, six TLRs (TLR3, -4, -5, -7, -8, -9), and did not interfere with macrophage polarization induced by the immunosuppressive IL-10, suggesting that the inflammatory activity evoked by Mag-Pam2Cys could be regulated to avoid potentially harmful consequences. We hope that our in vitro results will lay the foundation for the further evaluation of this diacylated lipopeptide as an immunopotentiator in vivo.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
- Correspondence: (G.F.); (B.C.)
| | - Antonio Anfossi
- School of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; (C.G.D.C.); (F.F.); (G.V.); (E.R.)
| | - Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, 06123 Perugia, Italy;
| | - Tania Carta
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
- School of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
| | - Floriana Fruscione
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; (C.G.D.C.); (F.F.); (G.V.); (E.R.)
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
| | - Guendalina Vito
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; (C.G.D.C.); (F.F.); (G.V.); (E.R.)
| | | | - Annalisa Oggiano
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (T.C.); (S.D.G.); (S.Z.); (A.O.)
| | - Bernardo Chessa
- School of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
- Correspondence: (G.F.); (B.C.)
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; (C.G.D.C.); (F.F.); (G.V.); (E.R.)
| |
Collapse
|
25
|
Comparative Phenotypic and Functional Analyses of the Effects of IL-10 or TGF-β on Porcine Macrophages. Animals (Basel) 2021; 11:ani11041098. [PMID: 33921388 PMCID: PMC8069609 DOI: 10.3390/ani11041098] [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] [Received: 03/04/2021] [Revised: 04/05/2021] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Macrophages play a central role in innate immune response to both infectious and non-infectious stressors. They respond to different agonists modifying their phenotype and functions. In humans and mice, the regulatory cytokines IL-10 or TGF-β are both known to drive macrophage polarization into an anti-inflammatory phenotype, referred to as M2c. However, the immune systems of animal species each have their own peculiarities and the M2c subsets has never been investigated in pigs. A deep knowledge of the porcine immune system is required to design vaccines or control strategies against pathogens, which are a major constraint to pork production. Due to anatomical, physiological, and immunological similarities, swine are attracting increasing attention as a model for human diseases. To better characterize porcine macrophages, we evaluated the effects of IL-10 or TGF-β on the phenotype and function of monocyte-derived macrophages. Both cytokines downregulated the expression of MHC II DR and CD14. IL-10, but not TGF-β, statistically significantly reduced the ability of macrophages to respond to Toll-like receptor 2 (TLR2) or TLR4 agonists. Whilst these data suggest differentiation to an M2c-like immunosuppressive phenotype, the responses, and differences between IL-10 and TGF-β also reveals species-specific differences. Abstract Macrophages are phagocytic cells involved in maintaining tissue homeostasis and defense against pathogens. Macrophages may be polarized into different functionally specialized subsets. M2c macrophages arise following stimulation with IL-10 or TGF-β and mediate anti-inflammatory and tissue repair functions. M2c macrophages remain poorly characterized in the pig, thus we investigated the impact of these regulatory cytokines on porcine monocyte-derived macrophages (moMΦ). The phenotype and functionality of these cells was characterized though confocal microscopy, flow cytometry, ELISA, and RT-qPCR. Both cytokines induced CD14 and MHC II DR down-regulation and reduced IL-6, TNF-α, and CD14 expression, suggestive of an anti-inflammatory phenotype. Interestingly, neither IL-10 or TGF-β were able to trigger IL-10 induction or release by moMΦ. Differences between these cytokines were observed: stimulation with IL-10, but not TGF-β, induced up-regulation of both CD16 and CD163 on moMΦ. In addition, IL-10 down-regulated expression of IL-1β and IL-12p40 4h post-stimulation and induced a stronger impairment of moMΦ ability to respond to either TLR2 or TLR4 agonists. Overall, our results provide an overview of porcine macrophage polarization by two immunosuppressive cytokines, revealing differences between IL-10 and TGF-β, and reporting some peculiarity of swine, which should be considered in translational studies.
