1
|
Zhao T, Gussak A, van der Hee B, Brugman S, van Baarlen P, Wells JM. Identification of plasminogen-binding sites in Streptococcus suis enolase that contribute to bacterial translocation across the blood-brain barrier. Front Cell Infect Microbiol 2024; 14:1356628. [PMID: 38456079 PMCID: PMC10919400 DOI: 10.3389/fcimb.2024.1356628] [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: 12/15/2023] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
Streptococcus suis is an emerging zoonotic pathogen that can cause invasive disease commonly associated with meningitis in pigs and humans. To cause meningitis, S. suis must cross the blood-brain barrier (BBB) comprising blood vessels that vascularize the central nervous system (CNS). The BBB is highly selective due to interactions with other cell types in the brain and the composition of the extracellular matrix (ECM). Purified streptococcal surface enolase, an essential enzyme participating in glycolysis, can bind human plasminogen (Plg) and plasmin (Pln). Plg has been proposed to increase bacterial traversal across the BBB via conversion to Pln, a protease which cleaves host proteins in the ECM and monocyte chemoattractant protein 1 (MCP1) to disrupt tight junctions. The essentiality of enolase has made it challenging to unequivocally demonstrate its role in binding Plg/Pln on the bacterial surface and confirm its predicted role in facilitating translocation of the BBB. Here, we report on the CRISPR/Cas9 engineering of S. suis enolase mutants eno261, eno252/253/255, eno252/261, and eno434/435 possessing amino acid substitutions at in silico predicted binding sites for Plg. As expected, amino acid substitutions in the predicted Plg binding sites reduced Plg and Pln binding to S. suis but did not affect bacterial growth in vitro compared to the wild-type strain. The binding of Plg to wild-type S. suis enhanced translocation across the human cerebral microvascular endothelial cell line hCMEC/D3 but not for the eno mutant strains tested. To our knowledge, this is the first study where predicted Plg-binding sites of enolase have been mutated to show altered Plg and Pln binding to the surface of S. suis and attenuation of translocation across an endothelial cell monolayer in vitro.
Collapse
Affiliation(s)
| | | | | | | | | | - Jerry M. Wells
- Host-Microbe Interactomics, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
2
|
Kumar A, Boradia VM, Mahajan A, Kumaran S, Raje M, Raje CI. Mycobacterium tuberculosis H37Rv enolase (Rv1023)- expression, characterization and effect of host dependent modifications on protein functionality. Biochimie 2023; 214:102-113. [PMID: 37385399 DOI: 10.1016/j.biochi.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/01/2023]
Abstract
Mycobacterium tuberculosis enolase is an essential glycolytic enzyme that catalyzes the conversion of 2, phosphoglycerate (PGA) to phosphoenol pyruvate (PEP). It is also a crucial link between glycolysis and the tricarboxylic acid (TCA) pathway. The depletion of PEP has recently been associated with the emergence of non-replicating drug resistant bacteria. Enolase is also known to exhibit multiple alternate functions, such as promoting tissue invasion via its role as a plasminogen (Plg) receptor. In addition, proteomic studies have identified the presence of enolase in the Mtb degradosome and in biofilms. However, the precise role in these processes has not been elaborated. The enzyme was recently identified as a target for 2-amino thiazoles - a novel class of anti-mycobacterials. In vitro assays and characterization of this enzyme were unsuccessful due to the inability to obtain functional recombinant protein. In the present study, we report the expression and characterization of enolase using Mtb H37Ra as a host strain. Our study demonstrates that the enzyme activity and alternate functions of this protein are significantly impacted by the choice of expression host (Mtb H37Ra or E. coli). Detailed analysis of the protein from each source revealed subtle differences in the post-translational modifications. Lastly, our study confirms the role of enolase in Mtb biofilm formation and describes the potential for inhibiting this process.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Biotechnology National Institute of Pharmaceutical Education and Research (NIPER), Phase X, Sector 67, SAS Nagar, Punjab, 160062, India
| | - Vishant Mahendra Boradia
- Department of Biotechnology National Institute of Pharmaceutical Education and Research (NIPER), Phase X, Sector 67, SAS Nagar, Punjab, 160062, India
| | - Apurwa Mahajan
- Council of Scientific and Industrial Research -Institute of Microbial Technology (CSIR-IMTECH), Sector 39 A, Chandigarh, 160036, India
| | - S Kumaran
- Council of Scientific and Industrial Research -Institute of Microbial Technology (CSIR-IMTECH), Sector 39 A, Chandigarh, 160036, India
| | - Manoj Raje
- Council of Scientific and Industrial Research -Institute of Microbial Technology (CSIR-IMTECH), Sector 39 A, Chandigarh, 160036, India
| | - Chaaya Iyengar Raje
- Department of Biotechnology National Institute of Pharmaceutical Education and Research (NIPER), Phase X, Sector 67, SAS Nagar, Punjab, 160062, India.
| |
Collapse
|
3
|
Xie Q, Xing H, Wen X, Liu B, Wei Y, Yu Y, Xie X, Song D, Shao G, Xiong Q, Feng Z. Identification of the multiple roles of enolase as an plasminogen receptor and adhesin in Mycoplasma hyopneumoniae. Microb Pathog 2023; 174:105934. [PMID: 36481292 DOI: 10.1016/j.micpath.2022.105934] [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: 08/01/2022] [Revised: 11/07/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Mycoplasma hyopneumoniae is the etiological agent underlying porcine enzootic pneumonia, a chronic respiratory disease worldwide. The recruitment of plasminogen to the surface and subsequently promotion of plasmin conversion by the surface-located receptor, have been reported to assist the adhesion and invasion of Mycoplasmas. The surface localization and plasminogen-binding ability of M. hyopneumoniae enolase were previously confirmed; however, the biological functions were not be determined, especially the role as a plasminogen receptor. Here, using ELISA and SPR analyses, we confirmed the stable binding of M. hyopneumoniae enolase to plasminogen in a dose-dependent manner. The facilitation of the activation of plasminogen in the presence of tPA and direct activation of plasminogen at low efficiency without tPA addition by M. hyopneumoniae enolase were also determined using a plasmin-specific chromogenic substrate. Notably, the C-terminal and N-terminal regions located in M. hyopneumoniae enolase play an important role in plasminogen binding and activation. Additionally, we demonstrate that M. hyopneumoniae enolase can competitively inhibit the adherence of M. hyopneumoniae to PK15 cells. These results provide insight into the role of enolase in M. hyopneumoniae infection, a mechanism that manipulates the proteolytic system of the host.
Collapse
Affiliation(s)
- Qingyun Xie
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Huixuan Xing
- Institute of Animal Science, Tibet Agricultural and Animal Husbandry College, Linzhi, 860000, China
| | - Xiaoyun Wen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Beibei Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Yanna Wei
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Yanfei Yu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Xing Xie
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Daesub Song
- College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Guoqing Shao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Qiyan Xiong
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China.
| | - Zhixin Feng
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China.
| |
Collapse
|
4
|
Hu Z, Li H, Zhao Y, Wang G, Shang Y, Chen Y, Wang S, Tian M, Qi J, Yu S. NADH oxidase of Mycoplasma synoviae is a potential diagnostic antigen, plasminogen/fibronectin binding protein and a putative adhesin. BMC Vet Res 2022; 18:455. [PMID: 36581820 PMCID: PMC9798693 DOI: 10.1186/s12917-022-03556-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Mycoplasma synoviae (MS) is an important pathogen causing respiratory diseases and arthritis in chickens and turkeys, thus, resulting in serious economic losses to the poultry industry. Membrane-associated proteins are thought to play important roles in cytoadherence and pathogenesis. NADH oxidase (NOX) is an oxidoreductase involved in glycolysis, which is thought to be a multifunctional protein and potential virulence factor in some pathogens. However, little is known regarding the NOX of MS (MSNOX). We previously demonstrated that MSNOX was a metabolic enzyme distributed in not only the cytoplasm but also the MS membrane. This study was aimed at exploring NOX's potential as a diagnostic antigen and its role in MS cytoadherence. RESULTS Western blots and ELISAs indicated that recombinant MSNOX (rMSNOX) protein reacted with sera positive for various MS isolates, but not MG isolates or other avian pathogens, thus, suggesting that rMSNOX is a potential diagnostic antigen. In addition, rabbit anti-rMSNOX serum showed substantial complement-dependent mycoplasmacidal activity toward various MS isolates and MG Rlow. MSNOX protein was found not only in the cytoplasm but also on the membrane of MS through suspension immunofluorescence and immunogold electron microscopy assays. Indirect immunofluorescence assays indicated that rMSNOX adhered to DF-1 cells, and this adherence was inhibited by rabbit anti-rMSNOX, but not anti-MG serum. Furthermore, indirect immunofluorescence and colony counting assays confirmed that the rabbit anti-rMSNOX serum inhibited the adherence of various MS isolates but not MG Rlow to DF-1 cells. Moreover, plasminogen (Plg)- and fibronectin (Fn)-binding assays demonstrated that rMSNOX bound Plg and Fn in a dose-dependent manner, thereby further confirming that MSNOX may be a putative adhesin. CONCLUSION MSNOX was identified to be a surface immunogenic protein that has good immunoreactivity and specificity in Western blot and ELISA, and therefore, may be used as a potential diagnostic antigen in the future. In addition, rMSNOX adhered to DF-1 cells, an effect inhibited by rabbit anti-rMSNOX, but not anti-MG serum, and anti-rMSNOX serum inhibited the adherence of various MS isolates, but not MG Rlow, to DF-1 cells, thus indicating that the inhibition of adherence by anti-MSNOX serum was MS specific. Moreover, rMSNOX adhered to extracellular matrix proteins including Plg and Fn, thus suggesting that NOX may play important roles in MS cytoadherence and pathogenesis. Besides, rabbit anti-rMSNOX serum presented complement-dependent mycoplasmacidal activity toward both MS and MG, indicating the MSNOX may be further studied as a potential protective vaccine candidate.
