1
|
Kumari K, Dey J, Mahapatra SR, Ma Y, Sharma PK, Misra N, Singh RP. Protein profiling and immunoinformatic analysis of the secretome of a metal-resistant environmental isolate Pseudomonas aeruginosa S-8. Folia Microbiol (Praha) 2024; 69:1095-1122. [PMID: 38457114 DOI: 10.1007/s12223-024-01152-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 02/03/2024] [Indexed: 03/09/2024]
Abstract
The bacterial secretome represents a comprehensive catalog of proteins released extracellularly that have multiple important roles in virulence and intercellular communication. This study aimed to characterize the secretome of an environmental isolate Pseudomonas aeruginosa S-8 by analyzing trypsin-digested culture supernatant proteins using nano-LC-MS/MS tool. Using a combined approach of bioinformatics and mass spectrometry, 1088 proteins in the secretome were analyzed by PREDLIPO, SecretomeP 2.0, SignalP 4.1, and PSORTb tool for their subcellular localization and further categorization of secretome proteins according to signal peptides. Using the gene ontology tool, secretome proteins were categorized into different functional categories. KEGG pathway analysis identified the secreted proteins into different metabolic functional pathways. Moreover, our LC-MS/MS data revealed the secretion of various CAZymes into the extracellular milieu, which suggests its strong biotechnological applications to breakdown complex carbohydrate polymers. The identified immunodominant epitopes from the secretome of P. aeruginosa showed the characteristic of being non-allergenic, highly antigenic, nontoxic, and having a low risk of triggering autoimmune responses, which highlights their potential as successful vaccine targets. Overall, the identification of secreted proteins of P. aeruginosa could be important for both diagnostic purposes and the development of an effective candidate vaccine.
Collapse
Affiliation(s)
- Kiran Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Ranchi, 835215, India
| | - Jyotirmayee Dey
- School of Biotechnology, Deemed to Be University, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, 751024, India
| | - Soumya Ranjan Mahapatra
- School of Biotechnology, Deemed to Be University, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, 751024, India
| | - Ying Ma
- College of Resources and Environment, Southwest University, Chongqing, China
| | - Parva Kumar Sharma
- Department of Plant Sciences and Landscape Architecture, University of Maryland, College Park, MD, 20742, USA
| | - Namrata Misra
- School of Biotechnology, Deemed to Be University, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, 751024, India
| | - Rajnish Prakash Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, 201309, India.
| |
Collapse
|
2
|
Del Carpio AMG, Freire CA, Andrade FB, Piazza RMF, Silva RM, Carvalho E, Elias WP. Genomic Dissection of an Enteroaggregative Escherichia coli Strain Isolated from Bacteremia Reveals Insights into Its Hybrid Pathogenic Potential. Int J Mol Sci 2024; 25:9238. [PMID: 39273188 PMCID: PMC11394720 DOI: 10.3390/ijms25179238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 09/15/2024] Open
Abstract
Escherichia coli is a frequent pathogen isolated from bloodstream infections. This study aimed to characterize the genetic features of EC092, an E. coli strain isolated from bacteremia that harbors enteroaggregative E. coli (EAEC) genetic markers, indicating its hybrid pathogenic potential. Whole-genome sequencing showed that EC092 belongs to phylogroup B1, ST278, and serotype O165:H4. Genes encoding virulence factors such as fimbriae, toxins, iron-uptake systems, autotransporter proteins (Pet, Pic, Sat, and SepA), and secretion systems were detected, as well as EAEC virulence genes (aggR, aatA, aaiC, and aap). EC092 was found to be closely related to the other EAEC prototype strains and highly similar in terms of virulence to three EAEC strains isolated from diarrhea. The genomic neighborhood of pet, pic, sat, sepA, and the EAEC virulence genes of EC092 and its three genetically related fecal EAEC strains showed an identical genomic organization and nucleotide sequences. Also, EC092 produced and secreted Pet, Pic, Sat, and SepA in the culture supernatant and resisted the bactericidal activity of normal human serum. Our results demonstrate that the strain EC092, isolated from bacteremia, is a hybrid pathogenic extraintestinal E. coli (ExPEC)/EAEC with virulence features that could mediate both extraintestinal and intestinal infections.
Collapse
Affiliation(s)
| | - Claudia A Freire
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Fernanda B Andrade
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Roxane M F Piazza
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Rosa M Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Eneas Carvalho
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Waldir P Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| |
Collapse
|
3
|
Sil BK, Jamiruddin MR, Paul PK, Aekwattanaphol N, Nakpheng T, Haq MA, Buatong W, Srichana T. Ascorbic acid as serine protease inhibitor in lung cancer cell line and human serum albumin. PLoS One 2024; 19:e0303706. [PMID: 39042609 PMCID: PMC11265676 DOI: 10.1371/journal.pone.0303706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/30/2024] [Indexed: 07/25/2024] Open
Abstract
Serine proteases (SPs) are distributed among all living cells accounting for almost one-third of all proteases. Dysregulation of SPs during inflammation and/or infection can result in devastating consequences, such as skin and lung inflammation, neuroinflammation, arthritis, as well as metastasis of cancerous cells. Such activities are tightly regulated by various inhibitors known as serine protease inhibitors (SERPIN). The thermodynamic investigations previously revealed that L-ascorbic acid binds to trypsin more firmly than pepsin and the binding force of L-ascorbic acid is driven by hydrogen bonds and van der Waals forces. However, the physiochemical effects of such interaction on trypsin and/or pepsin have not yet been reported. Ascorbic acid, also known as vitamin C, is one of the essential nutrients and most common food supplements, fortificants, and preservatives. The aim of this study was to explore the inhibitory effects of ascorbic acid on serine proteases at various concentrations on the in-vitro digestion and/or hydrolysis of intercellular matrix of cell monolayer and human serum albumin (HSA). The inhibitory effects of ascorbic on trypsin are investigated by qualitative and quantitative analysis using SDS-PAGE imaging and NIH densitometric software. Upon the addition of ascorbic acid in both indicator systems, the detachment and/or dissociation of cell monolayer and the digestion of HSA were inhibited in the presence of EDTA-Trypsin. The inhibitory effect of ascorbic acid on the digestion of intercellular matrix and/or hydrolysis of HSA showed a dose-dependent trend until it reached the maximum extent of inhibition. At an equal concentration (2.5mg/mL) ascorbic acid and EDTA-Trypsin exhibited the most potent inhibitory effect on the in vitro digestion of protein either in the form of intercellular matrix in cell monolayer and/or HSA respectively. Overall, our results based on two indicator systems strongly indicate that ascorbic acid may function as a serine protease inhibitor (SERPIN) beyond other important functions.
Collapse
Affiliation(s)
- Bijon Kumar Sil
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
| | | | - Pijush Kumar Paul
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Nattanit Aekwattanaphol
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Titpawan Nakpheng
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Md. Ahsanul Haq
- Immunobiology, Nutrition and Toxicology Lab, Nutrition Research Division, icddr,b, Dhaka, Bangladesh
| | - Wilaiporn Buatong
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Teerapol Srichana
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
4
|
Sangal V, Marrs ECL, Nelson A, Perry JD. Phylogenomic analyses of multidrug resistant Corynebacterium striatum strains isolated from patients in a tertiary care hospital in the UK. Eur J Clin Microbiol Infect Dis 2024; 43:1495-1501. [PMID: 38801486 PMCID: PMC11271431 DOI: 10.1007/s10096-024-04857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
Corynebacterium striatum is an emerging nosocomial pathogen. This is the first report showing the presence of three distinct multidrug resistant lineages of C. striatum among patients in a UK hospital. The presence of ErmX, Tet(W), Bla and AmpC proteins, and mutations in gyrA gene are associated with the resistance to clindamycin, doxycycline, penicillin and moxifloxacin, respectively. These strains are equipped with several corynebacterial virulence genes including two SpaDEF-type and a novel pilus gene cluster, which needs further molecular characterisation. This study highlights a need of developing an active surveillance strategy for routine monitoring and preventing potential cross-transmission among susceptible patients.
Collapse
Affiliation(s)
- Vartul Sangal
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK.
| | - Emma C L Marrs
- Microbiology Research Department, Freeman Hospital, Newcastle upon Tyne, UK
| | - Andrew Nelson
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - John D Perry
- Microbiology Research Department, Freeman Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
5
|
Méndez LR, Rodríguez-Cornejo T, Rodríguez-Ramos T, Al-Hussinee L, Velázquez J, Campbell JH, Carpio Y, Estrada MP, Dixon B. PACAP sequence modifications modulate the peptide antimicrobial activity against bacterial pathogens affecting aquaculture. FISH & SHELLFISH IMMUNOLOGY 2024; 148:109512. [PMID: 38499216 DOI: 10.1016/j.fsi.2024.109512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
The global aquaculture industry has significant losses each year due to disease outbreaks. Antibiotics are one of the common methods to treat fish infections, but prolonged use can lead to the emergence of resistant strains. Aeromonas spp. Infections are a common and problematic disease in fish, and members of this genera can produce antibiotic resistant strains. Antimicrobial peptides (AMPs) have emerged as an alternative method to treat and prevent infections and pituitary adenylate cyclase activating polypeptide (PACAP) is a prominent member of this family. The objective of this research was to study PACAP's direct antimicrobial activity and its toxicity in fish cells. Four synthetic variants of the natural PACAP from Clarias gariepinus were tested in addition to the natural variant. The experimental results show a different antimicrobial activity against A. salmonicida and A. hydrophila of each PACAP variant, and for the first time show dependence on the culture broth used. Furthermore, the results suggest that the underlying mechanism of PACAP antimicrobial activity includes a bacterial membrane permeabilizing effect, classifying PACAP as a membrane disruptive AMP. This study also demonstrated that the five PACAP variants evaluated showed low toxicity in vitro, at concentrations relevant for in vivo applications. Therefore, PACAP could be a promising alternative to antibiotics in the aquaculture sector.
Collapse
Affiliation(s)
- Laura Rivera Méndez
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, Canada
| | | | - Tania Rodríguez-Ramos
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, Canada
| | - Lowia Al-Hussinee
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, Canada; Animal Health Laboratory, OVC, Guelph University, Canada
| | - Janet Velázquez
- Animal Biotechnology Department, Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, Havana, 10600, Cuba
| | - James Hugh Campbell
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, Canada
| | - Yamila Carpio
- Animal Biotechnology Department, Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, Havana, 10600, Cuba
| | - Mario Pablo Estrada
- Animal Biotechnology Department, Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, Havana, 10600, Cuba
| | - Brian Dixon
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, Canada.
| |
Collapse
|
6
|
Hou JJ, Ding L, Yang T, Yang YF, Jin YP, Zhang XP, Ma AH, Qin YH. The proteolytic activity in inflammatory bowel disease: insight from gut microbiota. Microb Pathog 2024; 188:106560. [PMID: 38272327 DOI: 10.1016/j.micpath.2024.106560] [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: 08/02/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent inflammatory disease caused by the destruction of the intestinal mucosal epithelium that affects a growing number of people worldwide. Although the etiology of IBD is complex and still elucidated, the role of dysbiosis and dysregulated proteolysis is well recognized. Various studies observed altered composition and diversity of gut microbiota, as well as increased proteolytic activity (PA) in serum, plasma, colonic mucosa, and fecal supernatant of IBD compared to healthy individuals. The imbalance of intestinal microecology and intestinal protein hydrolysis were gradually considered to be closely related to IBD. Notably, the pivotal role of intestinal microbiota in maintaining proteolytic balance received increasing attention. In summary, we have speculated a mesmerizing story, regarding the hidden role of PA and microbiota-derived PA hidden in IBD. Most importantly, we provided the diagnosis and therapeutic targets for IBD as well as the formulation of new treatment strategies for other digestive diseases and protease-related diseases.
Collapse
Affiliation(s)
- Jun-Jie Hou
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Liang Ding
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Tao Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yan-Fei Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Ping Jin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Xiao-Ping Zhang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - A-Huo Ma
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China.
| |
Collapse
|
7
|
Correa GB, Freire CA, Dibo M, Huerta-Cantillo J, Navarro-Garcia F, Barbosa AS, Elias WP, Moraes CTP. Plasmid-encoded toxin of Escherichia coli cleaves complement system proteins and inhibits complement-mediated lysis in vitro. Front Cell Infect Microbiol 2024; 14:1327241. [PMID: 38371299 PMCID: PMC10869522 DOI: 10.3389/fcimb.2024.1327241] [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: 10/24/2023] [Accepted: 01/08/2024] [Indexed: 02/20/2024] Open
Abstract
Plasmid-encoded toxin (Pet) is an autotransporter protein of the serine protease autotransporters of Enterobacteriaceae (SPATE) family, important in the pathogenicity of Escherichia coli. The pet gene was initially found in the enteroaggregative E. coli (EAEC) virulence plasmid, pAA2. Although this virulence factor was initially described in EAEC, an intestinal E. coli pathotype, pet may also be present in other pathotypes, including extraintestinal pathogenic strains (ExPEC). The complement system is an important defense mechanism of the immune system that can be activated by invading pathogens. Proteases produced by pathogenic bacteria, such as SPATEs, have proteolytic activity and can cleave components of the complement system, promoting bacterial resistance to human serum. Considering these factors, the proteolytic activity of Pet and its role in evading the complement system were investigated. Proteolytic assays were performed by incubating purified components of the complement system with Pet and Pet S260I (a catalytic site mutant) proteins. Pet, but not Pet S260I, could cleave C3, C5 and C9 components, and also inhibited the natural formation of C9 polymers. Furthermore, a dose-dependent inhibition of ZnCl2-induced C9 polymerization in vitro was observed. E. coli DH5α survived incubation with human serum pre-treated with Pet. Therefore, Pet can potentially interfere with the alternative and the terminal pathways of the complement system. In addition, by cleaving C9, Pet may inhibit membrane attack complex (MAC) formation on the bacterial outer membrane. Thus, our data are suggestive of a role of Pet in resistance of E. coli to human serum.