Collapse
|
26
|
Identification of conserved Mycoplasma agalactiae surface antigens by immunoproteomics. Vet Immunol Immunopathol 2021; 236:110239. [PMID: 33845295 DOI: 10.1016/j.vetimm.2021.110239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/16/2021] [Accepted: 04/02/2021] [Indexed: 10/21/2022]
Abstract
Contagious agalactia represents one of the most relevant infectious diseases of dairy sheep, with Mycoplasma agalactiae being the primary etiological agent. The early, sensitive, and specific identification of infected animals, as well as the development of efficient prophylactic tools, remain challenging. Here, we present a comprehensive characterization of M. agalactiae antigens focusing on those shared among different isolates. Leveraging on previous proteomic data obtained on individual strains, we adopted a strategy entailing sample pooling to optimize the identification of conserved proteins that induce an immune response. The liposoluble proteins from previously characterized field isolates and the type strain PG2T were enriched by Triton X-114 fractionation, pooled, analysed by one-dimensional (1D) and two-dimensional (2D) electrophoresis, and subjected to western immunoblotting against sheep sera collected during natural infection with M. agalactiae. Immunodominant antigens were identified by Matrix-Assisted Laser Desorption-Time-Of-Flight-Mass Spectrometry (MALDI-TOF-MS). This combined immunoproteomic approach confirmed the role of several known immunogens, including P80, P48, and P40, and most variable surface proteins (Vpmas), and unveiled novel immunodominant, conserved antigens, including MAG_1000, MAG_2220, MAG_1980, phnD, MAG_4740, and MAG_2430. Genomic context, functional prediction, subcellular localization, and invariable expression of these proteins in all isolates suggest their possible involvement in bacterial pathogenicity and metabolism. Moreover, most of the identified antigens elicit a host humoral response since the early stages of infection, persisting for at least 270 days. The immunodominant, conserved antigen panel identified in this work supports the development of effective vaccines and diagnostic tools with higher sensitivity and specificity in all the natural infection stages.
Collapse
|
27
|
Optimized in vitro isolation of different subpopulation of immune cells from peripheral blood and comparative techniques for generation of monocyte-derived macrophages in small ruminants. Vet Immunol Immunopathol 2020; 230:110131. [PMID: 33129192 DOI: 10.1016/j.vetimm.2020.110131] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022]
Abstract
Peripheral blood from healthy sheep (n = 3) and goats (n = 3) were employed to establish an efficient method for simultaneous isolation of peripheral blood mononuclear cells (PBMCs) and neutrophils and to standardize protocols for monocyte purification and generation of monocyte-derived macrophages (MDMs). In both species, a significantly enriched population of PBMCs, with higher purity and number of cells determined by flow cytometry, was achieved when processing through a density gradient a mixture of buffy-coat and red blood cell layer (RBC) in comparison to the use of just the buffy-coat (p < 0.05). Neutrophils could be subsequently isolated from the layer, located underneath PBMCs fraction with significant higher purity rates, higher than 85 % determined by flow cytometry, than those obtained with protocols without density gradients (< 60 %) (p < 0.05). This technique would allow the isolation of both cell populations from the same sample of blood. A pure cell population of monocytes, CD14+ cells, was purified from PBMCs when using immunomagnetic columns, which allow for 17 % (nº monocytes/nº PBMCs) of yield and high percentages of expression of CD14+ (88 %), MHC-II+ (91.5 %) and CD11b+ (94 %) established by flow cytometry. On the other hand, the classical and non-expensive purification of monocytes from PBMCs based on the adherence capacity of the former, allowed significantly lower yield of monocytes (4.6 %), with percentages of surface markers expression that dropped to 35 %, 65 % and 55 %, respectively (p < 0.001), suggesting the isolation of a mixed population of cells. The addition of GM-CSF to the culture, at concentration from 25 to 125 ng/mL, enhanced proportionally the number of MDMs generated compared to the absence of supplementation or the use of autologous serum from 5% to 20 %. However, purification of monocytes through the adherence method achieved higher yields of MDMs than those isolated through immunomagnetic columns in both species (p < 0.001). Under the conditions of this study, the use of centrifugation in density gradients allow for the simultaneous purification of PBMCs and neutrophils, with high purity of both populations, from the same sample of blood. The isolation of monocytes could be subsequently achieved through two different methods, i.e. based on immunomagnetic columns or adherence. The preference between both methods would depend on the necessities of the experiment, the initial sample with high purity of monocytes or a final population of MDMs required.