Collapse
Affiliation(s)
- Zengjin Hu
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China ,grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiangxilu, Hefei, Anhui 230061 People’s Republic of China
| | - Haoran Li
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China ,grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiangxilu, Hefei, Anhui 230061 People’s Republic of China
| | - Yuxin Zhao
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China ,grid.268415.cCollege of Veterinary Medicine, Yangzhou University, No. 88 University South Road, Yangzhou, Jiangsu 225009 People’s Republic of China
| | - Guijun Wang
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiangxilu, Hefei, Anhui 230061 People’s Republic of China
| | - Yuanbing Shang
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Yuetong Chen
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Shaohui Wang
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Mingxing Tian
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Jingjing Qi
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| | - Shengqing Yu
- grid.464410.30000 0004 1758 7573Shanghai Veterinary Research Institute, the Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai, 200241 People’s Republic of China
| |
Collapse
|
5
|
Muacevic A, Adler JR, Toor D, Lyngdoh V, Nongrum G, Kapoor M, Chakraborti A. Group A Streptococcus Infections: Their Mechanisms, Epidemiology, and Current Scope of Vaccines. Cureus 2022; 14:e33146. [PMID: 36721580 PMCID: PMC9884514 DOI: 10.7759/cureus.33146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2022] [Indexed: 01/01/2023] Open
Abstract
Group A streptococci (GAS) are gram-positive, cocci-shaped bacteria that cause a wide variety of infections and are a cause of significant health burden, particularly in lower- and middle-income nations. The GAS genome contains a number of virulence factors such as the M-protein, hyaluronic acid, C5a peptidase, etc. Despite its significant health burden across the globe, a proper vaccine against GAS infections is not yet available. Various candidates for an effective GAS vaccine are currently being researched. These are based on various parts of the streptococcal genome. These include candidates based on the N-terminal region of the M protein, the conserved C-terminal region of the M protein, and other parts of the streptococcal genome. The development of a vaccine against GAS infections is hampered by certain challenges, such as extensive genetic heterogeneity and high protein sequence variation. This review paper sheds light on the various virulence factors of GAS, their epidemiology, the different vaccine candidates currently being researched, and the challenges associated with M-protein and non-M-protein-based vaccines. This review also sheds light on the current scenario regarding the status of vaccine development against GAS-related infections.
Collapse
|
6
|
The Contribution of Viral Proteins to the Synergy of Influenza and Bacterial Co-Infection. Viruses 2022; 14:v14051064. [PMID: 35632805 PMCID: PMC9143653 DOI: 10.3390/v14051064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
A severe course of acute respiratory disease caused by influenza A virus (IAV) infection is often linked with subsequent bacterial superinfection, which is difficult to cure. Thus, synergistic influenza-bacterial co-infection represents a serious medical problem. The pathogenic changes in the infected host are accelerated as a consequence of IAV infection, reflecting its impact on the host immune response. IAV infection triggers a complex process linked with the blocking of innate and adaptive immune mechanisms required for effective antiviral defense. Such disbalance of the immune system allows for easier initiation of bacterial superinfection. Therefore, many new studies have emerged that aim to explain why viral-bacterial co-infection can lead to severe respiratory disease with possible fatal outcomes. In this review, we discuss the key role of several IAV proteins-namely, PB1-F2, hemagglutinin (HA), neuraminidase (NA), and NS1-known to play a role in modulating the immune defense of the host, which consequently escalates the development of secondary bacterial infection, most often caused by Streptococcus pneumoniae. Understanding the mechanisms leading to pathological disorders caused by bacterial superinfection after the previous viral infection is important for the development of more effective means of prevention; for example, by vaccination or through therapy using antiviral drugs targeted at critical viral proteins.
Collapse
|
7
|
The C-Terminal Domain of Staphylococcus aureus Zinc Transport Protein AdcA Binds Plasminogen and Factor H In Vitro. Pathogens 2022; 11:pathogens11020240. [PMID: 35215183 PMCID: PMC8878332 DOI: 10.3390/pathogens11020240] [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: 12/22/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Bacterial acquisition of metals from a host is an essential attribute to facilitate survival and colonization within an infected organism. Staphylococcus aureus, a bacterial pathogen of medical importance, has evolved its strategies to acquire multiple metals, including iron, manganese, and zinc. Other important strategies for the colonization and infection of the host have been reported for staphylococci and include the expression of adhesins on the bacterial surface, as well as the acquisition of host plasminogen and complement regulatory proteins. Here we assess the ability of the zinc transport protein AdcA from Staphylococcus aureus, first characterized elsewhere as a zinc-binding protein of the ABC (ATP-binding cassette) transporters, to bind to host molecules. Like other staphylococcus ion-scavenging proteins, such as MntC, a manganese-binding protein, AdcA interacts with human plasminogen. Once activated, plasmin bound to AdcA cleaves fibrinogen and vitronectin. In addition, AdcA interacts with the human negative complement regulator factor H (FH). Plasminogen and FH have been shown to bind to distinct sites on the AdcA C-terminal portion. In conclusion, our in vitro data pave the way for future studies addressing the relevance of AdcA interactions with host molecules in vivo.
Collapse
|
8
|
Osorio-Aguilar Y, Gonzalez-Vazquez MC, Hernandez-Ceron DE, Lozano-Zarain P, Martinez-Laguna Y, Gonzalez-Bonilla CR, Rocha-Gracia RDC, Carabarin-Lima A. Structural Characterization of Haemophilus influenzae Enolase and Its Interaction with Human Plasminogen by In Silico and In Vitro Assays. Pathogens 2021; 10:pathogens10121614. [PMID: 34959569 PMCID: PMC8707213 DOI: 10.3390/pathogens10121614] [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: 11/10/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 12/02/2022] Open
Abstract
Haemophilus influenzae is the causal agent of invasive pediatric diseases, such as meningitis, epiglottitis, pneumonia, septic arthritis, pericarditis, cellulitis, and bacteremia (serotype b). Non-typeable H. influenzae (NTHi) strains are associated with localized infections, such as otitis media, conjunctivitis, sinusitis, bronchitis, and pneumonia, and can cause invasive diseases, such as as meningitis and sepsis in immunocompromised hosts. Enolase is a multifunctional protein and can act as a receptor for plasminogen, promoting its activation to plasmin, which leads to the degradation of components of the extracellular matrix, favoring host tissue invasion. In this study, using molecular docking, three important residues involved in plasminogen interaction through the plasminogen-binding motif (251EFYNKENGMYE262) were identified in non-typeable H. influenzae enolase (NTHiENO). Interaction with the human plasminogen kringle domains is conformationally stable due to the formation of four hydrogen bonds corresponding to enoTYR253-plgGLU1 (K2), enoTYR253-plgGLY310 (K3), and enoLYS255-plgARG471/enoGLU251-plgLYS468 (K5). On the other hand, in vitro assays, such as ELISA and far-western blot, showed that NTHiENO is a plasminogen-binding protein. The inhibition of this interaction using polyclonal anti-NTHiENO antibodies was significant. With these results, we can propose that NTHiENO–plasminogen interaction could be one of the mechanisms used by H. influenzae to adhere to and invade host cells.
Collapse
Affiliation(s)
- Yesenia Osorio-Aguilar
- Posgrado en Microbiología, Laboratorio de Microbiología Hospitalaria y de la Comunidad, Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (Y.O.-A.); (M.C.G.-V.); (P.L.-Z.); (Y.M.-L.); (R.d.C.R.-G.)
| | - Maria Cristina Gonzalez-Vazquez
- Posgrado en Microbiología, Laboratorio de Microbiología Hospitalaria y de la Comunidad, Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (Y.O.-A.); (M.C.G.-V.); (P.L.-Z.); (Y.M.-L.); (R.d.C.R.-G.)
| | | | - Patricia Lozano-Zarain
- Posgrado en Microbiología, Laboratorio de Microbiología Hospitalaria y de la Comunidad, Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (Y.O.-A.); (M.C.G.-V.); (P.L.-Z.); (Y.M.-L.); (R.d.C.R.-G.)
| | - Ygnacio Martinez-Laguna
- Posgrado en Microbiología, Laboratorio de Microbiología Hospitalaria y de la Comunidad, Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (Y.O.-A.); (M.C.G.-V.); (P.L.-Z.); (Y.M.-L.); (R.d.C.R.-G.)
| | | | - Rosa del Carmen Rocha-Gracia
- Posgrado en Microbiología, Laboratorio de Microbiología Hospitalaria y de la Comunidad, Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (Y.O.-A.); (M.C.G.-V.); (P.L.-Z.); (Y.M.-L.); (R.d.C.R.-G.)
| | - Alejandro Carabarin-Lima
- Posgrado en Microbiología, Laboratorio de Microbiología Hospitalaria y de la Comunidad, Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (Y.O.-A.); (M.C.G.-V.); (P.L.-Z.); (Y.M.-L.); (R.d.C.R.-G.)