Collapse
Affiliation(s)
| | | | - Miriam Dibo
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Jazmin Huerta-Cantillo
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | | | - Waldir P. Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | | |
Collapse
|
8
|
Soto Perezchica MM, Guerrero Barrera AL, Avelar Gonzalez FJ, Quezada Tristan T, Macias Marin O. Actinobacillus pleuropneumoniae, surface proteins and virulence: a review. Front Vet Sci 2023; 10:1276712. [PMID: 38098987 PMCID: PMC10720984 DOI: 10.3389/fvets.2023.1276712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/17/2023] [Indexed: 12/17/2023] Open
Abstract
Actinobacillus pleuropneumoniae (App) is a globally distributed Gram-negative bacterium that produces porcine pleuropneumonia. This highly contagious disease produces high morbidity and mortality in the swine industry. However, no effective vaccine exists to prevent it. The infection caused by App provokes characteristic lesions, such as edema, inflammation, hemorrhage, and necrosis, that involve different virulence factors. The colonization and invasion of host surfaces involved structures and proteins such as outer membrane vesicles (OMVs), pili, flagella, adhesins, outer membrane proteins (OMPs), also participates proteases, autotransporters, and lipoproteins. The recent findings on surface structures and proteins described in this review highlight them as potential immunogens for vaccine development.
Collapse
Affiliation(s)
- María M. Soto Perezchica
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Alma L. Guerrero Barrera
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Francisco J. Avelar Gonzalez
- Laboratorio de Estudios Ambientales, Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Teodulo Quezada Tristan
- Departamento de Ciencias Veterinaria, Centro de Ciencias Agropecuarias, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Osvaldo Macias Marin
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
9
|
Sakaguchi K, Tanabe M, Takizawa S, Kasahara S, Denda T, Koide S, Hayashi W, Nagano Y, Nagano N. Zoonotic potential and antimicrobial resistance of Escherichia spp. in urban crows in Japan-first detection of E. marmotae and E. ruysiae. Comp Immunol Microbiol Infect Dis 2023; 100:102040. [PMID: 37619490 DOI: 10.1016/j.cimid.2023.102040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/26/2023]
Abstract
Little is known about the prevalence of antimicrobial-resistant bacteria and pathogenic Escherichia coli in crows (carrion and jungle crows). We studied the phylogeny, virulence and antimicrobial resistance gene profiles of crow E. coli isolates to investigate their zoonotic potential and molecular epidemiology. During the winter of 2021-2022, 34 putative E. coli isolates were recovered from 27 of the 65 fresh fecal samples collected in urban areas. Three strains of the B1-O88:H8-ST446-fimH54 lineage, classified as extraintestinal pathogenic E. coli (ExPEC) and necrotoxigenic E. coli type 2, were colistin-resistant and harbored mcr-1.1-carrying IncI2 plasmids. The blaCTX-M-55 was identified in a multidrug-resistant B1-O non-typeable:H23-ST224-fimH39 strain. In phylogroup B2, two lineages of O6:H1-ST73-fimH30 and O6:H5-ST83-fimH21 were classified as ExPEC, uropathogenic E. coli, and necrotoxigenic E. coli type 1 (O6:H5-ST83-fimH21), and contained several virulence genes associated with avian pathogenic E. coli. Noteworthy is that three isolates, identified as E. coli by MALDI-TOF MS, were confirmed to be two Escherichia marmotae (cryptic clade V) and one Escherichia ruysiae (cryptic clade III) based on ANI and dDDH analyses. Our results provide the first evidence of these new species in crows. E. marmotae and E. ruysiae isolates in this study were classified as ExPEC and contained the enteroaggregative E. coli heat-stable toxin 1 gene. In addition, these two E. marmotae isolates displayed a close genetic relationship with human isolates associated with septicemia. This study provides the first insights into the prevalence and zoonotic significance of Escherichia spp. in urban crows in Japan, posing a significant risk for their transmission to humans.
Collapse
Affiliation(s)
- Kanae Sakaguchi
- Department of Health and Medical Sciences, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Mizuki Tanabe
- Department of Health and Medical Sciences, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Shino Takizawa
- Department of Health and Medical Sciences, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Satoe Kasahara
- Suwa Hydrobiological Station, Faculty of Science, Shinshu University, 5-2-4 Kogan-dori, Suwa, Nagano 392-0027, Japan
| | - Tomohiro Denda
- Department of Health and Medical Sciences, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Shota Koide
- Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Wataru Hayashi
- Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Yukiko Nagano
- Department of Health and Medical Sciences, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Noriyuki Nagano
- Department of Health and Medical Sciences, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
10
|
Navarro-Garcia F. Serine proteases autotransporter of Enterobacteriaceae: Structures, subdomains, motifs, functions, and targets. Mol Microbiol 2023; 120:178-193. [PMID: 37392318 DOI: 10.1111/mmi.15116] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/03/2023]
Abstract
Serine protease autotransporters of Enterobacteriaceae (SPATE) constitute a superfamily of virulence factors, resembling the trypsin-like superfamily of serine proteases. SPATEs accomplish multiple functions associated to disease development of their hosts, which could be the consequence of SPATE cleavage of host cell components. SPATEs have been divided into class-1 and class-2 based on structural differences and biological effects, including similar substrate specificity, cytotoxic effects on cultured cells, and enterotoxin activity on intestinal tissues for class-1 SPATEs, whereas most class-2 SPATEs exhibit a lectin-like activity with a predilection to degrade a variety of mucins, including leukocyte surface O-glycoproteins and soluble host proteins, resulting in mucosal colonization and immune modulation. In this review, the structure of class-1 and class-2 are analyzed, making emphasis on their putative functional subdomains as well as a description of their function is provided, including prototypical mechanism of action.
Collapse
Affiliation(s)
- Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN, Mexico, Mexico
| |
Collapse
|
11
|
Liang Q, Ma C, Crowley SM, Allaire JM, Han X, Chong RWW, Packer NH, Yu HB, Vallance BA. Sialic acid plays a pivotal role in licensing Citrobacter rodentium's transition from the intestinal lumen to a mucosal adherent niche. Proc Natl Acad Sci U S A 2023; 120:e2301115120. [PMID: 37399418 PMCID: PMC10334811 DOI: 10.1073/pnas.2301115120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/30/2023] [Indexed: 07/05/2023] Open
Abstract
Enteric bacterial pathogens pose significant threats to human health; however, the mechanisms by which they infect the mammalian gut in the face of daunting host defenses and an established microbiota remain poorly defined. For the attaching and effacing (A/E) bacterial family member and murine pathogen Citrobacter rodentium, its virulence strategy likely involves metabolic adaptation to the host's intestinal luminal environment, as a necessary precursor to reach and infect the mucosal surface. Suspecting this adaptation involved the intestinal mucus layer, we found that C. rodentium was able to catabolize sialic acid, a monosaccharide derived from mucins, and utilize it as its sole carbon source for growth. Moreover, C. rodentium also sensed and displayed chemotactic activity toward sialic acid. These activities were abolished when the nanT gene, encoding a sialic acid transporter, was deleted (ΔnanT). Correspondingly, the ΔnanT C. rodentium strain was significantly impaired in its ability to colonize the murine intestine. Intriguingly, sialic acid was also found to induce the secretion of two autotransporter proteins, Pic and EspC, which possess mucinolytic and host-adherent properties. As a result, sialic acid enhanced the ability of C. rodentium to degrade intestinal mucus (through Pic), as well as to adhere to intestinal epithelial cells (through EspC). We thus demonstrate that sialic acid, a monosaccharide constituent of the intestinal mucus layer, functions as an important nutrient and a key signal for an A/E bacterial pathogen to escape the colonic lumen and directly infect its host's intestinal mucosa.
Collapse
Affiliation(s)
- Qiaochu Liang
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Caixia Ma
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Shauna M. Crowley
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Joannie M. Allaire
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Xiao Han
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Raymond W. W. Chong
- ARC Centre of Excellence for Synthetic Biology, School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, North Ryde, Sydney, NSW2109, Australia
| | - Nicolle H. Packer
- ARC Centre of Excellence for Synthetic Biology, School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, North Ryde, Sydney, NSW2109, Australia
| | - Hong Bing Yu
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| | - Bruce A. Vallance
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, BCV5Z 4H4, Canada
| |
Collapse
|
12
|
Rivera FP, Medina A, Riveros M, Ochoa TJ, Pons MJ, Ruiz J. Colonizing and Virulence Factors in Enterotoxigenic Escherichia coli from Peru. Am J Trop Med Hyg 2023; 108:948-953. [PMID: 36972692 PMCID: PMC10160877 DOI: 10.4269/ajtmh.22-0677] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/12/2023] [Indexed: 03/29/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) ranks among the most relevant diarrheagenic pathogens. Efforts to design vaccines to fight ETEC have been focused on colonizing factors (CFs) and atypical virulence factors (AVF). An effective vaccine must account for differences in the regional prevalence of these CFs and AVFs to be truly effective in a given area. In the present study, the presence of 16 CFs and 9 AVFs, as well as the heat-stable (ST) variants (STh or STp), was established by polymerase chain reaction in 205 Peruvian ETEC isolates (120 from diarrhea cases and 85 from healthy controls). Ninety-nine (48.3%) isolates were heat-labile, 63 (30.7%) ST, and 43 (21.0%) presented both toxins. Of ST isolates, 59 (28.8%) possessed STh, 30 (14.6%) STp, five (2.4%) both STh and STp, and 12 (5.8%) were not amplified for any variant tested. The presence of CFs was associated with diarrhea (P < 0.0001). The presence of eatA as well as concomitant presence of CSI, CS3, and CS21 and of C5 and C6 was statistically related to diarrhea cases. The present results suggests that, if effective, a vaccine considering CS6, CS20, and CS21, together with EtpA, would provide protection against 64.4% of the isolates analyzed, whereas the addition of CS12 and EAST1 would lead to 83.9% coverage. Large studies are needed to establish both the ideal candidates to be considered to develop a vaccine effective in the area, and continuous surveillance is needed to detect displacement of circulating isolates that may compromise future vaccines.
Collapse
Affiliation(s)
- Fulton P. Rivera
- Laboratorio de Enfermedades Entéricas, Nutrición y Resistencia Antimicrobiana, Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Ciencias de la Salud, Universidad Científica del Sur, Lima, Peru
| | - Anicia Medina
- Laboratorio de Enfermedades Entéricas, Nutrición y Resistencia Antimicrobiana, Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Maribel Riveros
- Laboratorio de Enfermedades Entéricas, Nutrición y Resistencia Antimicrobiana, Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
- Facultad de Ciencias Naturales y Matemática, Universidad Nacional Federico Villarreal, Lima, Peru
- Laboratorio de Infectología Pediátrica, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Theresa J. Ochoa
- Laboratorio de Enfermedades Entéricas, Nutrición y Resistencia Antimicrobiana, Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
- Laboratorio de Infectología Pediátrica, Universidad Peruana Cayetano Heredia, Lima, Peru
- University of Texas School of Public Health, Houston, Texas
| | - Maria J. Pons
- Grupo de Enfermedades Emergentes y Reemergentes, Universidad Científica del Sur, Lima, Peru
| | - Joaquim Ruiz
- Grupo de Enfermedades Emergentes y Reemergentes, Universidad Científica del Sur, Lima, Peru
| |
Collapse
|
13
|
Ta A, Ricci-Azevedo R, Vasudevan SO, Wright SS, Kumari P, Havira MS, Surendran Nair M, Rathinam VA, Vanaja SK. A bacterial autotransporter impairs innate immune responses by targeting the transcription factor TFE3. Nat Commun 2023; 14:2035. [PMID: 37041208 PMCID: PMC10090168 DOI: 10.1038/s41467-023-37812-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/29/2023] [Indexed: 04/13/2023] Open
Abstract
Type I interferons (IFNs) are consequential cytokines in antibacterial defense. Whether and how bacterial pathogens inhibit innate immune receptor-driven type I IFN expression remains mostly unknown. By screening a library of enterohemorrhagic Escherichia coli (EHEC) mutants, we uncovered EhaF, an uncharacterized protein, as an inhibitor of innate immune responses including IFNs. Further analyses identified EhaF as a secreted autotransporter-a type of bacterial secretion system with no known innate immune-modulatory function-that translocates into host cell cytosol and inhibit IFN response to EHEC. Mechanistically, EhaF interacts with and inhibits the MiT/TFE family transcription factor TFE3 resulting in impaired TANK phosphorylation and consequently, reduced IRF3 activation and type I IFN expression. Notably, EhaF-mediated innate immune suppression promotes EHEC colonization and pathogenesis in vivo. Overall, this study has uncovered a previously unknown autotransporter-based bacterial strategy that targets a specific transcription factor to subvert innate host defense.