Collapse
|
28
|
Kamminga T, Benis N, Martins Dos Santos V, Bijlsma JJE, Schaap PJ. Combined Transcriptome Sequencing of Mycoplasma hyopneumoniae and Infected Pig Lung Tissue Reveals Up-Regulation of Bacterial F1-Like ATPase and Down-Regulation of the P102 Cilium Adhesin in vivo. Front Microbiol 2020; 11:1679. [PMID: 32765473 PMCID: PMC7379848 DOI: 10.3389/fmicb.2020.01679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/26/2020] [Indexed: 12/21/2022] Open
Abstract
Mycoplasma hyopneumoniae (M. hyopneumoniae) causes enzootic pneumonia in pigs but it is still largely unknown which host-pathogen interactions enable persistent infection and cause disease. In this study, we analyzed the host and bacterial transcriptomes during infection using RNA sequencing. Comparison of the transcriptome of lung lesion tissue from infected pigs with lung tissue from non-infected animals, identified 424 differentially expressed genes (FDR < 0.01 and fold change > 1.5LOG2). These genes were part of the following major pathways of the immune system: interleukin signaling (type 4, 10, 13, and 18), regulation of Toll-like receptors by endogenous ligand and activation of C3 and C5 in the complement system. Besides analyzing the lung transcriptome, a sampling protocol was developed to obtain enough bacterial mRNA from infected lung tissue for RNA sequencing. This was done by flushing infected lobes in the lung, and subsequently enriching for bacterial RNA. On average, 2.2 million bacterial reads were obtained per biological replicate to analyze the bacterial in vivo transcriptome. We compared the in vivo bacterial transcriptome with the transcriptome of bacteria grown in vitro and identified 22 up-regulated and 30 down-regulated genes (FDR < 0.01 and fold change > 2LOG2). Six out of seven genes in the operon encoding the mycoplasma specific F1-like ATPase (MHP_RS02445-MHP_RS02475) and all genes in the operon MHP_RS01965-MHP_RS01990 with functions related to nucleotide metabolism, spermidine transport and glycerol-3-phoshate transport were up-regulated in vivo. Down-regulated in vivo were genes related to glycerol uptake, cilium adhesion (P102), cell division and myo-inositol metabolism. In addition to providing a novel method to isolate bacterial mRNA from infected lung, this study provided insights into changes in gene expression during infection, which could help development of novel treatment strategies against enzootic pneumonia caused by M. hyopneumoniae.