- Licenciatura en Biotecnología, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico
- Correspondence: ; Tel.: +52-(222)-229-5500 (ext. 3965)
| |
Collapse
|
9
|
Esmail GA, Al-Dhabi NA, AlDawood B, Somily AM. Shotgun whole genome sequencing of drug-resistance Streptococcus anginosus strain 47S1 isolated from a patient with pharyngitis in Saudi Arabia. J Infect Public Health 2021; 14:1740-1749. [PMID: 34836797 DOI: 10.1016/j.jiph.2021.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND Streptococcus anginosus is an emergence opportunistic pathogen that colonize the human upper respiratory tract (URT), S. anginosus alongside with S. intermedius and S. constellatus, members of S. anginosus group, are implicated in several human infections. However, our understanding this bacterium to the genotype level with determining the genes associated with pathogenicity and antimicrobial resistance (AMR) is scarce. S. anginosus 47S1 strain was isolated from sore throat infection, the whole genome was characterized and the virulence & AMR genes contributing in pathogenicity were investigated. METHODOLOGY The whole genome of 47S1 was sequenced by Illumina sequencing technology. Strain 47S1 genome was de novo assembled with different strategies and annotated via PGAP, PROKKA and RAST pipelines. Identifying the CRISPR-Cass system and prophages sequences was performed using CRISPRloci and PhiSpy tools respectively. Prediction the virulence genes were performed with the VFDB database. AMR genes were detected in silico using NCBI AMRFinderPlus pipeline and CARD database and compared with in vitro AST findings. RESULTS β-hemolytic strain 47S1 was identified with conventional microbiology techniques and confirmed by the sequences of 16S rRNA gene. Genome of 47S1 comprised of 1981512 bp. Type I-C CRISPR-Cas system and 4 prophages were detected among the genome of 47S1. Several virulence genes were predicted, most of these genes are found in other pathogenic streptococci, mainly lmb, pavA, htrA/degP, eno, sagA, psaA and cpsI which play a significant role in colonizing, invading host tissues and evade form immune system. In silico AMR findings showed that 47S1 gnome harbors (tetA, tetB &tet32), (aac(6')-I, aadK &aph(3')-IVa), fusC, and PmrA genes that mediated-resistance to tetracyclines, aminoglycosides, fusidic acid, and fluoroquinolone respectively which corresponds with in vitro AST obtained results. In conclusion, WGS is a key approach to predict the virulence and AMR genes, results obtained in this study may contribute for a better understanding of the opportunistic S. anginosus pathogenicity.
Collapse
Affiliation(s)
- Galal Ali Esmail
- Department of Botany and Microbiology, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Naif Abdullah Al-Dhabi
- Department of Botany and Microbiology, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - Badr AlDawood
- Department of Emergency Medicine, College of Medicine, King Saud University, King Saud University Medical City, Riyadh 11461, Saudi Arabia
| | - Ali Mohammed Somily
- Department of Pathology and Laboratory Medicine/Microbiology, College of Medicine, King Saud University, King Saud University Medical City, Riyadh 11461, Saudi Arabia.
| |
Collapse
|
10
|
Ali MQ, Kohler TP, Schulig L, Burchhardt G, Hammerschmidt S. Pneumococcal Extracellular Serine Proteases: Molecular Analysis and Impact on Colonization and Disease. Front Cell Infect Microbiol 2021; 11:763152. [PMID: 34790590 PMCID: PMC8592123 DOI: 10.3389/fcimb.2021.763152] [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: 08/23/2021] [Accepted: 10/08/2021] [Indexed: 11/24/2022] Open
Abstract
The pathobiont Streptococcus pneumoniae causes life-threatening diseases, including pneumonia, sepsis, meningitis, or non-invasive infections such as otitis media. Serine proteases are enzymes that have been emerged during evolution as one of the most abundant and functionally diverse group of proteins in eukaryotic and prokaryotic organisms. S. pneumoniae expresses up to four extracellular serine proteases belonging to the category of trypsin-like or subtilisin-like family proteins: HtrA, SFP, PrtA, and CbpG. These serine proteases have recently received increasing attention because of their immunogenicity and pivotal role in the interaction with host proteins. This review is summarizing and focusing on the molecular and functional analysis of pneumococcal serine proteases, thereby discussing their contribution to pathogenesis.
Collapse
Affiliation(s)
- Murtadha Q Ali
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Lukas Schulig
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Gerhard Burchhardt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| |
Collapse
|
11
|
Coppolino F, Romeo L, Pietrocola G, Lentini G, De Gaetano GV, Teti G, Galbo R, Beninati C. Lysine Residues in the MK-Rich Region Are Not Required for Binding of the PbsP Protein From Group B Streptococci to Plasminogen. Front Cell Infect Microbiol 2021; 11:679792. [PMID: 34568085 PMCID: PMC8455988 DOI: 10.3389/fcimb.2021.679792] [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: 03/16/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Binding to plasminogen (Plg) enables bacteria to associate with and invade host tissues. The cell wall protein PbsP significantly contributes to the ability of group B streptococci, a frequent cause of invasive infection, to bind Plg. Here we sought to identify the molecular regions involved in the interactions between Plg and PbsP. The K4 Kringle domain of the Plg molecule was required for binding of Plg to whole PbsP and to a PbsP fragment encompassing a region rich in methionine and lysine (MK-rich domain). These interactions were inhibited by free L-lysine, indicating the involvement of lysine binding sites in the Plg molecule. However, mutation to alanine of all lysine residues in the MK-rich domain did not decrease its ability to bind Plg. Collectively, our data identify a novel bacterial sequence that can interact with lysine binding sites in the Plg molecule. Notably, such binding did not require the presence of lysine or other positively charged amino acids in the bacterial receptor. These data may be useful for developing alternative therapeutic strategies aimed at blocking interactions between group B streptococci and Plg.
Collapse
Affiliation(s)
- Francesco Coppolino
- Department of Biomedical, Dental and Imaging Sciences, University of Messina, Messina, Italy
| | - Letizia Romeo
- Department of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Giampiero Pietrocola
- Department Molecular Medicine, Biochemistry Section, University of Pavia, Pavia, Italy
| | - Germana Lentini
- Department of Human Pathology and Medicine, University of Messina, Messina, Italy
| | | | | | - Roberta Galbo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Concetta Beninati
- Department of Human Pathology and Medicine, University of Messina, Messina, Italy.,Scylla Biotech Srl, Messina, Italy
| |
Collapse
|
12
|
Shen J, Wu X, Yang Y, Lv Y, Li X, Ding X, Wang S, Yan Z, Yan Y, Yang F, Li H. Antimicrobial Resistance and Virulence Factor of Streptococcus dysgalactiae Isolated from Clinical Bovine Mastitis Cases in Northwest China. Infect Drug Resist 2021; 14:3519-3530. [PMID: 34511943 PMCID: PMC8418370 DOI: 10.2147/idr.s327924] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/24/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Streptococcus dysgalactiae is a major pathogen in bovine mastitis. The purpose of this study was to survey the prevalence, antimicrobial resistance, as well as the spread of resistance and virulence-associated gene of S. dysgalactiae. Methods A total of 60 S. dysgalactiae strains were obtained from 830 milk samples from Holstein cows with clinical mastitis. Antimicrobial resistance was examined by the disk diffusion method. Antimicrobial resistance and virulence genes were investigated by PCR, agarose gel electrophoresis and 16S rRNA gene sequencing. Results All isolates were resistant to tetracycline and showed a high level of resistance to aminoglycoside antibiotics, where 81.67% of the strains were multi-resistant to these ten sorts of antibiotics. In addition, the most prevalent resistance gene in S. dysgalactiae was aphA-1 (98.33%), followed by blaTEM (96.67%), ermB (83.3%), aadA1/aadA2 (78.33%) and tetL (73.33%). Totally, seven virulence genes with 25 combination patterns were detected in these isolates, and each isolates harbored at least one virulence gene. 21.67% of the isolates carried three or more virulence genes, while one strain with seven virulence-related genes and belonged to cfb+lmb+eno+napr+bca+scpB+cyl. Conclusion These findings indicate that S. dysgalactiae isolated from clinical bovine mastitis cases in Northwest China show a variety of molecular ecology and are highly resistant to antibiotics commonly used in dairy farms. This research will help investigators better understand the pathophysiology S. dysgalactiae in bovine mastitis and choose the appropriate antibiotics to treat mastitis.