Collapse
Affiliation(s)
- Atri Ta
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Rafael Ricci-Azevedo
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Swathy O Vasudevan
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Skylar S Wright
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Puja Kumari
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | | | - Meera Surendran Nair
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Sivapriya Kailasan Vanaja
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA.
| |
Collapse
|
14
|
Thunes NC, Mohammed HH, Evenhuis JP, Lipscomb RS, Pérez-Pascual D, Stevick RJ, Birkett C, Conrad RA, Ghigo JM, McBride MJ. Secreted peptidases contribute to virulence of fish pathogen Flavobacterium columnare. Front Cell Infect Microbiol 2023; 13:1093393. [PMID: 36816589 PMCID: PMC9936825 DOI: 10.3389/fcimb.2023.1093393] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Flavobacterium columnare causes columnaris disease in freshwater fish in both natural and aquaculture settings. This disease is often lethal, especially when fish population density is high, and control options such as vaccines are limited. The type IX secretion system (T9SS) is required for F. columnare virulence, but secreted virulence factors have not been fully identified. Many T9SS-secreted proteins are predicted peptidases, and peptidases are common virulence factors of other pathogens. T9SS-deficient mutants, such as ΔgldN and ΔporV, exhibit strong defects in secreted proteolytic activity. The F. columnare genome has many peptidase-encoding genes that may be involved in nutrient acquisition and/or virulence. Mutants lacking individual peptidase-encoding genes, or lacking up to ten peptidase-encoding genes, were constructed and examined for extracellular proteolytic activity, for growth defects, and for virulence in zebrafish and rainbow trout. Most of the mutants retained virulence, but a mutant lacking 10 peptidases, and a mutant lacking the single peptidase TspA exhibited decreased virulence in rainbow trout fry, suggesting that peptidases contribute to F. columnare virulence.
Collapse
Affiliation(s)
- Nicole C. Thunes
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Haitham H. Mohammed
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States,Department of Rangeland, Wildlife and Fisheries Management, Texas A&M University, College Station, TX, United States
| | - Jason P. Evenhuis
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, United States Department of Agriculture, Kearneysville, WV, United States
| | - Ryan S. Lipscomb
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, United States Department of Agriculture, Kearneysville, WV, United States
| | - David Pérez-Pascual
- Institut Pasteur, Université de Paris-Cité, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Genetics of Biofilms Laboratory, Paris, France
| | - Rebecca J. Stevick
- Institut Pasteur, Université de Paris-Cité, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Genetics of Biofilms Laboratory, Paris, France
| | - Clayton Birkett
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, United States Department of Agriculture, Kearneysville, WV, United States
| | - Rachel A. Conrad
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Jean-Marc Ghigo
- Institut Pasteur, Université de Paris-Cité, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Genetics of Biofilms Laboratory, Paris, France
| | - Mark J. McBride
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States,*Correspondence: Mark J. McBride,
| |
Collapse
|
15
|
Abstract
Proteases are an evolutionarily conserved family of enzymes that degrade peptide bonds and have been implicated in several common gastrointestinal (GI) diseases. Although luminal proteolytic activity is important for maintenance of homeostasis and health, the current review describes recent advances in our understanding of how overactivity of luminal proteases contributes to the pathophysiology of celiac disease, irritable bowel syndrome, inflammatory bowel disease and GI infections. Luminal proteases, many of which are produced by the microbiota, can modulate the immunogenicity of dietary antigens, reduce mucosal barrier function and activate pro-inflammatory and pro-nociceptive host signaling. Increased proteolytic activity has been ascribed to both increases in protease production and decreases in inhibitors of luminal proteases. With the identification of strains of bacteria that are important sources of proteases and their inhibitors, the stage is set to develop drug or microbial therapies to restore protease balance and alleviate disease.
Collapse
Affiliation(s)
- Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mabel Guzman
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen’s University, Kingston, Ontario, Canada
| | - Josie Libertucci
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Alan E. Lomax
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen’s University, Kingston, Ontario, Canada,CONTACT Alan E. Lomax Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, K7L 2V7, Canada
| |
Collapse
|
16
|
Pan X, Chen R, Zhang Y, Zhu Y, Zhao J, Yao H, Ma J. Porcine extraintestinal pathogenic Escherichia coli delivers two serine protease autotransporters coordinately optimizing the bloodstream infection. Front Cell Infect Microbiol 2023; 13:1138801. [PMID: 36875517 PMCID: PMC9978103 DOI: 10.3389/fcimb.2023.1138801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is one of the leading causes of bloodstream infections in a broad spectrum of birds and mammals, thus poses a great threat to public health, while its underlying mechanism causing sepsis is not fully understood. Here we reported a high virulent ExPEC strain PU-1, which has a robust ability to colonize within host bloodstream, while induced a low level of leukocytic activation. Two serine protease autotransporters of Enterobacteriaceae (SPATEs), VatPU-1 and TshPU-1, were found to play critical roles for the urgent blood infection of strain PU-1. Although the Vat and Tsh homologues have been identified as virulence factors of ExPEC, their contributions to bloodstream infection are still unclear. In this study, VatPU-1 and TshPU-1 were verified to interact with the hemoglobin (a well-known mucin-like glycoprotein in red blood cell), degrade the mucins of host respiratory tract, and cleave the CD43 (a major cell surface component sharing similar O-glycosylated modifications with other glycoprotein expressed on leukocytes), suggesting that these two SPATEs have the common activity to cleave a broad array of mucin-like O-glycoproteins. These cleavages significantly impaired the chemotaxis and transmigration of leukocytes, and then inhibited the activation of diverse immune responses coordinately, especially downregulated the leukocytic and inflammatory activation during bloodstream infection, thus might mediate the evasion of ExPEC from immune clearance of blood leukocytes. Taken together, these two SPATEs play critical roles to cause a heavy bacterial load within bloodstream via immunomodulation of leukocytes, which provides a more comprehensive understanding how ExPEC colonize within host bloodstream and cause severe sepsis.
Collapse
Affiliation(s)
- Xinming Pan
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
| | - Rong Chen
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
| | - Yating Zhang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
| | - Yinchu Zhu
- Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Jin Zhao
- Department of Animal Science, Yuxi Agriculture Vocation-Technical College, Yuxi, China
| | - Huochun Yao
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
| | - Jiale Ma
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Jiale Ma,
| |
Collapse
|
17
|
Alhammadi MM, Godfrey RE, Ingram JO, Singh G, Bathurst CL, Busby SJW, Browning DF. Novel organisation and regulation of the pic promoter from enteroaggregative and uropathogenic Escherichia coli. Virulence 2022; 13:1393-1406. [PMID: 35971774 PMCID: PMC9387333 DOI: 10.1080/21505594.2022.2111754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The serine protease autotransporters of the Enterobacteriaceae (SPATEs) are a large family of virulence factors commonly found in enteric bacteria. These secreted virulence factors have diverse functions during bacterial infection, including adhesion, aggregation and cell toxicity. One such SPATE, the Pic mucinase (protein involved in colonisation) cleaves mucin, allowing enteric bacterial cells to utilise mucin as a carbon source and to penetrate the gut mucus lining, thereby increasing mucosal colonisation. The pic gene is widely distributed within the Enterobacteriaceae, being found in human pathogens, such as enteroaggregative Escherichia coli (EAEC), uropathogenic E. coli (UPEC) and Shigella flexneri 2a. As the pic promoter regions from EAEC strain 042 and UPEC strain CFT073 differ, we have investigated the regulation of each promoter. Here, using in vivo and in vitro techniques, we show that both promoters are activated by the global transcription factor, CRP (cyclic AMP receptor protein), but the architectures of the EAEC and the UPEC pic promoter differ. Expression from both pic promoters is repressed by the nucleoid-associated factor, Fis, and maximal promoter activity occurs when cells are grown in minimal medium. As CRP activates transcription in conditions of nutrient depletion, whilst Fis levels are maximal in nutrient-rich environments, the regulation of the EAEC and UPEC pic promoters is consistent with Pic’s nutritional role in scavenging mucin as a suitable carbon source during colonisation and infection.
Collapse
Affiliation(s)
- Munirah M Alhammadi
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK.,Biology Department, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Rita E Godfrey
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Joseph O Ingram
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Gurdamanjit Singh
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Camilla L Bathurst
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Stephen J W Busby
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Douglas F Browning
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK.,College of Health & Life Sciences, Aston University, Birmingham, UK
| |
Collapse
|
18
|
Dubinsky V, Reshef L, Rabinowitz K, Wasserberg N, Dotan I, Gophna U. Escherichia coli Strains from Patients with Inflammatory Bowel Diseases have Disease-specific Genomic Adaptations. J Crohns Colitis 2022; 16:1584-1597. [PMID: 35560165 DOI: 10.1093/ecco-jcc/jjac071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Escherichia coli is over-abundant in the gut microbiome of patients with inflammatory bowel disease [IBD]. Here, we aimed to identify IBD-specific genomic functions of diverse E. coli lineages. METHODS We investigated E. coli genomes from patients with ulcerative colitis [UC], Crohn's disease [CD] or a pouch, and healthy subjects. The majority of genomes were reconstructed from metagenomic samples, including newly sequenced faecal metagenomes. Clinical metadata were collected. Functional analysis at the gene and mutation level were performed and integrated with IBD phenotypes and biomarkers. RESULTS Overall, 530 E. coli genomes were analysed. The E. coli B2 lineage was more prevalent in UC compared with other IBD phenotypes. Genomic metabolic capacities varied across E. coli lineages and IBD phenotypes. Host mucin utilisation enzymes were present in a single lineage and depleted in patients with a pouch, whereas those involved in inulin hydrolysis were enriched in patients with a pouch. E. coli strains from patients with UC were twice as likely to encode the genotoxic molecule colibactin than strains from patients with CD or a pouch. Strikingly, patients with a pouch showed the highest inferred E. coli growth rates, even in the presence of antibiotics. Faecal calprotectin did not correlate with the relative abundance of E. coli. Finally, we identified multiple IBD-specific non-synonymous mutations in E. coli genes encoding for bacterial cell envelope components. CONCLUSIONS Comparative genomics indicates that E. coli is a commensal species adapted to the overactive mucosal immune milieu in IBD, rather than causing it. Our results reveal mutations that may lead to attenuated antigenicity in some E. coli strains.
Collapse
Affiliation(s)
- Vadim Dubinsky
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Leah Reshef
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Keren Rabinowitz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel.,Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Nir Wasserberg
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Colorectal Unit, Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Uri Gophna
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
19
|
Clarke KR, Hor L, Pilapitiya A, Luirink J, Paxman JJ, Heras B. Phylogenetic Classification and Functional Review of Autotransporters. Front Immunol 2022; 13:921272. [PMID: 35860281 PMCID: PMC9289746 DOI: 10.3389/fimmu.2022.921272] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022] Open
Abstract
Autotransporters are the core component of a molecular nano-machine that delivers cargo proteins across the outer membrane of Gram-negative bacteria. Part of the type V secretion system, this large family of proteins play a central role in controlling bacterial interactions with their environment by promoting adhesion to surfaces, biofilm formation, host colonization and invasion as well as cytotoxicity and immunomodulation. As such, autotransporters are key facilitators of fitness and pathogenesis and enable co-operation or competition with other bacteria. Recent years have witnessed a dramatic increase in the number of autotransporter sequences reported and a steady rise in functional studies, which further link these proteins to multiple virulence phenotypes. In this review we provide an overview of our current knowledge on classical autotransporter proteins, the archetype of this protein superfamily. We also carry out a phylogenetic analysis of their functional domains and present a new classification system for this exquisitely diverse group of bacterial proteins. The sixteen phylogenetic divisions identified establish sensible relationships between well characterized autotransporters and inform structural and functional predictions of uncharacterized proteins, which may guide future research aimed at addressing multiple unanswered aspects in this group of therapeutically important bacterial factors.