Collapse
Affiliation(s)
- Tjerko Kamminga
- Laboratory of Systems and Synthetic Biology, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands.,Bioprocess Technology and Support, MSD Animal Health, Boxmeer, Netherlands
| | - Nirupama Benis
- Laboratory of Systems and Synthetic Biology, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| | - Vitor Martins Dos Santos
- Laboratory of Systems and Synthetic Biology, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| | | | - Peter J Schaap
- Laboratory of Systems and Synthetic Biology, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
29
|
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
|
30
|
Comparison of Macrophage Responses to African Swine Fever Viruses Reveals that the NH/P68 Strain is Associated with Enhanced Sensitivity to Type I IFN and Cytokine Responses from Classically Activated Macrophages. Pathogens 2020; 9:pathogens9030209. [PMID: 32178332 PMCID: PMC7157553 DOI: 10.3390/pathogens9030209] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
African swine fever (ASF) poses a severe threat to the global pig industry for which currently there is no available vaccine. The aetiological ASF virus (ASFV) has a predilection for cells of the myeloid lineage, however little is known about its interaction with polarised macrophages. This study focused on the in vitro interactions of porcine monocyte-derived un-activated (moMΦ), classically (moM1), alternatively (moM2), and IFN-α-activated macrophages with two genotype I ASFV strains: virulent 22653/14 and attenuated NH/P68. At a high multiplicity of infection, NH/P68, but not 22653/14, presented a reduced ability to infect moM1 and IFN−α-activated moMΦ compared to moMΦ. IFN-α activation resulted in a dose-dependent reduction in the proportion of ASFV-infected cells. Both strains replicated efficiently in all the subsets. While higher levels of IL-1α, IL-1β, and IL-18 were secreted by NH/P68-infected moM1 compared to 22653/14, both strains negatively affected moMΦ ability to release IL-6, IL-12, TNF-α in response to classical activation or stimulation with a TLR2 agonist. Our results suggest that ASFV 22653/14 covertly replicates in macrophages, compromising the development of effective immune responses. Attenuated NH/P68 has partially lost these mechanisms, which may enhance immune surveillance. A better understating of these mechanisms should aid the rational design of live attenuated ASFV vaccines.
Collapse
|
31
|
|
32
|
Gaurivaud P, Ganter S, Villard A, Manso-Silvan L, Chevret D, Boulé C, Monnet V, Tardy F. Mycoplasmas are no exception to extracellular vesicles release: Revisiting old concepts. PLoS One 2018; 13:e0208160. [PMID: 30485365 PMCID: PMC6261642 DOI: 10.1371/journal.pone.0208160] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022] Open
Abstract
Release of extracellular vesicles (EV) by Gram-negative and positive bacteria is being frequently reported. EV are nano-sized, membrane-derived, non-self-replicating, spherical structures shed into the extracellular environment that could play a role in bacteria-host interactions. Evidence of EV production in bacteria belonging to the class Mollicutes, which are wall-less, is mainly restricted to the genus Acholeplasma and is scanty for the Mycoplasma genus that comprises major human and animal pathogens. Here EV release by six Mycoplasma (sub)species of clinical importance was investigated. EV were obtained under nutritional stress conditions, purified by ultracentrifugation and observed by electron microscopy. The membrane proteins of EV from three different species were further identified by mass spectrometry as a preliminary approach to determining their potential role in host-pathogen interactions. EV were shown to be released by all six (sub)species although their quantities and sizes (30-220 nm) were very variable. EV purification was complicated by the minute size of viable mycoplasmal cells. The proteins of EV-membranes from three (sub)species included major components of host-pathogen interactions, suggesting that EV could contribute to make the host-pathogen interplay more complex. The process behind EV release has yet to be deciphered, although several observations demonstrated their active release from the plasma membrane of living cells. This work shed new light on old concepts of "elementary bodies" and "not-cell bound antigens".