Collapse
Affiliation(s)
- Jirao Shen
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xiaohu Wu
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Yayuan Yang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Yanan Lv
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xinpu Li
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xuezhi Ding
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Shengyi Wang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Zuoting Yan
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Yong Yan
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Feng Yang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Hongsheng Li
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| |
Collapse
|
13
|
Pathogen Moonlighting Proteins: From Ancestral Key Metabolic Enzymes to Virulence Factors. Microorganisms 2021; 9:microorganisms9061300. [PMID: 34203698 PMCID: PMC8232316 DOI: 10.3390/microorganisms9061300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 12/22/2022] Open
Abstract
Moonlighting and multitasking proteins refer to proteins with two or more functions performed by a single polypeptide chain. An amazing example of the Gain of Function (GoF) phenomenon of these proteins is that 25% of the moonlighting functions of our Multitasking Proteins Database (MultitaskProtDB-II) are related to pathogen virulence activity. Moreover, they usually have a canonical function belonging to highly conserved ancestral key functions, and their moonlighting functions are often involved in inducing extracellular matrix (ECM) protein remodeling. There are three main questions in the context of moonlighting proteins in pathogen virulence: (A) Why are a high percentage of pathogen moonlighting proteins involved in virulence? (B) Why do most of the canonical functions of these moonlighting proteins belong to primary metabolism? Moreover, why are they common in many pathogen species? (C) How are these different protein sequences and structures able to bind the same set of host ECM protein targets, mainly plasminogen (PLG), and colonize host tissues? By means of an extensive bioinformatics analysis, we suggest answers and approaches to these questions. There are three main ideas derived from the work: first, moonlighting proteins are not good candidates for vaccines. Second, several motifs that might be important in the adhesion to the ECM were identified. Third, an overrepresentation of GO codes related with virulence in moonlighting proteins were seen.
Collapse
|
14
|
Gani Z, Boradia VM, Kumar A, Patidar A, Talukdar S, Choudhary E, Singh R, Agarwal N, Raje M, Iyengar Raje C. Mycobacterium tuberculosis glyceraldehyde-3-phosphate dehydrogenase plays a dual role-As an adhesin and as a receptor for plasmin(ogen). Cell Microbiol 2021; 23:e13311. [PMID: 33486886 DOI: 10.1111/cmi.13311] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/30/2020] [Accepted: 01/19/2021] [Indexed: 11/30/2022]
Abstract
The spread of infection is directly determined by the ability of a pathogen to invade and infect host tissues. The process involves adherence due to host-pathogen interactions and traversal into deeper tissues. Mycobacterium tuberculosis (Mtb) primarily infects the lung but is unique in its ability to infect almost any other organ of the human host including immune privileged sites such as the central nervous system (CNS). The extreme invasiveness of this bacterium is not fully understood. In the current study, we report that cell surface Mtb glyceraldehyde-3-phosphate dehydrogenase (GAPDH) functions as a virulence factor by multiple mechanisms. Firstly, it serves as a dual receptor for both plasminogen (Plg) and plasmin (Plm). CRISPRi-mediated silencing of this essential enzyme confirmed its role in the recruitment of Plg/Plm. Our studies further demonstrate that soluble GAPDH can re-associate on Mtb bacilli to promote plasmin(ogen) recruitment. The direct association of plasmin(ogen) via cell surface GAPDH or by the re-association of soluble GAPDH enhanced bacterial adherence to and traversal across lung epithelial cells. Furthermore, the association of GAPDH with host extracellular matrix (ECM) proteins coupled with its ability to recruit plasmin(ogen) may endow cells with the ability of directed proteolytic activity vital for tissue invasion.
Collapse
Affiliation(s)
- Zahid Gani
- National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, India
| | - Vishant Mahendra Boradia
- National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, India.,Seattle Children's Research Institute, Global Center for Infectious Disease Research, Seattle, Washington, USA
| | - Ajay Kumar
- National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, India
| | - Anil Patidar
- Council of Scientific and Industrial Research-Institute of Microbial Technology (CSIR-IMTECH), Chandigarh, India
| | - Sharmila Talukdar
- Council of Scientific and Industrial Research-Institute of Microbial Technology (CSIR-IMTECH), Chandigarh, India
| | - Eira Choudhary
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad, India.,Symbiosis School of Biomedical Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Ranvir Singh
- National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, India
| | - Nisheeth Agarwal
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad, India
| | - Manoj Raje
- Council of Scientific and Industrial Research-Institute of Microbial Technology (CSIR-IMTECH), Chandigarh, India
| | - Chaaya Iyengar Raje
- National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, India
| |
Collapse
|
15
|
Kargarpour Z, Nasirzade J, Panahipour L, Miron RJ, Gruber R. Relative Centrifugal Force (RCF; G-Force) Affects the Distribution of TGF-β in PRF Membranes Produced Using Horizontal Centrifugation. Int J Mol Sci 2020; 21:E7629. [PMID: 33076376 PMCID: PMC7589083 DOI: 10.3390/ijms21207629] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 01/03/2023] Open
Abstract
Solid platelet-rich fibrin (PRF) is produced with centrifugation tubes designed to accelerate clotting. Thus, activated platelets may accumulate within the fibrin-rich extracellular matrix even before centrifugation is initiated. It can thus be assumed that platelets and their growth factors such as transforming growth factor-β (TGF-β) are trapped within PRF independent of their relative centrifugal force (RCF), the gravitation or g-force. To test this assumption, we prepared PRF membranes with tubes where clotting is activated by a silicone-coated interior. Tubes underwent 210 g, 650 g and 1500 g for 12 min in a horizontal centrifuge. The respective PRF membranes, either in total or separated into a platelet-poor plasma and buffy coat fraction, were subjected to repeated freeze-thawing to prepare lysates. Gingival fibroblasts were exposed to the PRF lysates to provoke the expression of TGF-β target genes. We show here that the expression of interleukin 11 (IL11) and NADPH oxidase 4 (NOX4), and Smad2/3 signaling were similarly activated by all lysates when normalized to the size of the PRF membranes. Notably, platelet-poor plasma had significantly less TGF-β activity than the buffy coat fraction at both high-speed protocols. In contrast to our original assumption, the TGF-β activity in PRF lysates produced using horizontal centrifugation follows a gradient with increasing concentration from the platelet-poor plasma towards the buffy coat layer.
Collapse
Affiliation(s)
- Zahra Kargarpour
- Department of Oral Biology, Medical University of Vienna, 1090 Vienna, Austria; (Z.K.); (J.N.); (L.P.)
| | - Jila Nasirzade
- Department of Oral Biology, Medical University of Vienna, 1090 Vienna, Austria; (Z.K.); (J.N.); (L.P.)
| | - Layla Panahipour
- Department of Oral Biology, Medical University of Vienna, 1090 Vienna, Austria; (Z.K.); (J.N.); (L.P.)
| | - Richard J. Miron
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland;
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, 1090 Vienna, Austria; (Z.K.); (J.N.); (L.P.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland;
| |
Collapse
|
16
|
Barrios-Arpi LM, Morales-Cauti SM. Cytomorphological characterization of lymphadenopathies in guinea pigs: study of 31 clinical cases. J Exot Pet Med 2020. [DOI: 10.1053/j.jepm.2019.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
17
|
Comparative Analysis of Blood Clot, Plasma Rich in Growth Factors and Platelet-Rich Fibrin Resistance to Bacteria-Induced Fibrinolysis. Microorganisms 2019; 7:microorganisms7090328. [PMID: 31500263 PMCID: PMC6780161 DOI: 10.3390/microorganisms7090328] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/22/2019] [Accepted: 09/06/2019] [Indexed: 12/14/2022] Open
Abstract
Alveolar osteitis (AO) is a common, painful postoperative complication after tooth extraction. Fibrinolytic activity in the extraction socket is one etiological factor. Platelet concentrates are used to prevent and treat AO. The aim of this study was to find out whether the positive effect of platelet concentrates can be related to resistance to bacteria-induced fibrinolysis. Blood from 45 human volunteers was used to prepare four media: blood clot medium as control group; PRF and PRGF first fraction (PRGF I) and PRGF second fraction (PRGF II) as study groups. Additionally, collected blood was used for blood plasma preparation on which evaluation of initial value of d-dimer concentration was performed. A solution of five different microbes (Staphylococcus aureus, Streptococcus pyogenes, Streptococcus pneumonia, Bacillus cereus, and Candida albicans) was adjusted to 0.5 McFarland (1 × 108 CFU/mL) and then diluted to 0.25 McFarland (0.5 × 108 CFU/mL). The d-dimer concentration was evaluated after one and three hours of bacteria exposure. The resistance to fibrinolysis was not statistically distinguished among any media groups at any time. S. pneumoniae was statistically active in PRF after three hours. C. albicans was statistically active in PRGF II after one hour and in PRF between the first and third hour and after three hours. S. aureus and B. cereus were statistically active in PRGF II after three hours. S. pyogenes was statistically active after one hour, between the first and third hour, and after the third hour in all groups. S. pyogenes was the most active bacterium. Different blood formulations were not distinguishable based on resistance to bacteria-induced fibrinolysis. Low fibrinolytic properties of the found major microbes suggests that bacteria-induced fibrinolysis is one of the leading causes of absence of a clot in a post-extraction socket to be clinically insignificant. The initial absence of a clot or its mechanical elimination during formation or the healing period are major causes of dry socket.