Collapse
Affiliation(s)
- Kaitlin R. Clarke
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Lilian Hor
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Akila Pilapitiya
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Joen Luirink
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, Amsterdam, Netherlands
| | - Jason J. Paxman
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- *Correspondence: Begoña Heras, ; Jason J. Paxman,
| | - Begoña Heras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- *Correspondence: Begoña Heras, ; Jason J. Paxman,
| |
Collapse
|
20
|
Freire CA, Silva RM, Ruiz RC, Pimenta DC, Bryant JA, Henderson IR, Barbosa AS, Elias WP. Secreted Autotransporter Toxin (Sat) Mediates Innate Immune System Evasion. Front Immunol 2022; 13:844878. [PMID: 35251044 PMCID: PMC8891578 DOI: 10.3389/fimmu.2022.844878] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Several strategies are used by Escherichia coli to evade the host innate immune system in the blood, such as the cleavage of complement system proteins by secreted proteases. Members of the Serine Proteases Autotransporters of Enterobacteriaceae (SPATE) family have been described as presenting proteolytic effects against complement proteins. Among the SPATE-encoding genes sat (secreted autotransporter toxin) has been detected in high frequencies among strains of E. coli isolated from bacteremia. Sat has been characterized for its cytotoxic action, but the possible immunomodulatory effects of Sat have not been investigated. Therefore, this study aimed to evaluate the proteolytic effects of Sat on complement proteins and the role in pathogenesis of BSI caused by extraintestinal E. coli (ExPEC). E. coli EC071 was selected as a Sat-producing ExPEC strain. Whole-genome sequencing showed that sat sequences of EC071 and uropathogenic E. coli CFT073 present 99% identity. EC071 was shown to be resistant to the bactericidal activity of normal human serum (NHS). Purified native Sat was used in proteolytic assays with proteins of the complement system and, except for C1q, all tested substrates were cleaved by Sat in a dose and time-dependent manner. Moreover, E. coli DH5α survived in NHS pre-incubated with Sat. EC071-derivative strains harboring sat knockout and in trans complementations producing either active or non-active Sat were tested in a murine sepsis model. Lethality was reduced by 50% when mice were inoculated with the sat mutant strain. The complemented strain producing active Sat partially restored the effect caused by the wild-type strain. The results presented in this study show that Sat presents immunomodulatory effects by cleaving several proteins of the three complement system pathways. Therefore, Sat plays an important role in the establishment of bloodstream infections and sepsis.
Collapse
Affiliation(s)
- Claudia A Freire
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Rosa M Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Rita C Ruiz
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Daniel C Pimenta
- Laboratório de Bioquímica, Instituto Butantan, São Paulo, Brazil
| | - Jack A Bryant
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Ian R Henderson
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom.,Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Angela S Barbosa
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Waldir P Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| |
Collapse
|
21
|
Genotypic assay to determine some virulence factors of Uropathogenic E. coli (UPEC) isolates. Int J Health Sci (Qassim) 2022. [DOI: 10.53730/ijhs.v6ns4.6289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A total of 179 urine samples were collected from patients suffering from urinary tract infections were admitted and visit Al-Hilla General Teaching Hospital in Al-Hilla City, during a period from April 2021 to December 2021, from both sex (male and female). Out of 179,123 (68.7%) were positive culture, whereas 56 (31.3%) samples showed no bacterial growth, To confirm the identification of E. coli by use selective media (EMB agar medium, biochemical tests, automated Vitek 2 system and 16s RNA specific primer by the presence of (1492 bp) compared with allelic ladder, it was found that, E. coli were deliberated the main an etiological causes UTI to other types bacteria which constitute 56/123 (45.5%), [45/56 (80.4%) from female and 11/56 (19.6%) from male], while 67/123 (54.4%) were related to other types of bacteria. Molecular detection of some virulence factors genes were studied, out of 56 E. coli isolates, hlyA gene was detected in 21/56 (37.5%) isolates by the presence of (1177 bp) and sat gene was detected in 35/56 (62.5%) isolates by the presence of (410 bp) compared with allelic ladder.
Collapse
|
22
|
Tontanahal A, Sperandio V, Kovbasnjuk O, Loos S, Kristoffersson AC, Karpman D, Arvidsson I. IgG Binds Escherichia coli Serine Protease EspP and Protects Mice From E. coli O157:H7 Infection. Front Immunol 2022; 13:807959. [PMID: 35250980 PMCID: PMC8894809 DOI: 10.3389/fimmu.2022.807959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/31/2022] [Indexed: 12/05/2022] Open
Abstract
Shiga toxin-producing Escherichia coli O157:H7 is a virulent strain causing severe gastrointestinal infection, hemolytic uremic syndrome and death. To date there are no specific therapies to reduce progression of disease. Here we investigated the effect of pooled immunoglobulins (IgG) on the course of disease in a mouse model of intragastric E. coli O157:H7 inoculation. Intraperitoneal administration of murine IgG on day 3, or both on day 3 and 6, post-inoculation improved survival and decreased intestinal and renal pathology. When given on both day 3 and 6 post-inoculation IgG treatment also improved kidney function in infected mice. Murine and human commercially available IgG preparations bound to proteins in culture filtrates from E. coli O157:H7. Bound proteins were extracted from membranes and peptide sequences were identified by mass spectrometry. The findings showed that murine and human IgG bound to E. coli extracellular serine protease P (EspP) in the culture filtrate, via the IgG Fc domain. These results were confirmed using purified recombinant EspP and comparing culture filtrates from the wild-type E. coli O157:H7 strain to a deletion mutant lacking espP. Culture filtrates from wild-type E. coli O157:H7 exhibited enzymatic activity, specifically associated with the presence of EspP and demonstrated as pepsin cleavage, which was reduced in the presence of murine and human IgG. EspP is a virulence factor previously shown to promote colonic cell injury and the uptake of Shiga toxin by intestinal cells. The results presented here suggest that IgG binds to EspP, blocks its enzymatic activity, and protects the host from E. coli O157:H7 infection, even when given post-inoculation.
Collapse
Affiliation(s)
- Ashmita Tontanahal
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Vanessa Sperandio
- Departments of Microbiology and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olga Kovbasnjuk
- Division of Gastroenterology, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, United States
| | - Sebastian Loos
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Diana Karpman
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
- *Correspondence: Diana Karpman,
| | - Ida Arvidsson
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
23
|
Hwang IS, Oh EJ, Song E, Park IW, Lee Y, Sohn KH, Choi D, Oh CS. An Apoplastic Effector Pat-1 Cm of the Gram-Positive Bacterium Clavibacter michiganensis Acts as Both a Pathogenicity Factor and an Immunity Elicitor in Plants. FRONTIERS IN PLANT SCIENCE 2022; 13:888290. [PMID: 35432427 PMCID: PMC9006514 DOI: 10.3389/fpls.2022.888290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/14/2022] [Indexed: 05/09/2023]
Abstract
Clavibacter michiganensis, a Gram-positive plant-pathogenic bacterium, utilizes apoplastic effectors for disease development in host plants. Here, we determine the roles of Pat-1Cm (a putative serine protease) in pathogenicity and plant immunity. Pat-1Cm was found to be a genuine secreted protein, and the secreted mature form did not carry the first 33 amino acids predicted to be a signal peptide (SP). The pat-1Cm mutant impaired to cause wilting, but still caused canker symptom in tomato. Moreover, this mutant failed to trigger the hypersensitive response (HR) in a nonhost Nicotiana tabacum. Among orthologs and paralogs of pat-1Cm , only chp-7Cs from Clavibacter sepedonicus, a potato pathogen, successfully complemented pat-1Cm function in pathogenicity in tomato, whereas all failed to complement pat-1Cm function in HR induction in N. tabacum. Based on the structural prediction, Pat-1Cm carried a catalytic triad for putative serine protease, and alanine substitution of any amino acids in the triad abolished both pathogenicity and HR-inducing activities of Pat-1Cm in C. michiganensis. Ectopic expression of pat-1Cm with an SP from tobacco secreted protein triggered HR in N. tabacum, but not in tomato, whereas a catalytic triad mutant failed to induce HR. Inoculation of the pat-1Cm mutant mixed with the mutant of another apoplastic effector CelA (cellulase) caused severe wilting in tomato, indicating that these two apoplastic effectors can functionally cooperate in pathogenicity. Overall, these results indicate that Pat-1Cm is a distinct secreted protein carrying a functional catalytic triad for serine protease and this enzymatic activity might be critical for both pathogenicity and HR-eliciting activities of Pat-1Cm in plants.
Collapse
Affiliation(s)
- In Sun Hwang
- Department of Horticultural Biotechnology, College of Life Science, Kyung Hee University, Yongin, South Korea
| | - Eom-Ji Oh
- Department of Horticultural Biotechnology, College of Life Science, Kyung Hee University, Yongin, South Korea
| | - Eunbee Song
- Department of Horticultural Biotechnology, College of Life Science, Kyung Hee University, Yongin, South Korea
| | - In Woong Park
- Department of Horticultural Biotechnology, College of Life Science, Kyung Hee University, Yongin, South Korea
| | - Yoonyoung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Kee Hoon Sohn
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Doil Choi
- Department of Plant Science, Plant Immunity Research Center, Plant Genomics and Breeding Institute, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Chang-Sik Oh
- Department of Horticultural Biotechnology, College of Life Science, Kyung Hee University, Yongin, South Korea
- Graduate School of Biotechnology, Kyung Hee University, Yongin, South Korea
- *Correspondence: Chang-Sik Oh,
| |
Collapse
|
24
|
Schüroff PA, Salvador FA, Abe CM, Wami HT, Carvalho E, Hernandes RT, Dobrindt U, Gomes TAT, Elias WP. The aggregate-forming pili (AFP) mediates the aggregative adherence of a hybrid-pathogenic Escherichia coli (UPEC/EAEC) isolated from a urinary tract infection. Virulence 2021; 12:3073-3093. [PMID: 34923895 PMCID: PMC8923075 DOI: 10.1080/21505594.2021.2007645] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) comprises an important diarrheagenic pathotype, while uropathogenic E. coli (UPEC) is the most important agent of urinary tract infection (UTI). Recently, EAEC virulence factors have been detected in E. coli strains causing UTI, showing the importance of these hybrid-pathogenic strains. Previously, we detected an E. coli strain isolated from UTI (UPEC-46) presenting characteristics of EAEC, e.g., the aggregative adherence (AA) pattern and EAEC-associated genes (aatA, aap, and pet). In this current study, we analyzed the whole genomic sequence of UPEC-46 and characterized some phenotypic traits. The AA phenotype was observed in cell lineages of urinary and intestinal origin. The production of curli, cellulose, bacteriocins, and Pet toxin was detected. Additionally, UPEC-46 was not capable of forming biofilm using different culture media and human urine. The genome sequence analysis showed that this strain belongs to serotype O166:H12, ST10, and phylogroup A, harbors the tet, aadA, and dfrA/sul resistance genes, and is phylogenetically more related to EAEC strains isolated from human feces. UPEC-46 harbors three plasmids. Plasmid p46-1 (~135 kb) carries some EAEC marker genes and those encoding the aggregate-forming pili (AFP) and its regulator (afpR). A mutation in afpA (encoding the AFP major pilin) led to the loss of pilin production and assembly, and notably, a strongly reduced adhesion to epithelial cells. In summary, the genetic background and phenotypic traits analyzed suggest that UPEC-46 is a hybrid strain (UPEC/EAEC) and highlights the importance of AFP adhesin in the adherence to colorectal and bladder cell lines.
Collapse
Affiliation(s)
- Paulo A Schüroff
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil.,Institute of Hygiene, University of Münster, Münster, Germany
| | - Fábia A Salvador
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Cecilia M Abe
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Haleluya T Wami
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Eneas Carvalho
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Rodrigo T Hernandes
- Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Ulrich Dobrindt
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Tânia A T Gomes
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Waldir P Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| |
Collapse
|
25
|
Oulas A, Zachariou M, Chasapis CT, Tomazou M, Ijaz UZ, Schmartz GP, Spyrou GM, Vlamis-Gardikas A. Putative Antimicrobial Peptides Within Bacterial Proteomes Affect Bacterial Predominance: A Network Analysis Perspective. Front Microbiol 2021; 12:752674. [PMID: 34867874 PMCID: PMC8636115 DOI: 10.3389/fmicb.2021.752674] [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: 08/03/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
The predominance of bacterial taxa in the gut, was examined in view of the putative antimicrobial peptide sequences (AMPs) within their proteomes. The working assumption was that compatible bacteria would share homology and thus immunity to their putative AMPs, while competing taxa would have dissimilarities in their proteome-hidden AMPs. A network-based method ("Bacterial Wars") was developed to handle sequence similarities of predicted AMPs among UniProt-derived protein sequences from different bacterial taxa, while a resulting parameter ("Die" score) suggested which taxa would prevail in a defined microbiome. T he working hypothesis was examined by correlating the calculated Die scores, to the abundance of bacterial taxa from gut microbiomes from different states of health and disease. Eleven publicly available 16S rRNA datasets and a dataset from a full shotgun metagenomics served for the analysis. The overall conclusion was that AMPs encrypted within bacterial proteomes affected the predominance of bacterial taxa in chemospheres.