Collapse
Affiliation(s)
- Patrice Gaurivaud
- Université de Lyon, Anses, Laboratoire de Lyon, UMR Mycoplasmoses des Ruminants, Lyon, France
- Université de Lyon, VetAgro Sup, UMR Mycoplasmoses des Ruminants, Marcy-L’étoile, France
- * E-mail:
| | - Sarah Ganter
- Université de Lyon, Anses, Laboratoire de Lyon, UMR Mycoplasmoses des Ruminants, Lyon, France
- Université de Lyon, VetAgro Sup, UMR Mycoplasmoses des Ruminants, Marcy-L’étoile, France
| | - Alexandre Villard
- Université de Lyon, Anses, Laboratoire de Lyon, UMR Mycoplasmoses des Ruminants, Lyon, France
- Université de Lyon, VetAgro Sup, UMR Mycoplasmoses des Ruminants, Marcy-L’étoile, France
| | - Lucia Manso-Silvan
- CIRAD, UMR ASTRE, Montpellier, France
- INRA, UMR ASTRE, Montpellier, France
| | - Didier Chevret
- PAPPSO, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Christelle Boulé
- Université Claude Bernard Lyon 1, Centre Technologique des Microstructures, Service « Etudes à façon » EZUS Lyon, Villeurbanne, France
| | - Véronique Monnet
- PAPPSO, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Florence Tardy
- Université de Lyon, Anses, Laboratoire de Lyon, UMR Mycoplasmoses des Ruminants, Lyon, France
- Université de Lyon, VetAgro Sup, UMR Mycoplasmoses des Ruminants, Marcy-L’étoile, France
| |
Collapse
|
33
|
Christodoulides A, Gupta N, Yacoubian V, Maithel N, Parker J, Kelesidis T. The Role of Lipoproteins in Mycoplasma-Mediated Immunomodulation. Front Microbiol 2018; 9:1682. [PMID: 30108558 PMCID: PMC6080569 DOI: 10.3389/fmicb.2018.01682] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 07/05/2018] [Indexed: 01/16/2023] Open
Abstract
Mycoplasma infections, such as walking pneumonia or pelvic inflammatory diseases, are a major threat to public health. Despite their relatively small physical and genomic size, mycoplasmas are known to elicit strong host immune responses, generally inflammatory, while also being able to evade the immune system. The mycoplasma membrane is composed of approximately two-thirds protein and one-third lipid and contains several lipoproteins that are known to regulate host immune responses. Herein, the immunomodulatory effects of mycoplasma lipoproteins are reviewed. A better understanding of the immunomodulatory effects, both activating and evasive, of Mycoplasma surface lipoproteins will contribute to understanding mechanisms potentially relevant to mycoplasma disease vaccine development and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Theodoros Kelesidis
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
34
|
Mitiku F, Hartley CA, Sansom FM, Coombe JE, Mansell PD, Beggs DS, Browning GF. The major membrane nuclease MnuA degrades neutrophil extracellular traps induced by Mycoplasma bovis. Vet Microbiol 2018; 218:13-19. [DOI: 10.1016/j.vetmic.2018.03.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 12/19/2022]
|
35
|
Dang G, Cui Y, Wang L, Li T, Cui Z, Song N, Chen L, Pang H, Liu S. Extracellular Sphingomyelinase Rv0888 of Mycobacterium tuberculosis Contributes to Pathological Lung Injury of Mycobacterium smegmatis in Mice via Inducing Formation of Neutrophil Extracellular Traps. Front Immunol 2018; 9:677. [PMID: 29670633 PMCID: PMC5893642 DOI: 10.3389/fimmu.2018.00677] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/19/2018] [Indexed: 12/16/2022] Open
Abstract
Mycobacterium tuberculosis is the causative agent of tuberculosis (TB), which mainly causes pulmonary injury and tubercles. Although macrophages are generally considered to harbor the main cells of M. tuberculosis, new evidence suggests that neutrophils are rapidly recruited to the infected lung. M. tuberculosis itself, or its early secreted antigenic target protein 6 (ESAT-6), can induce formation of neutrophil extracellular traps (NETs). However, NETs trap mycobacteria but are unable to kill them. The role of NETs’ formation in the pathogenesis of mycobacteria remains unclear. Here, we report a new M. tuberculosis extracellular factor, bifunctional enzyme Rv0888, with both nuclease and sphingomyelinase activities. Rv0888 sphingomyelinase activity can induce NETs’ formation in vitro and in the lung of the mice and enhance the colonization ability of Mycobacterium smegmatis in the lungs of mice. Mice infected by M. smegmatis harboring Rv0888 sphingomyelinase induced pathological injury and inflammation of the lung, which was mainly mediated by NETs, induced by Rv0888 sphingomyelinase, associated protein (myeloperoxidase) triggered caspase-3. In summary, the study sheds new light on the pathogenesis of mycobacteria and reveals a novel target for TB treatment.