Collapse
|
18
|
Abstract
ABSTRACT
Of the eight phylogenetic groups comprising the genus
Streptococcus
, Lancefield group C and G streptococci (GCS and GGS, resp.) occupy four of them, including the Pyogenic, Anginosus, and Mitis groups, and one Unnamed group so far. These organisms thrive as opportunistic commensals in both humans and animals but may also be associated with clinically serious infections, often resembling those due to their closest genetic relatives, the group A streptoccci (GAS). Advances in molecular genetics, taxonomic approaches and phylogenomic studies have led to the establishment of at least 12 species, several of which being subdivided into subspecies. This review summarizes these advances, citing 264 early and recent references. It focuses on the molecular structure and genetic regulation of clinically important proteins associated with the cell wall, cytoplasmic membrane and extracellular environment. The article also addresses the question of how, based on the current knowledge, basic research and translational medicine might proceed to further advance our understanding of these multifaceted organisms. Particular emphasis in this respect is placed on streptokinase as the protein determining the host specificity of infection and the Rsh-mediated stringent response with its potential for supporting bacterial survival under nutritional stress conditions.
Collapse
|
19
|
Jagau H, Behrens IK, Lahme K, Lorz G, Köster RW, Schneppenheim R, Obser T, Brehm MA, König G, Kohler TP, Rohde M, Frank R, Tegge W, Fulde M, Hammerschmidt S, Steinert M, Bergmann S. Von Willebrand Factor Mediates Pneumococcal Aggregation and Adhesion in Blood Flow. Front Microbiol 2019; 10:511. [PMID: 30972039 PMCID: PMC6443961 DOI: 10.3389/fmicb.2019.00511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/27/2019] [Indexed: 12/24/2022] Open
Abstract
Streptococcus pneumoniae is a major cause of community acquired pneumonia and septicaemia in humans. These diseases are frequently associated with thromboembolic cardiovascular complications. Pneumococci induce the exocytosis of endothelial Weibel-Palade Bodies and thereby actively stimulate the release of von Willebrand factor (VWF), which is an essential glycoprotein of the vascular hemostasis. Both, the pneumococcus induced pulmonary inflammation and the thromboembolytic complications are characterized by a dysbalanced hemostasis including a marked increase in VWF plasma concentrations. Here, we describe for the first time VWF as a novel interaction partner of capsulated and non-encapsulated pneumococci. Moreover, cell culture infection analyses with primary endothelial cells characterized VWF as bridging molecule that mediates bacterial adherence to endothelial cells in a heparin-sensitive manner. Due to the mechanoresponsive changes of the VWF protein conformation and multimerization status, which occur in the blood stream, we used a microfluidic pump system to generate shear flow-induced multimeric VWF strings on endothelial cell surfaces and analyzed attachment of RFP-expressing pneumococci in flow. By applying immunofluorescence visualization and additional electron microscopy, we detected a frequent and enduring bacterial attachment to the VWF strings. Bacterial attachment to the endothelium was confirmed in vivo using a zebrafish infection model, which is described in many reports and acknowledged as suitable model to study hemostasis mechanisms and protein interactions of coagulation factors. Notably, we visualized the recruitment of zebrafish-derived VWF to the surface of pneumococci circulating in the blood stream and detected a VWF-dependent formation of bacterial aggregates within the vasculature of infected zebrafish larvae. Furthermore, we identified the surface-exposed bacterial enolase as pneumococcal VWF binding protein, which interacts with the VWF domain A1 and determined the binding kinetics by surface plasmon resonance. Subsequent epitope mapping using an enolase peptide array indicates that the peptide 181YGAEIFHALKKILKS195 might serve as a possible core sequence of the VWF interaction site. In conclusion, we describe a VWF-mediated mechanism for pneumococcal anchoring within the bloodstream via surface-displayed enolase, which promotes intravascular bacterial aggregation.
Collapse
Affiliation(s)
- Hilger Jagau
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ina-Kristin Behrens
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Karen Lahme
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Georgina Lorz
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Reinhard W Köster
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Reinhard Schneppenheim
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf (UKE Hamburg), Hamburg, Germany
| | - Tobias Obser
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf (UKE Hamburg), Hamburg, Germany
| | - Maria A Brehm
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf (UKE Hamburg), Hamburg, Germany
| | - Gesa König
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf (UKE Hamburg), Hamburg, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Universität Greifswald, Greifswald, Germany
| | - Manfred Rohde
- Helmholtz Centre for Infection Research, Central Facility for Microscopy, Braunschweig, Germany
| | - Ronald Frank
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Werner Tegge
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marcus Fulde
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Universität Greifswald, Greifswald, Germany
| | - Michael Steinert
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Simone Bergmann
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
20
|
Qiu C, Yuan Y, Liang Z, Lee SW, Ploplis VA, Castellino FJ. Variations in the secondary structures of PAM proteins influence their binding affinities to human plasminogen. J Struct Biol 2019; 206:193-203. [PMID: 30880082 DOI: 10.1016/j.jsb.2019.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/04/2019] [Accepted: 03/12/2019] [Indexed: 11/25/2022]
Abstract
M-proteins (M-Prts) are major virulence determinants of Group A Streptococcus pyogenes (GAS) that are covalently anchored to the cell wall at their conserved COOH-termini while the NH2-terminal regions extend through the capsule into extracellular space. Functional M-Prts are also secreted and/or released from GAS cells where they exist as helical coiled-coil dimers in solution. Certain GAS strains (Pattern D) uniquely express an M-protein (plasminogen-binding group A streptococcal M-protein; PAM) that directly interacts with human plasminogen (hPg), a process strongly implicated in the virulence of these strains. M-Prt expressed by the emm gene is employed to serotype over 250 known strains of GAS, ∼20 of which are hitherto found to express PAMs. We have developed a modular structural model of the PAM dimer that describes the roles of different domains of this protein in various functions. While the helical COOH-terminal domains of PAM are essential for dimerization in solution, regions of its NH2-terminal domains also exhibit a weak potential to dimerize. We find that temperature controls the open (unwound) or closed (wound) states of the functional NH2-terminal domains of PAM. As temperature increases, α-helices are dramatically reduced, which concomitantly destabilizes the helical coiled-coil PAM dimers. PAMs with two a-repeats within the variable NH2-terminal A-domain (class I/III) bind to hPg tightly, but natural PAM isolates with a single a-repeat in this domain (class II) display dramatic changes in hPg binding with temperature. We conclude that coexistence of two a-repeats in PAM is critical to achieve optimal binding to hPg, especially in its monomeric form, at the biologically relevant temperature.
Collapse
Affiliation(s)
- Cunjia Qiu
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Yue Yuan
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Zhong Liang
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Shaun W Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Victoria A Ploplis
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Francis J Castellino
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
21
|
Organization of multi-binding to host proteins: The glyceraldehyde-3-phosphate dehydrogenase (GAPDH) of Mycoplasma pneumoniae. Microbiol Res 2019; 218:22-31. [DOI: 10.1016/j.micres.2018.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/29/2018] [Accepted: 09/21/2018] [Indexed: 12/28/2022]
|
22
|
Crystal structure of GAPDH of Streptococcus agalactiae and characterization of its interaction with extracellular matrix molecules. Microb Pathog 2018; 127:359-367. [PMID: 30553015 DOI: 10.1016/j.micpath.2018.12.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/10/2018] [Indexed: 11/21/2022]
Abstract
GAPDH being a key enzyme in the glycolytic pathway is one of the surface adhesins of many Gram-positive bacteria including Streptococcus agalactiae. This anchorless adhesin is known to bind to host plasminogen (PLG) and fibrinogen (Fg), which enhances the virulence and modulates the host immune system. The crystal structure of the recombinant GAPDH from S. agalactiae (SagGAPDH) was determined at 2.6 Å resolution by molecular replacement. The structure was found to be highly conserved with a typical NAD binding domain and a catalytic domain. In this paper, using biolayer interferometry studies, we report that the multifunctional SagGAPDH enzyme binds to a variety of host molecules such as PLG, Fg, laminin, transferrin and mucin with a KD value of 4.4 × 10-7 M, 9.8 × 10-7 M, 1 × 10-5 M, 9.7 × 10-12 M and 1.4 × 10-7 M respectively. The ligand affinity blots reveal that SagGAPDH binds specifically to α and β subunits of Fg and the competitive binding ELISA assay reveals that the Fg and PLG binding sites on GAPDH does not overlap each other. The PLG binding motif of GAPDH varies with organisms, however positively charged residues in the hydrophobic surroundings is essential for PLG binding. The lysine analogue competitive binding assay and lysine succinylation experiments deciphered the role of SagGAPDH lysines in PLG binding. On structural comparison with S. pneumoniae GAPDH, K171 of SagGAPDH is being predicted to be involved in PLG binding. Further SagGAPDH exhibited enzymatic activity in the presence of Fg, PLG and transferrin. This suggests that these host molecules does not mask the active site and bind at some other region of GAPDH.