Collapse
Affiliation(s)
- Anastasis Oulas
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,The Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Margarita Zachariou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,The Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Christos T Chasapis
- NMR Center, Instrumental Analysis Laboratory, School of Natural Sciences, University of Patras, Patras, Greece
| | - Marios Tomazou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,The Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Umer Z Ijaz
- School of Engineering, University of Glasgow, Glasgow, United Kingdom
| | | | - George M Spyrou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,The Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Alexios Vlamis-Gardikas
- Division of Organic Chemistry, Biochemistry and Natural Products, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
26
|
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
|
27
|
Structural and functional significance of the amino acid differences Val 35Thr, Ser 46Ala, Asn 65Ser, and Ala 94Ser in 3C-like proteinases from SARS-CoV-2 and SARS-CoV. Int J Biol Macromol 2021; 193:2113-2120. [PMID: 34774600 PMCID: PMC8580570 DOI: 10.1016/j.ijbiomac.2021.11.043] [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: 09/04/2021] [Revised: 10/07/2021] [Accepted: 11/05/2021] [Indexed: 11/28/2022]
Abstract
Three dimensional structures of (chymo)trypsin-like proteinase (3CLpro) from SARS-CoV-2 and SARS-CoV differ at 8 positions. We previously found that the Val86Leu, Lys88Arg, Phe134His, and Asn180Lys mutations in these enzymes can change the orientation of the N- and C-terminal domains of 3CLpro relative to each other, which leads to a change in catalytic activity. This conclusion was derived from the comparison of the structural catalytic core in 169 (chymo)trypsin-like proteinases with the serine/cysteine fold. Val35Thr, Ser46Ala, Asn65Ser, Ala94Ser mutations were not included in that analysis, since they are located far from the catalytic tetrad. In the present work, the structural and functional roles of these variable amino acids at positions 35, 46, 65, and 94 in the 3CLpro sequences of SARS-CoV-2 and SARS-CoV have been established using a comparison of the same set of proteinases leading to the identification of new conservative elements. Comparative analysis showed that, in addition to interdomain mobility, which could modulate catalytic activity, the 3CLpro(s) can use for functional regulation an autolytic loop and the unique Asp33-Asn95 region (the Asp33-Asn95 Zone) in the N-terminal domain. Therefore, all 4 analyzed mutation sites are associated with the unique structure-functional features of the 3CLpro from SARS-CoV-2 and SARS-CoV. Strictly speaking, the presented structural results are hypothetical, since at present there is not a single experimental work on the identification and characterization of autolysis sites in these proteases.
Collapse
|
28
|
SepA Enhances Shigella Invasion of Epithelial Cells by Degrading Alpha-1 Antitrypsin and Producing a Neutrophil Chemoattractant. mBio 2021; 12:e0283321. [PMID: 34724811 PMCID: PMC8561385 DOI: 10.1128/mbio.02833-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Shigella spp. are highly adapted pathogens that cause bacillary dysentery in human and nonhuman primates. An unusual feature of Shigella pathogenesis is that this organism invades the colonic epithelia from the basolateral pole. Therefore, it has evolved the ability to disrupt the intestinal epithelial barrier to reach the basolateral surface. We have shown previously that the secreted serine protease A (SepA), which belongs to the family of serine protease autotransporters of Enterobacteriaceae, is responsible for the initial destabilization of the intestinal epithelial barrier that facilitates Shigella invasion. However, the mechanisms used by SepA to regulate this process remain unknown. To investigate the protein targets cleaved by SepA in the intestinal epithelium, we incubated a sample of homogenized human colon with purified SepA or with a catalytically inactive mutant of this protease. We discovered that SepA targets an array of 18 different proteins, including alpha-1 antitrypsin (AAT), a major circulating serine proteinase inhibitor in humans. In contrast to other serine proteases, SepA cleaved AAT without forming an inhibiting complex, which resulted in the generation of a neutrophil chemoattractant. We demonstrated that the products of the AAT-SepA reaction induce a mild but significant increase in neutrophil transepithelial migration in vitro. Moreover, the presence of AAT during Shigella infection stimulated neutrophil migration and dramatically enhanced the number of bacteria invading the intestinal epithelium in a SepA-dependent manner. We conclude that by cleaving AAT, SepA releases a chemoattractant that promotes neutrophil migration, which in turn disrupts the intestinal epithelial barrier to enable Shigella invasion. IMPORTANCE Shigella is the second leading cause of diarrheal death globally. In this study, we identified the host protein targets of SepA, Shigella's major protein secreted in culture. We demonstrated that by cleaving AAT, a serine protease inhibitor important to protect surrounding tissue at inflammatory sites, SepA releases a neutrophil chemoattractant that enhances Shigella invasion. Moreover, SepA degraded AAT without becoming inhibited by the cleaved product, and SepA catalytic activity was enhanced at higher concentrations of AAT. Activation of SepA by an excess of AAT may be physiologically relevant at the early stages of Shigella infection, when the amount of synthesized SepA is very low compared to the concentration of AAT in the intestinal lumen. This observation may also help to explain the adeptness of Shigella infectivity at low dose, despite the requirement of reaching the basolateral side to invade and colonize the colonic epithelium.
Collapse
|
29
|
Sudan S, Flick R, Nong L, Li J. Potential Probiotic Bacillus subtilis Isolated from a Novel Niche Exhibits Broad Range Antibacterial Activity and Causes Virulence and Metabolic Dysregulation in Enterotoxic E. coli. Microorganisms 2021; 9:1483. [PMID: 34361918 PMCID: PMC8307078 DOI: 10.3390/microorganisms9071483] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
Microbial life in extreme environments, such as deserts and deep oceans, is thought to have evolved to overcome constraints of nutrient availability, temperature, and suboptimal hygiene environments. Isolation of probiotic bacteria from such niche may provide a competitive edge over traditional probiotics. Here, we tested the survival, safety, and antimicrobial effect of a recently isolated and potential novel strain of Bacillus subtilis (CP9) from desert camel in vitro. Antimicrobial assays were performed via radial diffusion, agar spot, and co-culture assays. Cytotoxic analysis was performed using pig intestinal epithelial cells (IPEC-J2). Real time-PCR was performed for studying the effect on ETEC virulence genes and metabolomic analysis was performed using LC-MS. The results showed that CP9 cells were viable in varied bile salts and in low pH environments. CP9 showed no apparent cytotoxicity in IPEC-J2 cells. CP9 displayed significant bactericidal effect against Enterotoxic E. coli (ETEC), Salmonella Typhimurium, and Methicillin-resistant Staphylococcus aureus (MRSA) in a contact inhibitory fashion. CP9 reduced the expression of ETEC virulent genes during a 5 h co-culture. Additionally, a unique emergent metabolic signature in co-culture samples was observed by LC-MS analysis. Our findings indicate that CP9 exhibits a strong antibacterial property and reveals potential mechanisms behind.
Collapse
Affiliation(s)
- Sudhanshu Sudan
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Robert Flick
- Biozone, Mass Spectrometry and Metabolomics, Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E5, Canada;
| | - Linda Nong
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Julang Li
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| |
Collapse
|
30
|
Genome Mining and Comparative Genome Analysis Revealed Niche-Specific Genome Expansion in Antibacterial Bacillus pumilus Strain SF-4. Genes (Basel) 2021; 12:genes12071060. [PMID: 34356076 PMCID: PMC8303946 DOI: 10.3390/genes12071060] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 01/21/2023] Open
Abstract
The present study reports the isolation of antibacterial exhibiting Bacillus pumilus (B. pumilus) SF-4 from soil field. The genome of this strain SF-4 was sequenced and analyzed to acquire in-depth genomic level insight related to functional diversity, evolutionary history, and biosynthetic potential. The genome of the strain SF-4 harbor 12 Biosynthetic Gene Clusters (BGCs) including four Non-ribosomal peptide synthetases (NRPSs), two terpenes, and one each of Type III polyketide synthases (PKSs), hybrid (NRPS/PKS), lipopeptide, β-lactone, and bacteriocin clusters. Plant growth-promoting genes associated with de-nitrification, iron acquisition, phosphate solubilization, and nitrogen metabolism were also observed in the genome. Furthermore, all the available complete genomes of B. pumilus strains were used to highlight species boundaries and diverse niche adaptation strategies. Phylogenetic analyses revealed local diversification and indicate that strain SF-4 is a sister group to SAFR-032 and 150a. Pan-genome analyses of 12 targeted strains showed regions of genome plasticity which regulate function of these strains and proposed direct strain adaptations to specific habitats. The unique genome pool carries genes mostly associated with “biosynthesis of secondary metabolites, transport, and catabolism” (Q), “replication, recombination and repair” (L), and “unknown function” (S) clusters of orthologous groups (COG) categories. Moreover, a total of 952 unique genes and 168 exclusively absent genes were prioritized across the 12 genomes. While newly sequenced B. pumilus SF-4 genome consists of 520 accessory, 59 unique, and seven exclusively absent genes. The current study demonstrates genomic differences among 12 B. pumilus strains and offers comprehensive knowledge of the respective genome architecture which may assist in the agronomic application of this strain in future.
Collapse
|
31
|
Massinga AJ, Garrine M, Messa A, Nobela NA, Boisen N, Massora S, Cossa A, Varo R, Sitoe A, Hurtado JC, Ordi J, Mucavele H, Nhampossa T, Breiman RF, Whitney CG, Blau DM, Bassat Q, Mandomando I. Klebsiella spp. cause severe and fatal disease in Mozambican children: antimicrobial resistance profile and molecular characterization. BMC Infect Dis 2021; 21:526. [PMID: 34090384 PMCID: PMC8178901 DOI: 10.1186/s12879-021-06245-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/20/2021] [Indexed: 12/31/2022] Open
Abstract
Background Klebsiella spp. are important pathogens associated with bacteremia among admitted children and is among the leading cause of death in children < 5 years in postmortem studies, supporting a larger role than previously considered in childhood mortality. Herein, we compared the antimicrobial susceptibility, mechanisms of resistance, and the virulence profile of Klebsiella spp. from admitted and postmortem children. Methods Antimicrobial susceptibility and virulence factors of Klebsiella spp. recovered from blood samples collected upon admission to the hospital (n = 88) and postmortem blood (n = 23) from children < 5 years were assessed by disk diffusion and multiplex PCR. Results Klebsiella isolates from postmortem blood were likely to be ceftriaxone resistant (69.6%, 16/23 vs. 48.9%, 43/88, p = 0.045) or extended-spectrum β-lactamase (ESBL) producers (60.9%, 14/23 vs. 25%, 22/88, p = 0.001) compared to those from admitted children. blaCTX-M-15 was the most frequent ESBL gene: 65.3%, 9/14 in postmortem isolates and 22.7% (5/22) from admitted children. We found higher frequency of genes associated with hypermucoviscosity phenotype and invasin in postmortem isolates than those from admitted children: rmpA (30.4%; 7/23 vs. 9.1%, 8/88, p = 0.011), wzi-K1 (34.7%; 8/23 vs. 8%; 7/88, p = 0.002) and traT (60.8%; 14/23 vs. 10.2%; 9/88, p < 0.0001), respectively. Additionally, serine protease auto-transporters of Enterobacteriaceae were detected from 1.8% (pic) to 12.6% (pet) among all isolates. Klebsiella case fatality rate was 30.7% (23/75). Conclusion Multidrug resistant Klebsiella spp. harboring genes associated with hypermucoviscosity phenotype has emerged in Mozambique causing invasive fatal disease in children; highlighting the urgent need for prompt diagnosis, appropriate treatment and effective preventive measures for infection control. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06245-x.
Collapse
Affiliation(s)
| | - Marcelino Garrine
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa (IHMT, UNL), Lisbon, Portugal
| | - Augusto Messa
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Nélio A Nobela
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Nadia Boisen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Sergio Massora
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Anélsio Cossa
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Rosauro Varo
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - António Sitoe
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | | | - Jaume Ordi
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Hélio Mucavele
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Tacilta Nhampossa
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
| | - Robert F Breiman
- Emory Global Health Institute, Emory University, Atlanta, GA, USA
| | | | - Dianna M Blau
- Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Quique Bassat
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Inácio Mandomando
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique. .,Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique.
| |
Collapse
|
32
|
Mohebi S, Nave HH, Javadi K, Amanati A, Kholdi S, Hadadi M, Hashemizadeh Z, Motamedifar M. Evaluate the distribution of virulence genes and to investigate antibiotic resistance pattern among Shigella species isolated from children with shigellosis in Iran. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
33
|
Hou JJ, Wang X, Li Y, Su S, Wang YM, Wang BM. The relationship between gut microbiota and proteolytic activity in irritable bowel syndrome. Microb Pathog 2021; 157:104995. [PMID: 34048892 DOI: 10.1016/j.micpath.2021.104995] [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: 12/07/2020] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023]
Abstract
Irritable bowel syndrome (IBS) is a common functional gastrointestinal disease that affects 3.8-9.2% of the world population. It affects the physiology and psychology of patients and increases the burden on families, the healthcare system, society, and economic development. Presently, a large number of studies have shown that compared to healthy individuals, the composition and diversity of gut microbiota in IBS patients have changed, and the proteolytic activity (PA) in fecal supernatant and colonic mucosa of IBS patients has also increased. These findings indicate that the imbalance of intestinal microecology and intestinal protein hydrolysis is closely related to IBS. Furthermore, the intestinal flora is a key substance that regulates the PA and is associated with IBS. The current review described the intestinal microecology and intestinal proteolytic activity of patients with IBS and also discussed the effect of intestinal flora on PA. In summary, this study proposed a pivotal role of gut microbiota and PA in IBS, respectively, and provided an in-depth insight into the diagnosis and treatment targets of IBS as well as the formulation of new treatment strategies for other digestive diseases and protease-related diseases.