Collapse
Affiliation(s)
- Guanghui Dang
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yingying Cui
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Lei Wang
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tiantian Li
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ziyin Cui
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ningning Song
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Liping Chen
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hai Pang
- School of Medicine, Tsinghua University, Beijing, China
| | - Siguo Liu
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
36
|
Gomez-Lopez N, Romero R, Xu Y, Miller D, Unkel R, Shaman M, Jacques SM, Panaitescu B, Garcia-Flores V, Hassan SS. Neutrophil Extracellular Traps in the Amniotic Cavity of Women with Intra-Amniotic Infection: A New Mechanism of Host Defense. Reprod Sci 2017; 24:1139-1153. [PMID: 27884950 PMCID: PMC6343453 DOI: 10.1177/1933719116678690] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) control microbial infections through their antimicrobial activities attributed to DNA, histones, granules, and cytoplasmic proteins (eg, elastase). Intra-amniotic infection is characterized by the influx of neutrophils into the amniotic cavity; therefore, the aim of this study was to determine whether amniotic fluid neutrophils form NETs in this inflammatory process. METHODS Amniotic fluid samples from women with intra-amniotic infection (n = 15) were stained for bacteria detection using fluorescent dyes. Amniotic fluid neutrophils were purified by filtration. As controls, neutrophils from maternal blood samples (n = 3) were isolated by density gradients. Isolated neutrophils were plated onto glass cover slips for culture with and without 100 nM of phorbol-12-myristate-13-acetate (PMA). NET formation was assessed by 4',6-diamidino-2-phenylindole (DAPI) staining and scanning electron microscopy. Different stages of NET formation were visualized using antibodies against elastase and histone H3, in combination with DAPI staining, by confocal microscopy. Finally, maternal or neonatal neutrophils were added to amniotic fluid samples from women without intra-amniotic infection (n = 4), and NET formation was evaluated by DAPI staining. RESULTS (1) NETs were present in the amniotic fluid of women with intra-amniotic infection; (2) all of the amniotic fluid samples had detectable live and dead bacteria associated with the presence of NETs; (3) in contrast to neutrophils from the maternal circulation, amniotic fluid neutrophils did not require PMA stimulation to form NETs; (4) different stages of NET formation were observed by co-localizing elastase, histone H3, and DNA in amniotic fluid neutrophils; and (5) neither maternal nor neonatal neutrophils form NETs in the amniotic fluid of women without intra-amniotic infection. CONCLUSION NETs are detectable in the amniotic fluid of women with intra-amniotic infection.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
- Department of Immunology and Microbiology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor,
MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University,
East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit,
MI, USA
| | - Yi Xu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Derek Miller
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
- Department of Immunology and Microbiology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Ronald Unkel
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Majid Shaman
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Suzanne M. Jacques
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Pathology, Hutzel Women’s Hospital/Harper University Hospital,
Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| |
Collapse
|
37
|
Cubeddu T, Cacciotto C, Pisanu S, Tedde V, Alberti A, Pittau M, Dore S, Cannas A, Uzzau S, Rocca S, Addis MF. Cathelicidin production and release by mammary epithelial cells during infectious mastitis. Vet Immunol Immunopathol 2017; 189:66-70. [PMID: 28669389 DOI: 10.1016/j.vetimm.2017.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/25/2017] [Accepted: 06/13/2017] [Indexed: 10/19/2022]
Abstract
Cathelicidins are well-characterized antimicrobial peptides (AMPs) that are present in significant amounts in mastitic milk. Neutrophils are believed to be the main producers of these AMPs, while the role of mammary epithelial cells (MECs) in their production and release is still unclear. In this work, cathelicidin production patterns were investigated in mammary tissues of ewes infected by Staphylococcus aureus, Streptococcus uberis, or Mycoplasma agalactiae, with a combined approach including immunohistochemistry, immune-colocalization, and fluorescent in situ hybridization. Our results confirm that MECs produce and release cathelicidins in response to different mastitis pathogens. As opposed to neutrophils, however, MECs do not seem to store the preformed protein precursor in their cytoplasm, but appear to synthesize and release it only upon exposure to the microorganisms. Cathelicidin production by MECs appears to occur before leukocyte influx in the milk, suggesting a role for these cells in the initial response of the mammary epithelium to microbial infection. Once in the milk, infiltrating neutrophils release massive amounts of cathelicidin by degranulation and production of neutrophil extracellular traps, acting as the main contributor for cathelicidin abundance in mastitic milk. Taken together, our results support the active contribution of MECs to cathelicidin production and release, and reinforce the value of cathelicidins as sensitive and pathogen-independent mastitis markers.