Collapse
|
23
|
Desvaux M, Candela T, Serror P. Surfaceome and Proteosurfaceome in Parietal Monoderm Bacteria: Focus on Protein Cell-Surface Display. Front Microbiol 2018; 9:100. [PMID: 29491848 PMCID: PMC5817068 DOI: 10.3389/fmicb.2018.00100] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 01/16/2018] [Indexed: 12/12/2022] Open
Abstract
The cell envelope of parietal monoderm bacteria (archetypal Gram-positive bacteria) is formed of a cytoplasmic membrane (CM) and a cell wall (CW). While the CM is composed of phospholipids, the CW is composed at least of peptidoglycan (PG) covalently linked to other biopolymers, such as teichoic acids, polysaccharides, and/or polyglutamate. Considering the CW is a porous structure with low selective permeability contrary to the CM, the bacterial cell surface hugs the molecular figure of the CW components as a well of the external side of the CM. While the surfaceome corresponds to the totality of the molecules found at the bacterial cell surface, the proteinaceous complement of the surfaceome is the proteosurfaceome. Once translocated across the CM, secreted proteins can either be released in the extracellular milieu or exposed at the cell surface by associating to the CM or the CW. Following the gene ontology (GO) for cellular components, cell-surface proteins at the CM can either be integral (GO: 0031226), i.e., the integral membrane proteins, or anchored to the membrane (GO: 0046658), i.e., the lipoproteins. At the CW (GO: 0009275), cell-surface proteins can be covalently bound, i.e., the LPXTG-proteins, or bound through weak interactions to the PG or wall polysaccharides, i.e., the cell wall binding proteins. Besides monopolypeptides, some proteins can associate to each other to form supramolecular protein structures of high molecular weight, namely the S-layer, pili, flagella, and cellulosomes. After reviewing the cell envelope components and the different molecular mechanisms involved in protein attachment to the cell envelope, perspectives in investigating the proteosurfaceome in parietal monoderm bacteria are further discussed.
Collapse
Affiliation(s)
- Mickaël Desvaux
- Université Clermont Auvergne, INRA, UMR454 MEDiS, Clermont-Ferrand, France
| | - Thomas Candela
- EA4043 Unité Bactéries Pathogènes et Santé, Châtenay-Malabry, France
| | - Pascale Serror
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
24
|
Siemens N, Oehmcke-Hecht S, Mettenleiter TC, Kreikemeyer B, Valentin-Weigand P, Hammerschmidt S. Port d'Entrée for Respiratory Infections - Does the Influenza A Virus Pave the Way for Bacteria? Front Microbiol 2017; 8:2602. [PMID: 29312268 PMCID: PMC5742597 DOI: 10.3389/fmicb.2017.02602] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022] Open
Abstract
Bacterial and viral co-infections of the respiratory tract are life-threatening and present a global burden to the global community. Staphylococcus aureus, Streptococcus pneumoniae, and Streptococcus pyogenes are frequent colonizers of the upper respiratory tract. Imbalances through acquisition of seasonal viruses, e.g., Influenza A virus, can lead to bacterial dissemination to the lower respiratory tract, which in turn can result in severe pneumonia. In this review, we summarize the current knowledge about bacterial and viral co-infections of the respiratory tract and focus on potential experimental models suitable for mimicking this disease. Transmission of IAV and pneumonia is mainly modeled by mouse infection. Few studies utilizing ferrets, rats, guinea pigs, rabbits, and non-human primates are also available. The knowledge gained from these studies led to important discoveries and advances in understanding these infectious diseases. Nevertheless, mouse and other infection models have limitations, especially in translation of the discoveries to humans. Here, we suggest the use of human engineered lung tissue, human ex vivo lung tissue, and porcine models to study respiratory co-infections, which might contribute to a greater translation of the results to humans and improve both, animal and human health.
Collapse
Affiliation(s)
- Nikolai Siemens
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sonja Oehmcke-Hecht
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Thomas C. Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Peter Valentin-Weigand
- Center for Infection Medicine, Institute for Microbiology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
25
|
Bergmann S, Eichhorn I, Kohler TP, Hammerschmidt S, Goldmann O, Rohde M, Fulde M. SCM, the M Protein of Streptococcus canis Binds Immunoglobulin G. Front Cell Infect Microbiol 2017; 7:80. [PMID: 28401063 PMCID: PMC5368172 DOI: 10.3389/fcimb.2017.00080] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 02/28/2017] [Indexed: 11/13/2022] Open
Abstract
The M protein of Streptococcus canis (SCM) is a virulence factor and serves as a surface-associated receptor with a particular affinity for mini-plasminogen, a cleavage product of the broad-spectrum serine protease plasmin. Here, we report that SCM has an additional high-affinity immunoglobulin G (IgG) binding activity. The ability of a particular S. canis isolate to bind to IgG significantly correlates with a scm-positive phenotype, suggesting a dominant role of SCM as an IgG receptor. Subsequent heterologous expression of SCM in non-IgG binding S. gordonii and Western Blot analysis with purified recombinant SCM proteins confirmed its IgG receptor function. As expected for a zoonotic agent, the SCM-IgG interaction is species-unspecific, with a particular affinity of SCM for IgGs derived from human, cats, dogs, horses, mice, and rabbits, but not from cows and goats. Similar to other streptococcal IgG-binding proteins, the interaction between SCM and IgG occurs via the conserved Fc domain and is, therefore, non-opsonic. Interestingly, the interaction between SCM and IgG-Fc on the bacterial surface specifically prevents opsonization by C1q, which might constitute another anti-phagocytic mechanism of SCM. Extensive binding analyses with a variety of different truncated SCM fragments defined a region of 52 amino acids located in the central part of the mature SCM protein which is important for IgG binding. This binding region is highly conserved among SCM proteins derived from different S. canis isolates but differs significantly from IgG-Fc receptors of S. pyogenes and S. dysgalactiae sub. equisimilis, respectively. In summary, we present an additional role of SCM in the pathogen-host interaction of S. canis. The detailed analysis of the SCM-IgG interaction should contribute to a better understanding of the complex roles of M proteins in streptococcal pathogenesis.
Collapse
Affiliation(s)
- Simone Bergmann
- Department of Medical Microbiology, Helmholtz Center for Infection Research Braunschweig, Germany
| | - Inga Eichhorn
- Institute of Microbiology and Epizootics, Centre for Infection Medicine, Freie Universität Berlin Berlin, Germany
| | - Thomas P Kohler
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt Universität Greifswald Greifswald, Germany
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt Universität Greifswald Greifswald, Germany
| | - Oliver Goldmann
- Department of Infection Immunology, Helmholtz Center for Infection Research Braunschweig, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Center for Infection Research Braunschweig, Germany
| | - Marcus Fulde
- Department of Medical Microbiology, Helmholtz Center for Infection ResearchBraunschweig, Germany; Institute of Microbiology and Epizootics, Centre for Infection Medicine, Freie Universität BerlinBerlin, Germany
| |
Collapse
|
26
|
Rahi A, Matta SK, Dhiman A, Garhyan J, Gopalani M, Chandra S, Bhatnagar R. Enolase of Mycobacterium tuberculosis is a surface exposed plasminogen binding protein. Biochim Biophys Acta Gen Subj 2016; 1861:3355-3364. [PMID: 27569900 DOI: 10.1016/j.bbagen.2016.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/12/2016] [Accepted: 08/24/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Enolase, a glycolytic enzyme, has long been studied as an anchorless protein present on the surface of many pathogenic bacteria that aids in tissue remodeling and invasion by binding to host plasminogen. METHODS Anti-Mtb enolase antibodies in human sera were detected using ELISA. Immunoelectron microscopy, immunofluorescence microscopy and flow cytometry were used to show surface localization of Mtb enolase. SPR was used to determine the affinity of enolase-plasminogen interaction. Plasmin formation upon plasminogen binding to enolase and Mtb surface was measured by ELISA. Mice challenge and histopathological studies were undertaken to determine the protective efficacy of enolase immunization. RESULTS Enolase of Mtb is present on its surface and binds human plasminogen with high affinity. There was an average of 2-fold increase in antibody mediated recognition of Mtb enolase in human sera from TB patients with an active disease over control individuals. Substitution of C-terminal lysine to alanine in rEno decreased its binding affinity with human plasminogen by >2-folds. Enolase bound plasminogen showed urokinase mediated conversion into plasmin. Binding of plasminogen to the surface of Mtb and its conversion into fibrinolytic plasmin was significantly reduced in the presence of anti-rEno antibodies. Immunization with rEno also led to a significant decrease in lung CFU counts of mice upon infection with Mtb H37Rv. CONCLUSIONS Mtb enolase is a surface exposed plasminogen binding protein which upon immunization confers significant protection against Mtb challenge. GENERAL SIGNIFICANCE Plasminogen binding has been recognized for Mtb, however, proteins involved have not been characterized. We show here that Mtb enolase is a moonlighting plasminogen binding protein.