Collapse
Affiliation(s)
- Jun-Jie Hou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Xin Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Ying Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Shuai Su
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yu-Ming Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Bang-Mao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| |
Collapse
|
34
|
Urmi AS, Inaba H, Nomura R, Yoshida S, Ohara N, Asai F, Nakano K, Matsumoto-Nakano M. Roles of Porphyromonas gulae proteases in bacterial and host cell biology. Cell Microbiol 2021; 23:e13312. [PMID: 33486854 DOI: 10.1111/cmi.13312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 01/19/2023]
Abstract
Porphyromonas gulae, an animal-derived periodontal pathogen, expresses several virulence factors, including fimbria, lipopolysaccharide (LPS) and proteases. We previously reported that its invasive efficiency was dependent on fimbriae types. In addition, P. gulae LPS increased inflammatory responses via toll-like receptors. The present study was conducted to investigate the involvement of P. gulae proteases in bacterial and host cell biology. Porphyromonas gulae strains showed an ability to agglutinate mouse erythrocytes and also demonstrated co-aggregation with Actinomyces viscosus, while the protease inhibitors antipain, PMSF, TLCK and leupeptin diminished P. gulae proteolytic activity, resulting in inhibition of haemagglutination and co-aggregation with A. viscosus. In addition, specific proteinase inhibitors were found to reduce bacterial cell growth. Porphyromonas gulae inhibited Ca9-22 cell proliferation in a multiplicity of infection- and time-dependent manner. Additionally, P. gulae-induced decreases in cell contact and adhesion-related proteins were accompanied by a marked change in cell morphology from well spread to rounded. In contrast, inhibition of protease activity prevented degradation of proteins, such as E-cadherin, β-catenin and focal adhesion kinase, and also blocked inhibition of cell proliferation. Together, these results indicate suppression of the amount of human proteins, such as γ-globulin, fibrinogen and fibronectin, by P. gulae proteases, suggesting that a novel protease complex contributes to bacterial virulence.
Collapse
Affiliation(s)
- Alam Saki Urmi
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroaki Inaba
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ryota Nomura
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Sho Yoshida
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Naoya Ohara
- Department of Oral Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and the Advanced Research Center for Oral and Craniofacial Sciences, Dental School, Okayama University, Okayama, Japan
| | - Fumitoshi Asai
- Department of Pharmacology, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Michiyo Matsumoto-Nakano
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
35
|
Classification, structural biology, and applications of mucin domain-targeting proteases. Biochem J 2021; 478:1585-1603. [DOI: 10.1042/bcj20200607] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022]
Abstract
Epithelial surfaces throughout the body are coated by mucins, a class of proteins carrying domains characterized by a high density of O-glycosylated serine and threonine residues. The resulting mucosal layers form crucial host-microbe interfaces that prevent the translocation of microbes while also selecting for distinct bacteria via the presented glycan repertoire. The intricate interplay between mucus production and breakdown thus determines the composition of the microbiota maintained within these mucosal environments, which can have a large influence on the host during both homeostasis and disease. Most research to date on mucus breakdown has focused on glycosidases that trim glycan structures to release monosaccharides as a source of nutrients. More recent work has uncovered the existence of mucin-type O-glycosylation-dependent proteases that are secreted by pathogens, commensals, and mutualists to facilitate mucosal colonization and penetration. Additionally, immunoglobulin A (IgA) proteases promote bacterial colonization in the presence of neutralizing secretory IgA through selective cleavage of the heavily O-glycosylated hinge region. In this review, we summarize families of O-glycoproteases and IgA proteases, discuss known structural features, and review applications of these enzymes to glycobiology.
Collapse
|
36
|
Ageorges V, Monteiro R, Leroy S, Burgess CM, Pizza M, Chaucheyras-Durand F, Desvaux M. Molecular determinants of surface colonisation in diarrhoeagenic Escherichia coli (DEC): from bacterial adhesion to biofilm formation. FEMS Microbiol Rev 2021; 44:314-350. [PMID: 32239203 DOI: 10.1093/femsre/fuaa008] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/31/2020] [Indexed: 12/11/2022] Open
Abstract
Escherichia coli is primarily known as a commensal colonising the gastrointestinal tract of infants very early in life but some strains being responsible for diarrhoea, which can be especially severe in young children. Intestinal pathogenic E. coli include six pathotypes of diarrhoeagenic E. coli (DEC), namely, the (i) enterotoxigenic E. coli, (ii) enteroaggregative E. coli, (iii) enteropathogenic E. coli, (iv) enterohemorragic E. coli, (v) enteroinvasive E. coli and (vi) diffusely adherent E. coli. Prior to human infection, DEC can be found in natural environments, animal reservoirs, food processing environments and contaminated food matrices. From an ecophysiological point of view, DEC thus deal with very different biotopes and biocoenoses all along the food chain. In this context, this review focuses on the wide range of surface molecular determinants acting as surface colonisation factors (SCFs) in DEC. In the first instance, SCFs can be broadly discriminated into (i) extracellular polysaccharides, (ii) extracellular DNA and (iii) surface proteins. Surface proteins constitute the most diverse group of SCFs broadly discriminated into (i) monomeric SCFs, such as autotransporter (AT) adhesins, inverted ATs, heat-resistant agglutinins or some moonlighting proteins, (ii) oligomeric SCFs, namely, the trimeric ATs and (iii) supramolecular SCFs, including flagella and numerous pili, e.g. the injectisome, type 4 pili, curli chaperone-usher pili or conjugative pili. This review also details the gene regulatory network of these numerous SCFs at the various stages as it occurs from pre-transcriptional to post-translocational levels, which remains to be fully elucidated in many cases.
Collapse
Affiliation(s)
- Valentin Ageorges
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France
| | - Ricardo Monteiro
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France.,GSK, Via Fiorentina 1, 53100 Siena, Italy
| | - Sabine Leroy
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France
| | - Catherine M Burgess
- Food Safety Department, Teagasc Food Research Centre, Ashtown, Dublin 15, Ireland
| | | | - Frédérique Chaucheyras-Durand
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France.,Lallemand Animal Nutrition SAS, F-31702 Blagnac Cedex, France
| | - Mickaël Desvaux
- Université Clermont Auvergne, INRAE, MEDiS, F-63000 Clermont-Ferrand, France
| |
Collapse
|
37
|
Javadi K, Mohebi S, Motamedifar M, Hadi N. Characterization and antibiotic resistance pattern of diffusely adherent Escherichia coli (DAEC), isolated from paediatric diarrhoea in Shiraz, southern Iran. New Microbes New Infect 2020; 38:100780. [PMID: 33163200 PMCID: PMC7607503 DOI: 10.1016/j.nmni.2020.100780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 11/26/2022] Open
Abstract
Diarrhoea is a major health concern, especially in developing countries. Research has implicated diffusely adherent Escherichia coli (DAEC) strains as a cause of diarrhoea. In this study, we investigated the prevalence, adherence assay, virulence gene profiles and antimicrobial resistance of DAEC at a hospital in southern Iran. In this cross-sectional study, 309 infants and children under the age of 13 years with diarrhoea who had been referred to Shahid Dastgheib Hospital, Shiraz between October 2018 and May 2019 were recruited. Microbiological methods, PCR, HEp-2 adherence assay and antimicrobial susceptibility test were used. Of the 309 stool samples, 207 (66.9%) were found to contain E. coli by biochemical tests and culture. Molecular analysis of Afa/Dr and AIDA-I adhesin-encoding genes showed that 14 (6.7%) out of 207 E. coli isolates were DAEC. All DAEC isolates in HEp-2 cells showed a diffusely adherent pattern. The virulence genes sat, pet, sigA, pic, astA and fimH were found in 50%, 0%, 14.2%, 14.2%, 21.4% and 100% of DAEC isolates, respectively. The most effective antibiotic against the DAEC isolates was imipenem (92.8%) and the least effective was ampicillin (0%). Our findings expand the knowledge on DAEC prevalence and its characteristics in Iran. It also explains the role of virulence genes in DAEC pathogenesis. The results showed that although the prevalence of DAEC is low, these strains exhibit a high rate of antimicrobial resistance as well as high frequency for carrying virulence genes.
Collapse
Affiliation(s)
- K Javadi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - S Mohebi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - M Motamedifar
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Shiraz HIV/AIDS Research Centre, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - N Hadi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Bioinformatics and Computational Biology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
38
|
Rajan A, Robertson MJ, Carter HE, Poole NM, Clark JR, Green SI, Criss ZK, Zhao B, Karandikar U, Xing Y, Margalef-Català M, Jain N, Wilson RL, Bai F, Hyser JM, Petrosino J, Shroyer NF, Blutt SE, Coarfa C, Song X, Prasad BVV, Amieva MR, Grande-Allen J, Estes MK, Okhuysen PC, Maresso AW. Enteroaggregative E. coli Adherence to Human Heparan Sulfate Proteoglycans Drives Segment and Host Specific Responses to Infection. PLoS Pathog 2020; 16:e1008851. [PMID: 32986782 PMCID: PMC7553275 DOI: 10.1371/journal.ppat.1008851] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/13/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is a significant cause of acute and chronic diarrhea, foodborne outbreaks, infections of the immunocompromised, and growth stunting in children in developing nations. There is no vaccine and resistance to antibiotics is rising. Unlike related E. coli pathotypes that are often associated with acute bouts of infection, EAEC is associated with persistent diarrhea and subclinical long-term colonization. Several secreted virulence factors have been associated with EAEC pathogenesis and linked to disease in humans, less certain are the molecular drivers of adherence to the intestinal mucosa. We previously established human intestinal enteroids (HIEs) as a model system to study host-EAEC interactions and aggregative adherence fimbriae A (AafA) as a major driver of EAEC adherence to HIEs. Here, we report a large-scale assessment of the host response to EAEC adherence from all four segments of the intestine across at least three donor lines for five E. coli pathotypes. The data demonstrate that the host response in the duodenum is driven largely by the infecting pathotype, whereas the response in the colon diverges in a patient-specific manner. Major pathways altered in gene expression in each of the four enteroid segments differed dramatically, with responses observed for inflammation, apoptosis and an overwhelming response to different mucin genes. In particular, EAEC both associated with large mucus droplets and specific mucins at the epithelial surface, binding that was ameliorated when mucins were removed, a process dependent on AafA. Pan-screening for glycans for binding to purified AafA identified the human ligand as heparan sulfate proteoglycans (HSPGs). Removal of HSPG abrogated EAEC association with HIEs. These results may mean that the human intestine responds remarkably different to distinct pathobionts that is dependent on the both the individual and intestinal segment in question, and uncover a major role for surface heparan sulfate proteoglycans as tropism-driving factor in adherence and/or colonization.
Collapse
Affiliation(s)
- Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Matthew J. Robertson
- Molecular and Cell Biology-Mol. Regulation, Baylor College of Medicine, Houston, TX, United States of America
| | - Hannah E. Carter
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Nina M. Poole
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Sabrina I. Green
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Zachary K. Criss
- Department of Medicine Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States of America
| | - Boyang Zhao
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Umesh Karandikar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Yikun Xing
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Mar Margalef-Català
- Department of Pediatrics, Division of Infectious Diseases, Stanford University, Stanford, CA, United States of America
| | - Nikhil Jain
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Reid L. Wilson
- Department of Bioengineering, Rice University, Houston, TX, United States of America
| | - Fan Bai
- Department of Biochemistry, Emory Comprehensive Glycomics Core, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Joseph M. Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Joseph Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Noah F. Shroyer
- Department of Medicine Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States of America
| | - Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Cristian Coarfa
- Molecular and Cell Biology-Mol. Regulation, Baylor College of Medicine, Houston, TX, United States of America
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Xuezheng Song
- Department of Biochemistry, Emory Comprehensive Glycomics Core, Emory University School of Medicine, Atlanta, GA, United States of America
| | - BV Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Manuel R. Amieva
- Department of Pediatrics, Division of Infectious Diseases, Stanford University, Stanford, CA, United States of America
| | - Jane Grande-Allen
- Department of Bioengineering, Rice University, Houston, TX, United States of America
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Pablo C. Okhuysen
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| |
Collapse
|
39
|
The Bartonella autotransporter BafA activates the host VEGF pathway to drive angiogenesis. Nat Commun 2020; 11:3571. [PMID: 32678094 PMCID: PMC7366657 DOI: 10.1038/s41467-020-17391-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/26/2020] [Indexed: 12/29/2022] Open
Abstract
Pathogenic bacteria of the genus Bartonella can induce vasoproliferative lesions during infection. The underlying mechanisms are unclear, but involve secretion of an unidentified mitogenic factor. Here, we use functional transposon-mutant screening in Bartonella henselae to identify such factor as a pro-angiogenic autotransporter, called BafA. The passenger domain of BafA induces cell proliferation, tube formation and sprouting of microvessels, and drives angiogenesis in mice. BafA interacts with vascular endothelial growth factor (VEGF) receptor-2 and activates the downstream signaling pathway, suggesting that BafA functions as a VEGF analog. A BafA homolog from a related pathogen, Bartonella quintana, is also functional. Our work unveils the mechanistic basis of vasoproliferative lesions observed in bartonellosis, and we propose BafA as a key pathogenic factor contributing to bacterial spread and host adaptation. Pathogenic bacteria of the genus Bartonella can induce vasoproliferative lesions during infection. Here, Tsukamoto et al. show that this effect is caused by a secreted protein that induces cell proliferation and angiogenesis by acting as an analog of the host’s vascular endothelial growth factor (VEGF).