Collapse
Affiliation(s)
- Tiziana Cubeddu
- Università degli Studi di Sassari, Dipartimento di Medicina Veterinaria, Via Vienna 2, 07100 Sassari, Italy
| | - Carla Cacciotto
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy
| | - Salvatore Pisanu
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy
| | - Vittorio Tedde
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy
| | - Alberto Alberti
- Università degli Studi di Sassari, Dipartimento di Medicina Veterinaria, Via Vienna 2, 07100 Sassari, Italy
| | - Marco Pittau
- Università degli Studi di Sassari, Dipartimento di Medicina Veterinaria, Via Vienna 2, 07100 Sassari, Italy
| | - Simone Dore
- C.Re.N.M.O.C., Centro di Referenza Nazionale per le Mastopatie degli Ovini e dei Caprini - Istituto Zooprofilattico Sperimentale della Sardegna, Via Duca degli Abruzzi 8, 07100 Sassari, Italy
| | - Agnese Cannas
- C.Re.N.M.O.C., Centro di Referenza Nazionale per le Mastopatie degli Ovini e dei Caprini - Istituto Zooprofilattico Sperimentale della Sardegna, Via Duca degli Abruzzi 8, 07100 Sassari, Italy
| | - Sergio Uzzau
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy; Università degli Studi di Sassari, Dipartimento di Scienze Biomediche, Viale S. Pietro 43/B, 07100 Sassari, Italy
| | - Stefano Rocca
- Università degli Studi di Sassari, Dipartimento di Medicina Veterinaria, Via Vienna 2, 07100 Sassari, Italy
| | - Maria Filippa Addis
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy; Università degli Studi di Milano, Dipartimento di Medicina Veterinaria, Via Celoria 10, 20133 Milano, Italy.
| |
Collapse
|
38
|
Hoppenbrouwers T, Autar ASA, Sultan AR, Abraham TE, van Cappellen WA, Houtsmuller AB, van Wamel WJB, van Beusekom HMM, van Neck JW, de Maat MPM. In vitro induction of NETosis: Comprehensive live imaging comparison and systematic review. PLoS One 2017; 12:e0176472. [PMID: 28486563 PMCID: PMC5423591 DOI: 10.1371/journal.pone.0176472] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/11/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Multiple inducers of in vitro Neutrophil Extracellular Trap (NET) formation (NETosis) have been described. Since there is much variation in study design and results, our aim was to create a systematic review of NETosis inducers and perform a standardized in vitro study of NETosis inducers important in (cardiac) wound healing. METHODS In vitro NETosis was studied by incubating neutrophils with PMA, living and dead bacteria (S. aureus and E. coli), LPS, (activated) platelets (supernatant), glucose and calcium ionophore Ionomycin using 3-hour periods of time-lapse confocal imaging. RESULTS PMA is a consistent and potent inducer of NETosis. Ionomycin also consistently resulted in extrusion of DNA, albeit with a process that differs from the NETosis process induced by PMA. In our standardized experiments, living bacteria were also potent inducers of NETosis, but dead bacteria, LPS, (activated) platelets (supernatant) and glucose did not induce NETosis. CONCLUSION Our systematic review confirms that there is much variation in study design and results of NETosis induction. Our experimental results confirm that under standardized conditions, PMA, living bacteria and Ionomycin all strongly induce NETosis, but real-time confocal imaging reveal different courses of events.