Collapse
Affiliation(s)
- Amit Rahi
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Sumit Kumar Matta
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Alisha Dhiman
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Jaishree Garhyan
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Monisha Gopalani
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Subhash Chandra
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Rakesh Bhatnagar
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
27
|
Buscetta M, Firon A, Pietrocola G, Biondo C, Mancuso G, Midiri A, Romeo L, Galbo R, Venza M, Venza I, Kaminski PA, Gominet M, Teti G, Speziale P, Trieu-Cuot P, Beninati C. PbsP, a cell wall-anchored protein that binds plasminogen to promote hematogenous dissemination of group B Streptococcus. Mol Microbiol 2016; 101:27-41. [PMID: 26888569 DOI: 10.1111/mmi.13357] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2016] [Indexed: 02/04/2023]
Abstract
Streptococcus agalactiae (Group B Streptococcus or GBS) is a leading cause of invasive infections in neonates whose virulence is dependent on its ability to interact with cells and host components. We here characterized a surface protein with a critical function in GBS pathophysiology. This adhesin, designated PbsP, possesses two Streptococcal Surface Repeat domains, a methionine and lysine-rich region, and a LPXTG cell wall-anchoring motif. PbsP mediates plasminogen (Plg) binding both in vitro and in vivo and we showed that cell surface-bound Plg can be activated into plasmin by tissue plasminogen activator to increase the bacterial extracellular proteolytic activity. Absence of PbsP results in a decreased bacterial transmigration across brain endothelial cells and impaired virulence in a murine model of infection. PbsP is conserved among the main GBS lineages and is a major plasminogen adhesin in non-CC17 GBS strains. Importantly, immunization of mice with recombinant PbsP confers protective immunity. Our results indicate that GBS have evolved different strategies to recruit Plg which indicates that the ability to acquire cell surface proteolytic activity is essential for the invasiveness of this bacterium.
Collapse
Affiliation(s)
- Marco Buscetta
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy.,Institut Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS ERL3526, 75015, Paris, France
| | - Arnaud Firon
- Institut Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS ERL3526, 75015, Paris, France
| | - Giampiero Pietrocola
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | - Carmelo Biondo
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Giuseppe Mancuso
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Angelina Midiri
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Letizia Romeo
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Roberta Galbo
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Mario Venza
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Isabella Venza
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Pierre-Alexandre Kaminski
- Institut Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS ERL3526, 75015, Paris, France
| | - Myriam Gominet
- Institut Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS ERL3526, 75015, Paris, France
| | - Giuseppe Teti
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy
| | - Pietro Speziale
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | - Patrick Trieu-Cuot
- Institut Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS ERL3526, 75015, Paris, France
| | - Concetta Beninati
- Metchnikoff Laboratory, Departments of Human Pathology and Medicine, University of Messina, Messina, Italy.,Scylla Biotech Srl, Messina, Italy
| |
Collapse
|
28
|
Ermert D, Blom AM. C4b-binding protein: The good, the bad and the deadly. Novel functions of an old friend. Immunol Lett 2015; 169:82-92. [PMID: 26658464 DOI: 10.1016/j.imlet.2015.11.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 01/29/2023]
Abstract
C4b-binding protein (C4BP) is best known as a potent soluble inhibitor of the classical and lectin pathways of the complement system. This large 500 kDa multimeric plasma glycoprotein is expressed mainly in the liver but also in lung and pancreas. It consists of several identical 75 kDa α-chains and often also one 40 kDa β-chain, both of which are mainly composed of complement control protein (CCP) domains. Structure-function studies revealed that one crucial binding site responsible for inhibition of complement is located to CCP1-3 of the α-chain. Binding of anticoagulant protein S to the CCP1 of the β-chain provides C4BP with the ability to strongly bind apoptotic and necrotic cells in order to prevent inflammation arising from activation of complement by these cells. Further, C4BP interacts strongly with various types of amyloid and enhances fibrillation of islet amyloid polypeptide secreted from pancreatic beta cells, which may attenuate pro-inflammatory and cytotoxic effects of this amyloid. Full deficiency of C4BP has not been identified but non-synonymous alterations in its sequence have been found in haemolytic uremic syndrome and recurrent pregnancy loss. Furthermore, C4BP is bound by several bacterial pathogens, notably Streptococcus pyogenes, which due to inhibition of complement and enhancement of bacterial adhesion to endothelial cells provides these bacteria with a survival advantage in the host. Thus, depending on the context, C4BP has a protective or detrimental role in the organism.
Collapse
Affiliation(s)
- David Ermert
- Lund University, Department of Translational Medicine, Division of Medical Protein Chemistry, Inga Marie Nilssons Street 53, Malmö, 20502, Sweden.
| | - Anna M Blom
- Lund University, Department of Translational Medicine, Division of Medical Protein Chemistry, Inga Marie Nilssons Street 53, Malmö, 20502, Sweden.
| |
Collapse
|
29
|
Guinet F, Avé P, Filali S, Huon C, Savin C, Huerre M, Fiette L, Carniel E. Dissociation of Tissue Destruction and Bacterial Expansion during Bubonic Plague. PLoS Pathog 2015; 11:e1005222. [PMID: 26484539 PMCID: PMC4615631 DOI: 10.1371/journal.ppat.1005222] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 09/22/2015] [Indexed: 01/14/2023] Open
Abstract
Activation and/or recruitment of the host plasmin, a fibrinolytic enzyme also active on extracellular matrix components, is a common invasive strategy of bacterial pathogens. Yersinia pestis, the bubonic plague agent, expresses the multifunctional surface protease Pla, which activates plasmin and inactivates fibrinolysis inhibitors. Pla is encoded by the pPla plasmid. Following intradermal inoculation, Y. pestis has the capacity to multiply in and cause destruction of the lymph node (LN) draining the entry site. The closely related, pPla-negative, Y. pseudotuberculosis species lacks this capacity. We hypothesized that tissue damage and bacterial multiplication occurring in the LN during bubonic plague were linked and both driven by pPla. Using a set of pPla-positive and pPla-negative Y. pestis and Y. pseudotuberculosis strains in a mouse model of intradermal injection, we found that pPla is not required for bacterial translocation to the LN. We also observed that a pPla-cured Y. pestis caused the same extensive histological lesions as the wild type strain. Furthermore, the Y. pseudotuberculosis histological pattern, characterized by infectious foci limited by inflammatory cell infiltrates with normal tissue density and follicular organization, was unchanged after introduction of pPla. However, the presence of pPla enabled Y. pseudotuberculosis to increase its bacterial load up to that of Y. pestis. Similarly, lack of pPla strongly reduced Y. pestis titers in LNs of infected mice. This pPla-mediated enhancing effect on bacterial load was directly dependent on the proteolytic activity of Pla. Immunohistochemistry of Pla-negative Y. pestis-infected LNs revealed extensive bacterial lysis, unlike the numerous, apparently intact, microorganisms seen in wild type Y. pestis-infected preparations. Therefore, our study demonstrates that tissue destruction and bacterial survival/multiplication are dissociated in the bubo and that the primary action of Pla is to protect bacteria from destruction rather than to alter the tissue environment to favor Y. pestis propagation in the host. The hallmark of bubonic plague, a disease that ravaged Medieval Europe and is still prevalent in several countries, is the bubo, a highly inflammatory and painful lymph node, which is characterized by high concentrations of bacteria within a severely damaged organ. Yersinia pestis, the causative agent, expresses a surface protease, Pla, critical to the development of bubonic plague. This multitarget protease has the potential to activate the fibrinolytic pathway and to promote destruction of extracellular protein networks within tissues. Hence, it was expected that Pla was responsible for the tissue destructions of the bubo, and consequently, for bacterial propagation and virulence. However, we found, using various engineered Yersinia strains in a mouse model of bubonic plague, that Pla proteolytic activity was dispensable for lymph node alteration, but was required to achieve high bacterial loads in the organ. Further analysis showed that Pla is essential for preventing the bacteria from being destroyed in the host. Therefore, the role of Pla as a virulence factor is to protect Y. pestis survival and integrity in the host, rather than to assist its spread through tissue destruction.