Collapse
|
40
|
Mucus layer modeling of human colonoids during infection with enteroaggragative E. coli. Sci Rep 2020; 10:10533. [PMID: 32601325 PMCID: PMC7324601 DOI: 10.1038/s41598-020-67104-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/02/2020] [Indexed: 12/25/2022] Open
Abstract
EAEC is a common cause of diarrheal illness worldwide. Pathogenesis is believed to occur in the ileum and colon, where the bacteria adhere and form a robust aggregating biofilm. Among the multiple virulence factors produced by EAEC, the Pic serine protease has been implicated in bacterial colonization by virtue of its mucinolytic activity. Hence, a potential role of Pic in mucus barrier disruption during EAEC infection has been long postulated. In this study, we used human colonoids comprising goblet cells and a thick mucin barrier as an intestinal model to investigate Pic's roles during infection with EAEC. We demonstrated the ability of purified Pic, but not a protease defective Pic mutant to degrade MUC2. Western blot and confocal microscopy analysis revealed degradation of the MUC2 layer in colonoids infected with EAEC, but not with its isogenic EAECpic mutant. Wild-type and MUC2-knockdown colonoids infected with EAEC strains exposed a differential biofilm distribution, greater penetration of the mucus layer and increased colonization of the colonic epithelium by Wild-type EAEC than its isogenic Pic mutant. Higher secretion of pro-inflammatory cytokines was seen in colonoids infected with EAEC than EAECpic. Although commensal E. coli expressing Pic degraded MUC2, it did not show improved mucus layer penetration or colonization of the colonic epithelium. Our study demonstrates a role of Pic in MUC2 barrier disruption in the human intestine and shows that colonoids are a reliable system to study the interaction of pathogens with the mucus layer.
Collapse
|
41
|
Petro CD, Duncan JK, Seldina YI, Allué-Guardia A, Eppinger M, Riddle MS, Tribble DR, Johnson RC, Dalgard CL, Sukumar G, Connor P, Boisen N, Melton-Celsa AR. Genetic and Virulence Profiles of Enteroaggregative Escherichia coli (EAEC) Isolated From Deployed Military Personnel (DMP) With Travelers' Diarrhea. Front Cell Infect Microbiol 2020; 10:200. [PMID: 32509590 PMCID: PMC7251025 DOI: 10.3389/fcimb.2020.00200] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/16/2020] [Indexed: 02/01/2023] Open
Abstract
To discern if there was a particular genotype associated with clinical enteroaggregative Escherichia coli (EAEC) strains isolated from deployed military personnel (DMP) with travelers' diarrhea (TD), we characterized a collection of EAEC from DMP deployed to Afghanistan, Djibouti, Kenya, or Honduras. Although we did not identify a specific EAEC genotype associated with TD in DMP, we found that EAEC isolated at the first clinic visit were more likely to encode the dispersin gene aap than EAEC collected at follow-up visits. A majority of the EAEC isolates were typical EAEC that adhered to HEp-2 cells, formed biofilms, and harbored genes for aggregative adherence fimbriae (AAF), AggR, and serine protease autotransporters of Enterobacteriaceae (SPATEs). A separate subset of the EAEC had aggR and genes for SPATEs but encoded a gene highly homologous to that for CS22, a fimbriae more commonly found in enterotoxigenic E. coli. None of these CS22-encoding EAEC formed biofilms in vitro or adhered to HEp-2 cells. Whole genome sequence and single nucleotide polymorphism analyses demonstrated that most of the strains were genetically diverse, but that a few were closely related. Isolation of these related strains occurred within days to more than a year apart, a finding that suggests a persistent source and genomic stability. In an ampicillin-treated mouse model we found that an agg4A+ aar- isolate formed a biofilm in the intestine and caused reduced weight gain in mice, whereas a strain that did not form an in vivo biofilm caused no morbidity. Our diverse strain collection from DMP displays the heterogeneity of EAEC strains isolated from human patients, and our mouse model of infection indicated the genotype agg4A+ aar– and/or capacity to form biofilm in vivo may correlate to disease severity.
Collapse
Affiliation(s)
- Courtney D Petro
- Department of Microbiology and Immunolgy, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Jeffrey K Duncan
- Department of Microbiology and Immunolgy, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Yuliya I Seldina
- Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Anna Allué-Guardia
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX, United States.,South Texas Center for Emerging Infectious Diseases, San Antonio, TX, United States
| | - Mark Eppinger
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX, United States.,South Texas Center for Emerging Infectious Diseases, San Antonio, TX, United States
| | - Mark S Riddle
- Department of Preventative Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - David R Tribble
- Department of Preventative Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Ryan C Johnson
- Department of Preventative Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Gauthaman Sukumar
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Collaborative Health Initiative Research Program, Henry Jackson Foundation, Bethesda, MD, United States
| | - Patrick Connor
- Military Enteric Disease Group, Academic Department of Military Medicine, Birmingham, United Kingdom
| | - Nadia Boisen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Angela R Melton-Celsa
- Department of Microbiology and Immunolgy, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
42
|
Kriaa A, Jablaoui A, Mkaouar H, Akermi N, Maguin E, Rhimi M. Serine proteases at the cutting edge of IBD: Focus on gastrointestinal inflammation. FASEB J 2020; 34:7270-7282. [PMID: 32307770 DOI: 10.1096/fj.202000031rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022]
Abstract
Serine proteases have been long recognized to coordinate many physiological processes and play key roles in regulating the inflammatory response. Accordingly, their dysregulation has been regularly associated with several inflammatory disorders and suggested as a central mechanism in the pathophysiology of digestive inflammation. So far, studies addressing the proteolytic homeostasis in the gut have mainly focused on host serine proteases as candidates of interest, while largely ignoring the potential contribution of their bacterial counterparts. The human gut microbiota comprises a complex ecosystem that contributes to host health and disease. Yet, our understanding of microbially produced serine proteases and investigation of whether they are causally linked to IBD is still in its infancy. In this review, we highlight recent advances in the emerging roles of host and bacterial serine proteases in digestive inflammation. We also discuss the application of available tools in the gut to monitor disease-related serine proteases. An exhaustive representation and understanding of such functional potential would help in closing existing gaps in mechanistic knowledge.
Collapse
Affiliation(s)
- Aicha Kriaa
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Amin Jablaoui
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Héla Mkaouar
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Nizar Akermi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Emmanuelle Maguin
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Moez Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| |
Collapse
|
43
|
Denesyuk AI, Johnson MS, Salo-Ahen OMH, Uversky VN, Denessiouk K. NBCZone: Universal three-dimensional construction of eleven amino acids near the catalytic nucleophile and base in the superfamily of (chymo)trypsin-like serine fold proteases. Int J Biol Macromol 2020; 153:399-411. [PMID: 32151723 PMCID: PMC7124590 DOI: 10.1016/j.ijbiomac.2020.03.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 10/25/2022]
Abstract
(Chymo)trypsin-like serine fold proteases belong to the serine/cysteine proteases found in eukaryotes, prokaryotes, and viruses. Their catalytic activity is carried out using a triad of amino acids, a nucleophile, a base, and an acid. For this superfamily of proteases, we propose the existence of a universal 3D structure comprising 11 amino acids near the catalytic nucleophile and base - Nucleophile-Base Catalytic Zone (NBCZone). The comparison of NBCZones among 169 eukaryotic, prokaryotic, and viral (chymo)trypsin-like proteases suggested the existence of 15 distinct groups determined by the combination of amino acids located at two "key" structure-functional positions 54T and 55T near the catalytic base His57T. Most eukaryotic and prokaryotic proteases fell into two major groups, [ST]A and TN. Usually, proteases of [ST]A group contain a disulfide bond between cysteines Cys42T and Cys58T of the NBCZone. In contrast, viral proteases were distributed among seven groups, and lack this disulfide bond. Furthermore, only the [ST]A group of eukaryotic proteases contains glycine at position 43T, which is instrumental for activation of these enzymes. In contrast, due to the side chains of residues at position 43T prokaryotic and viral proteases do not have the ability to carry out the structural transition of the eukaryotic zymogen-zyme type.
Collapse
Affiliation(s)
- Alexander I Denesyuk
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290 Pushchino, Russia; Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland.
| | - Mark S Johnson
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland
| | - Outi M H Salo-Ahen
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland; Pharmaceutical Sciences Laboratory, Pharmacy, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland
| | - Vladimir N Uversky
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290 Pushchino, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Konstantin Denessiouk
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland; Pharmaceutical Sciences Laboratory, Pharmacy, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland
| |
Collapse
|
44
|
Habouria H, Pokharel P, Maris S, Garénaux A, Bessaiah H, Houle S, Veyrier FJ, Guyomard-Rabenirina S, Talarmin A, Dozois CM. Three new serine-protease autotransporters of Enterobacteriaceae (SPATEs) from extra-intestinal pathogenic Escherichia coli and combined role of SPATEs for cytotoxicity and colonization of the mouse kidney. Virulence 2020; 10:568-587. [PMID: 31198092 PMCID: PMC6592367 DOI: 10.1080/21505594.2019.1624102] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Serine protease autotransporters of Enterobacteriaceae (SPATEs) are secreted proteins that contribute to virulence and function as proteases, toxins, adhesins, and/or immunomodulators. An extra-intestinal pathogenic E. coli (ExPEC) O1:K1 strain, QT598, isolated from a turkey, was shown to contain vat, tsh, and three uncharacterized SPATE-encoding genes. Uncharacterized SPATEs: Sha (Serine-protease hemagglutinin autotransporter), TagB and TagC (tandem autotransporter genes B and C) were tested for activities including hemagglutination, autoaggregation, and cytotoxicity when expressed in E. coli K-12. Sha and TagB conferred autoaggregation and hemagglutination activities. TagB, TagC, and Sha all exhibited cytopathic effects on a bladder epithelial cell line. In QT598, tagB and tagC are tandemly encoded on a genomic island, and were present in 10% of UTI isolates and 4.7% of avian E. coli. Sha is encoded on a virulence plasmid and was present in 1% of UTI isolates and 20% of avian E. coli. To specifically examine the role of SPATEs for infection, the 5 SPATE genes were deleted from strain QT598 and tested for cytotoxicity. Loss of all five SPATEs abrogated the cytopathic effect on bladder epithelial cells, although derivatives producing any of the 5 SPATEs retained cytopathic activity. In mouse infections, sha gene-expression was up-regulated a mean of sixfold in the bladder compared to growth in vitro. Loss of either tagBC or sha did not reduce urinary tract colonization. Deletion of all 5 SPATEs, however, significantly reduced competitive colonization of the kidney supporting a cumulative role of SPATEs for QT598 in the mouse UTI model.