Collapse
Affiliation(s)
- Tamara Hoppenbrouwers
- Department of Plastic and Reconstructive Surgery, Erasmus MC, Rotterdam, The Netherlands
- Department of Hematology, Erasmus MC, Rotterdam, The Netherlands
| | - Anouchska S. A. Autar
- Department of Hematology, Erasmus MC, Rotterdam, The Netherlands
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Andi R. Sultan
- Department of Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - Tsion E. Abraham
- Optical Imaging Center, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Willem J. B. van Wamel
- Department of Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | | | - Johan W. van Neck
- Department of Plastic and Reconstructive Surgery, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
39
|
Addis M, Bronzo V, Puggioni G, Cacciotto C, Tedde V, Pagnozzi D, Locatelli C, Casula A, Curone G, Uzzau S, Moroni P. Relationship between milk cathelicidin abundance and microbiologic culture in clinical mastitis. J Dairy Sci 2017; 100:2944-2953. [DOI: 10.3168/jds.2016-12110] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/24/2016] [Indexed: 11/19/2022]
|
40
|
Chopra-Dewasthaly R, Korb M, Brunthaler R, Ertl R. Comprehensive RNA-Seq Profiling to Evaluate the Sheep Mammary Gland Transcriptome in Response to Experimental Mycoplasma agalactiae Infection. PLoS One 2017; 12:e0170015. [PMID: 28081235 PMCID: PMC5231372 DOI: 10.1371/journal.pone.0170015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/26/2016] [Indexed: 11/19/2022] Open
Abstract
Mycoplasma agalactiae is a worldwide serious pathogen of small ruminants that usually spreads through the mammary route causing acute to subacute mastitis progressing to chronic persistent disease that is hard to eradicate. Knowledge of mechanisms of its pathogenesis and persistence in the mammary gland are still insufficient, especially the host-pathogen interplay that enables it to reside in a chronic subclinical state. This study reports transcriptome profiling of mammary tissue from udders of sheep experimentally infected with M. agalactiae type strain PG2 in comparison with uninfected control animals using Illumina RNA-sequencing (RNA-Seq). Several differentially expressed genes (DEGs) were observed in the infected udders and RT-qPCR analyses of selected DEGs showed their expression profiles to be in agreement with results from RNA-Seq. Gene Ontology (GO) analysis revealed majority of the DEGs to be associated with mycoplasma defense responses that are directly or indirectly involved in host innate and adaptive immune responses. Similar RNA-Seq analyses were also performed with spleen cells of the same sheep to know the specific systemic transcriptome responses. Spleen cells exhibited a comparatively lower number of DEGs suggesting a less prominent host response in this organ. To our knowledge this is the first study that describes host transcriptomics of M. agalactiae infection and the related immune-inflammatory responses. The data provides useful information to further dissect the molecular genetic mechanisms underlying mycoplasma mastitis, which is a prerequisite for designing effective intervention strategies.
Collapse
Affiliation(s)
- Rohini Chopra-Dewasthaly
- Division of Clinical Microbiology and Infection Biology, Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Veterinaerplatz 1,Vienna, Austria
- * E-mail:
| | - Melanie Korb
- VetCore Facility for Research, University of Veterinary Medicine, Veterinaerplatz 1,Vienna, Austria
| | - René Brunthaler
- Institute of Pathology and Forensic Veterinary Medicine, Department of Pathobiology, University of Veterinary Medicine, Veterinaerplatz 1, Vienna, Austria
| | - Reinhard Ertl
- VetCore Facility for Research, University of Veterinary Medicine, Veterinaerplatz 1,Vienna, Austria
| |
Collapse
|