Collapse
Affiliation(s)
- Françoise Guinet
- Unité de Recherche Yersinia, Institut Pasteur, Paris, France
- * E-mail: (FG); (EC)
| | - Patrick Avé
- Unité d’Histopathologie Humaine et Modèles Animaux, Institut Pasteur, Paris, France
| | - Sofia Filali
- Unité de Recherche Yersinia, Institut Pasteur, Paris, France
| | - Christèle Huon
- Unité de Recherche Yersinia, Institut Pasteur, Paris, France
| | - Cyril Savin
- Unité de Recherche Yersinia, Institut Pasteur, Paris, France
| | - Michel Huerre
- Unité de Recherche et d’Expertise d’Histotechnologie et Pathologie, Institut Pasteur, Paris, France
| | - Laurence Fiette
- Unité d’Histopathologie Humaine et Modèles Animaux, Institut Pasteur, Paris, France
| | - Elisabeth Carniel
- Unité de Recherche Yersinia, Institut Pasteur, Paris, France
- * E-mail: (FG); (EC)
| |
Collapse
|
30
|
Gonzalez T, Gaultney RA, Floden AM, Brissette CA. Escherichia coli lipoprotein binds human plasminogen via an intramolecular domain. Front Microbiol 2015; 6:1095. [PMID: 26500634 PMCID: PMC4595779 DOI: 10.3389/fmicb.2015.01095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/22/2015] [Indexed: 11/13/2022] Open
Abstract
Escherichia coli lipoprotein (Lpp) is a major cellular component that exists in two distinct states, bound-form and free-form. Bound-form Lpp is known to interact with the periplasmic bacterial cell wall, while free-form Lpp is localized to the bacterial cell surface. A function for surface-exposed Lpp has yet to be determined. We hypothesized that the presence of C-terminal lysinses in the surface-exposed region of Lpp would facilitate binding to the host zymogen plasminogen (Plg), a protease commandeered by a number of clinically important bacteria. Recombinant Lpp was synthesized and the binding of Lpp to Plg, the effect of various inhibitors on this binding, and the effects of various mutations of Lpp on Lpp-Plg interactions were examined. Additionally, the ability of Lpp-bound Plg to be converted to active plasmin was analyzed. We determined that Lpp binds Plg via an atypical domain located near the center of mature Lpp that may not be exposed on the surface of intact E. coli according to the current localization model. Finally, we found that Plg bound by Lpp can be converted to active plasmin. While the consequences of Lpp binding Plg are unclear, these results prompt further investigation of the ability of surface exposed Lpp to interact with host molecules such as extracellular matrix components and complement regulators, and the role of these interactions in infections caused by E. coli and other bacteria.
Collapse
Affiliation(s)
- Tammy Gonzalez
- Brissette Laboratory, Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks ND, USA
| | - Robert A Gaultney
- Brissette Laboratory, Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks ND, USA
| | - Angela M Floden
- Brissette Laboratory, Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks ND, USA
| | - Catherine A Brissette
- Brissette Laboratory, Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks ND, USA
| |
Collapse
|
31
|
Chaves EGA, Weber SS, Báo SN, Pereira LA, Bailão AM, Borges CL, Soares CMDA. Analysis of Paracoccidioides secreted proteins reveals fructose 1,6-bisphosphate aldolase as a plasminogen-binding protein. BMC Microbiol 2015; 15:53. [PMID: 25888027 PMCID: PMC4357084 DOI: 10.1186/s12866-015-0393-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 02/18/2015] [Indexed: 12/26/2022] Open
Abstract
Background Despite being important thermal dimorphic fungi causing Paracoccidioidomycosis, the pathogenic mechanisms that underlie the genus Paracoccidioides remain largely unknown. Microbial pathogens express molecules that can interact with human plasminogen, a protein from blood plasma, which presents fibrinolytic activity when activated into plasmin. Additionally, plasmin exhibits the ability of degrading extracellular matrix components, favoring the pathogen spread to deeper tissues. Previous work from our group demonstrated that Paracoccidioides presents enolase, as a protein able to bind and activate plasminogen, increasing the fibrinolytic activity of the pathogen, and the potential for adhesion and invasion of the fungus to host cells. By using proteomic analysis, we aimed to identify other proteins of Paracoccidioides with the ability of binding to plasminogen. Results In the present study, we employed proteomic analysis of the secretome, in order to identify plasminogen-binding proteins of Paracoccidioides, Pb01. Fifteen proteins were present in the fungal secretome, presenting the ability to bind to plasminogen. Those proteins are probable targets of the fungus interaction with the host; thus, they could contribute to the invasiveness of the fungus. For validation tests, we selected the protein fructose 1,6-bisphosphate aldolase (FBA), described in other pathogens as a plasminogen-binding protein. The protein FBA at the fungus surface and the recombinant FBA (rFBA) bound human plasminogen and promoted its conversion to plasmin, potentially increasing the fibrinolytic capacity of the fungus, as demonstrated in fibrin degradation assays. The addition of rFBA or anti-rFBA antibodies was capable of reducing the interaction between macrophages and Paracoccidioides, possibly by blocking the binding sites for FBA. These data reveal the possible participation of the FBA in the processes of cell adhesion and tissue invasion/dissemination of Paracoccidioides. Conclusions These data indicate that Paracoccidioides is a pathogen that has several plasminogen-binding proteins that likely play important roles in pathogen-host interaction. In this context, FBA is a protein that might be involved somehow in the processes of invasion and spread of the fungus during infection.
Collapse
Affiliation(s)
- Edilânia Gomes Araújo Chaves
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICBII, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, Goiás, Brazil.
| | - Simone Schneider Weber
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICBII, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, Goiás, Brazil.
| | - Sonia Nair Báo
- Laboratório de Microscopia, Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Distrito Federal, Brazil.
| | - Luiz Augusto Pereira
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICBII, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, Goiás, Brazil.
| | - Alexandre Melo Bailão
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICBII, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, Goiás, Brazil.
| | - Clayton Luiz Borges
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICBII, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, Goiás, Brazil.
| | - Célia Maria de Almeida Soares
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICBII, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, Goiás, Brazil.
| |
Collapse
|
32
|
From plasminogen to plasmin: role of plasminogen receptors in human cancer. Int J Mol Sci 2014; 15:21229-52. [PMID: 25407528 PMCID: PMC4264222 DOI: 10.3390/ijms151121229] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 11/04/2014] [Accepted: 11/12/2014] [Indexed: 12/02/2022] Open
Abstract
Cell surface-associated proteolysis mediated by plasmin (PLA) is an essential feature of wound healing, angiogenesis and cell invasion, processes that are dysregulated in cancer development, progression and systemic spread. The generation of PLA, initiated by the binding of its precursor plasminogen (PLG) to the cell surface, is regulated by an array of activators, inhibitors and receptors. In this review, we will highlight the importance of the best-characterized components of the PLG/PLA cascade in the pathogenesis of cancer focusing on the role of the cell surface-PLG receptors (PLG-R). PLG-R overexpression has been associated with poor prognosis of cancer patients and resistance to chemotherapy. We will also discuss recent findings on the molecular mechanisms regulating cell surface expression and distribution of PLG-R.
Collapse
|
33
|
Abdelsalam M, Fujino M, Eissa A, Chen S, Warda M. Expression, genetic localization and phylogenic analysis of NAPlr in piscine Streptococcus dysgalactiae subspecies dysgalactiae isolates and their patterns of adherence. J Adv Res 2014; 6:747-55. [PMID: 26425363 PMCID: PMC4563591 DOI: 10.1016/j.jare.2014.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/16/2014] [Accepted: 05/16/2014] [Indexed: 01/19/2023] Open
Abstract
Streptococcus dysgalactiae, the long recognized mammalian pathogen, has currently received a major concern regarding fish bacterial infection. Adhesion to host epithelial cells and the presence of wall-associated plasminogen binding proteins are prerequisites to Streptococcus infection. This is the first study of the occurrence of nephritis-associated plasminogen-binding receptor (NAPlr) and α-enolase genes in piscine S. dysgalactiae subspecies dysgalactiae (SDSD) isolates. Further characterization of surface localized NAPlr of fish SDSD revealed a similar immune-reactive band of 43 KDa as that from porcine S. dysgalactiae subsp. equisimilis (SDSE). The phylogenetic analysis revealed that NAPlr of fish SDSD is more associated with those of mammalian SDSE and Streptococcus pyogenes rather than of other streptococci. Our findings warrant public attention to the possible implication of these virulence genes in dissemination of SDSD to different tissues of infected hosts and to get advantage to new niches. The SDSD adherence patterns were also studied to better understand their pathogenicity. The patterns of adherence of SDSD on two different cell lines showed a different pattern of adherence. Such difference gives an insight about the variance in host susceptibility to infection.
Collapse
Affiliation(s)
- M. Abdelsalam
- Department of Fish Diseases and Management, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
- Corresponding authors. Tel.: +20 2 1122671243, +2 2 35720399; fax: +20 2 35725240, +20 2 35710305.
| | - M. Fujino
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - A.E. Eissa
- Department of Fish Diseases and Management, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
- Departments of Poultry and Fish Diseases, Faculty of Veterinary Medicine, Tripoli University, Tripoli, Libya
| | - S.C. Chen
- Graduate Institute of Animal Vaccine Technology, National Pingtung University of Science and Technology, Pingtung, Taiwan
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - M. Warda
- Department of Biochemistry, Biotechnology Center for Services and Researches, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
- Corresponding authors. Tel.: +20 2 1122671243, +2 2 35720399; fax: +20 2 35725240, +20 2 35710305.
| |
Collapse
|
34
|
Raghunathan K, Harris PT, Spurbeck RR, Arvidson CG, Arvidson DN. Crystal structure of an efficacious gonococcal adherence inhibitor: An enolase fromLactobacillus gasseri. FEBS Lett 2014; 588:2212-6. [DOI: 10.1016/j.febslet.2014.05.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/09/2014] [Accepted: 05/09/2014] [Indexed: 12/01/2022]
|