Collapse
Affiliation(s)
- Hajer Habouria
- a Institut national de recherche scientifique (INRS)-Institut Armand Frappier , Laval , Quebec , Canada.,b Centre de recherche en infectiologie porcine et avicole (CRIPA)
| | - Pravil Pokharel
- a Institut national de recherche scientifique (INRS)-Institut Armand Frappier , Laval , Quebec , Canada.,b Centre de recherche en infectiologie porcine et avicole (CRIPA)
| | - Segolène Maris
- a Institut national de recherche scientifique (INRS)-Institut Armand Frappier , Laval , Quebec , Canada.,b Centre de recherche en infectiologie porcine et avicole (CRIPA)
| | - Amélie Garénaux
- a Institut national de recherche scientifique (INRS)-Institut Armand Frappier , Laval , Quebec , Canada.,b Centre de recherche en infectiologie porcine et avicole (CRIPA)
| | - Hicham Bessaiah
- a Institut national de recherche scientifique (INRS)-Institut Armand Frappier , Laval , Quebec , Canada.,b Centre de recherche en infectiologie porcine et avicole (CRIPA)
| | - Sébastien Houle
- a Institut national de recherche scientifique (INRS)-Institut Armand Frappier , Laval , Quebec , Canada.,b Centre de recherche en infectiologie porcine et avicole (CRIPA)
| | - Frédéric J Veyrier
- a Institut national de recherche scientifique (INRS)-Institut Armand Frappier , Laval , Quebec , Canada.,c Institut Pasteur International Network
| | - Stéphanie Guyomard-Rabenirina
- c Institut Pasteur International Network.,d Unité Environnement Santé , Institut Pasteur de Guadeloupe , Les Abymes , Guadeloupe , France
| | - Antoine Talarmin
- c Institut Pasteur International Network.,d Unité Environnement Santé , Institut Pasteur de Guadeloupe , Les Abymes , Guadeloupe , France
| | - Charles M Dozois
- a Institut national de recherche scientifique (INRS)-Institut Armand Frappier , Laval , Quebec , Canada.,b Centre de recherche en infectiologie porcine et avicole (CRIPA).,c Institut Pasteur International Network
| |
Collapse
|
45
|
Vieira PCG, Espinoza-Culupú AO, Nepomuceno R, Alves MR, Lebrun I, Elias WP, Ruiz RC. Secreted autotransporter toxin (Sat) induces cell damage during enteroaggregative Escherichia coli infection. PLoS One 2020; 15:e0228959. [PMID: 32084148 PMCID: PMC7034920 DOI: 10.1371/journal.pone.0228959] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 01/27/2020] [Indexed: 12/30/2022] Open
Abstract
Secreted autotransporter toxin (Sat) is a 107-kDa serine protease autotransporter of Enterobacteriaceae (SPATE) presenting cytotoxic activity in renal and bladder cells. Further studies have detected the Sat-encoding gene (sat) in enteroaggregative Escherichia coli (EAEC) and in E. coli strains isolated from neonatal septicemia and meningitis. Here, we investigated the role of Sat as a cytotoxin of EAEC. Sat was purified from a strain of E. coli harboring sat (DEC/Sat+, O126:H2) and used to raise antibodies in rabbit. The presence of Sat was detected by ELISA in the supernatant of 93.7% of EAEC strains harboring sat and in none lacking the gene. The effect of Sat during infection was investigated in polarized Caco-2 cells infected with Sat-producing EAEC (CV323/77, O125ab:H21). This strain induced intense cell detachment, which was inhibited by PMSF or Sat antiserum. Also, sat transcription and Sat production were detected during infection. Here we demonstrate that Sat is internalized in polarized cells leading to F-actin disruption which preceded cell detachment. A comparative study of the toxin action in cell lines corresponding to the infection sites in which bacteria carrying the sat gene have been isolated was performed. Cells originating from the gastrointestinal tract (Caco-2), urinary (LLC-PK1) and endothelium (HUVEC) were incubated with purified Sat. The time required for observation of cell damage differed according to the cell line. HUVEC cells were more sensitive to Sat than cells derived from urinary and intestinal tracts. The intense activity of Sat on the endothelial cells suggests that Sat could also be a virulence factor for the bacteria in the bloodstream. In addition, this is the first work demonstrating that Sat induces cytotoxic effect during EAEC infection in vitro. The cell damage observed during infection indicates that Sat may be another toxin with cytotoxic role in the EAEC pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Marina R. Alves
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP, Brazil
| | - Ivo Lebrun
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, SP, Brazil
| | - Waldir P. Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP, Brazil
| | - Rita C. Ruiz
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP, Brazil
| |
Collapse
|
46
|
Dubinsky V, Reshef L, Bar N, Keizer D, Golan N, Rabinowitz K, Godny L, Yadgar K, Zonensain K, Tulchinsky H, Gophna U, Dotan I. Predominantly Antibiotic-resistant Intestinal Microbiome Persists in Patients With Pouchitis Who Respond to Antibiotic Therapy. Gastroenterology 2020; 158:610-624.e13. [PMID: 31605691 DOI: 10.1053/j.gastro.2019.10.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 09/29/2019] [Accepted: 10/01/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Pouchitis that develops in patients with ulcerative colitis after total proctocolectomy and ileal pouch anal anastomosis is usually treated with antibiotics. Some patients have recurrence of flares, or become antibiotic-dependent, and require repeated courses or prolonged periods of antibiotic therapy. We investigated microbial factors associated with response to antibiotic treatment and development of antibiotic dependence in patients with pouchitis. METHODS We performed a prospective study of 49 patients who had undergone pouch surgery at a tertiary center. Disease activity was determined based on clinical, endoscopic, and histologic criteria. Pouch phenotype was defined as recurrent-acute pouchitis (n = 6), chronic pouchitis and Crohn's-like disease of the pouch (n = 27), normal pouch from patient with ulcerative colitis (n = 10), and normal pouch from patient with familial adenomatous polyposis (n = 6). Fecal samples (n = 234) were collected over time during or in the absence of antibiotic treatment (ciprofloxacin and/or metronidazole). Thirty-three patients were treated with antibiotics, for a median of 425 days of cumulative antibiotic therapy, during follow-up. Calprotectin was measured and fecal DNA was sequenced using shotgun metagenomics and analyzed with specifically designed bioinformatic pipelines. Bacterial strains were isolated from fecal samples. We assessed their ciprofloxacin resistance and ability to induce secretion of inflammatory cytokines by HT-29 intestinal epithelial cells. RESULTS Most antibiotic-treated patients (79%) had a clinical response to each course of antibiotics; however, 89% of those who completed a 4-week course relapsed within 3 months. Median calprotectin levels decreased by 40% in response to antibiotics. Antibiotic treatment reduced disease-associated bacteria such as Clostridium perfringens, Ruminococcus gnavus, and Klebsiella pneumoniae, but also beneficial species, such as Faecalibacterium prausnitzii. The microbiomes of antibiotic-responsive patients were dominated by facultative anaerobic genera (Escherichia, Enterococcus, and Streptococcus), with multiple ciprofloxacin-resistance mutations in drug target genes and confirmed drug resistance. However, these strains had lower potential for virulence and did not induce secretion of inflammatory cytokines by epithelial cells. After antibiotic cessation, patients had an abrupt shift in microbiome composition, with blooms of oral and disease-associated bacteria. In addition, antibiotic treatment enriched for strains that acquired multidrug resistance loci, encoding enzymes that confer resistance to nonrelated antibiotics, including extended-spectrum beta-lactamases. CONCLUSIONS The efficacy of antibiotic treatment of pouchitis might be attributed to the establishment of an antibiotic-resistant microbiome with low inflammatory potential. This microbiome might provide resistance against colonization by bacteria that promote inflammation. To avoid progression to antibiotic-dependent disease and its consequences, strategies such as short-term alternating antibiotics and nutrition- and microbiome-based interventions should be considered.
Collapse
Affiliation(s)
- Vadim Dubinsky
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University
| | - Leah Reshef
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University
| | - Nir Bar
- Department of Gastroenterology and Liver Disease, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Danielle Keizer
- The Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Felsenstein Medical Research Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Noam Golan
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University
| | - Keren Rabinowitz
- The Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Felsenstein Medical Research Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Lihi Godny
- The Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Felsenstein Medical Research Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Karin Yadgar
- The Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Felsenstein Medical Research Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Keren Zonensain
- The Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Felsenstein Medical Research Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Hagit Tulchinsky
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Proctology Unit, Division of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Uri Gophna
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University.
| | - Iris Dotan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel.
| |
Collapse
|
47
|
Freire CA, Santos ACM, Pignatari AC, Silva RM, Elias WP. Serine protease autotransporters of Enterobacteriaceae (SPATEs) are largely distributed among Escherichia coli isolated from the bloodstream. Braz J Microbiol 2020; 51:447-454. [PMID: 31965549 DOI: 10.1007/s42770-020-00224-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/07/2020] [Indexed: 12/29/2022] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is the major cause of Gram-negative-related sepsis. Bacterial survival in the bloodstream is mediated by a variety of virulence traits, including those mediating immune system evasion. Serine protease autotransporters of Enterobacteriaceae (SPATE) constitute a superfamily of virulence factors that can cause tissue damage and cleavage of molecules of the complement system, which is a key feature for the establishment of infection in the bloodstream. In this study, we analyzed 278 E. coli strains isolated from human bacteremia from inpatients of both genders, different ages, and clinical conditions. These strains were screened for the presence of SPATE-encoding genes as well as for phylogenetic classification and intrinsic virulence of ExPEC. SPATE-encoding genes were detected in 61.2% of the strains and most of these strains (44.6%) presented distinct SPATE-encoding gene profiles. sat was the most frequent gene among the entire collection, found in 34.2%, followed by vat (28.4%), pic (8.3%), and tsh (4.7%). Although in low frequencies, espC (0.7%), eatA (1.1%), and espI (1.1%) were detected and are being reported for the first time in extraintestinal isolates. The presence of SPATE-encoding genes was positively associated to phylogroup B2 and intrinsic virulent strains. These findings suggest that SPATEs are highly prevalent and involved in diverse steps of the pathogenesis of bacteremia caused by E. coli.
Collapse
Affiliation(s)
- Claudia A Freire
- Laboratório de Bacteriologia, Instituto Butantan, Avenida Vital Brazil 1500, São Paulo, SP, 05503-900, Brazil
| | - Ana Carolina M Santos
- Disciplina de Microbiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Antonio C Pignatari
- Laboratório Especial de Microbiologia Clínica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Rosa M Silva
- Disciplina de Microbiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Waldir P Elias
- Laboratório de Bacteriologia, Instituto Butantan, Avenida Vital Brazil 1500, São Paulo, SP, 05503-900, Brazil.
| |
Collapse
|
48
|
Yang Y, Sun J, Sun Y, Kwan YH, Wong WC, Zhang Y, Xu T, Feng D, Zhang Y, Qiu JW, Qian PY. Genomic, transcriptomic, and proteomic insights into the symbiosis of deep-sea tubeworm holobionts. THE ISME JOURNAL 2020; 14:135-150. [PMID: 31595051 PMCID: PMC6908572 DOI: 10.1038/s41396-019-0520-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/11/2019] [Accepted: 08/25/2019] [Indexed: 12/22/2022]
Abstract
Deep-sea hydrothermal vents and methane seeps are often densely populated by animals that host chemosynthetic symbiotic bacteria, but the molecular mechanisms of such host-symbiont relationship remain largely unclear. We characterized the symbiont genome of the seep-living siboglinid Paraescarpia echinospica and compared seven siboglinid-symbiont genomes. Our comparative analyses indicate that seep-living siboglinid endosymbionts have more virulence traits for establishing infections and modulating host-bacterium interaction than the vent-dwelling species, and have a high potential to resist environmental hazards. Metatranscriptome and metaproteome analyses of the Paraescarpia holobiont reveal that the symbiont is highly versatile in its energy use and efficient in carbon fixation. There is close cooperation within the holobiont in production and supply of nutrients, and the symbiont may be able to obtain nutrients from host cells using virulence factors. Moreover, the symbiont is speculated to have evolved strategies to mediate host protective immunity, resulting in weak expression of host innate immunity genes in the trophosome. Overall, our results reveal the interdependence of the tubeworm holobiont through mutual nutrient supply, a pathogen-type regulatory mechanism, and host-symbiont cooperation in energy utilization and nutrient production, which is a key adaptation allowing the tubeworm to thrive in deep-sea chemosynthetic environments.
Collapse
Affiliation(s)
- Yi Yang
- Department of Ocean Science, Division of Life Science and Hong Kong Branch of The Southern Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jin Sun
- Department of Ocean Science, Division of Life Science and Hong Kong Branch of The Southern Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yanan Sun
- Department of Ocean Science, Division of Life Science and Hong Kong Branch of The Southern Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yick Hang Kwan
- Department of Ocean Science, Division of Life Science and Hong Kong Branch of The Southern Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Wai Chuen Wong
- Department of Ocean Science, Division of Life Science and Hong Kong Branch of The Southern Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yanjie Zhang
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Ting Xu
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Dong Feng
- CAS Key Laboratory of Ocean and Marginal Sea Geology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301, Guangzhou, China
- Laboratory for Marine Geology, Qingdao National Laboratory for Marine Science and Technology, 266061, Qingdao, China
| | - Yu Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jian-Wen Qiu
- Department of Biology, Hong Kong Baptist University, Hong Kong, China.
| | - Pei-Yuan Qian
- Department of Ocean Science, Division of Life Science and Hong Kong Branch of The Southern Science and Engineering Guangdong Laboratory, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
49
|
Pokharel P, Habouria H, Bessaiah H, Dozois CM. Serine Protease Autotransporters of the Enterobacteriaceae (SPATEs): Out and About and Chopping It Up. Microorganisms 2019; 7:E594. [PMID: 31766493 PMCID: PMC6956023 DOI: 10.3390/microorganisms7120594] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Autotransporters are secreted proteins with multiple functions produced by a variety of Gram-negative bacteria. In Enterobacteriaceae, a subgroup of these autotransporters are the SPATEs (serine protease autotransporters of Enterobacteriaceae). SPATEs play a crucial role in survival and virulence of pathogens such as Escherichia coli and Shigella spp. and contribute to intestinal and extra-intestinal infections. These high molecular weight proteases are transported to the external milieu by the type Va secretion system and function as proteases with diverse substrate specificities and biological functions including adherence and cytotoxicity. Herein, we provide an overview of SPATEs and discuss recent findings on the biological roles of these secreted proteins, including proteolysis of substrates, adherence to cells, modulation of the immune response, and virulence in host models. In closing, we highlight recent insights into the regulation of expression of SPATEs that could be exploited to understand fundamental SPATE biology.
Collapse
Affiliation(s)
- Pravil Pokharel
- Institut National de Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada; (P.P.); (H.H.); (H.B.)
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Hajer Habouria
- Institut National de Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada; (P.P.); (H.H.); (H.B.)
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Hicham Bessaiah
- Institut National de Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada; (P.P.); (H.H.); (H.B.)
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Charles M. Dozois
- Institut National de Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada; (P.P.); (H.H.); (H.B.)
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Saint-Hyacinthe, QC J2S 2M2, Canada
- Institut Pasteur International Network, Laval, QC H7V 1B7, Canada
| |
Collapse
|
50
|
Guerrieri CG, Pereira MF, Galdino ACM, Dos Santos ALS, Elias WP, Schuenck RP, Spano LC. Typical and Atypical Enteroaggregative Escherichia coli Are Both Virulent in the Galleria mellonella Model. Front Microbiol 2019; 10:1791. [PMID: 31456762 PMCID: PMC6700222 DOI: 10.3389/fmicb.2019.01791] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/19/2019] [Indexed: 12/16/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is an emerging pathotype responsible for acute and persistent diarrhea. It can be classified as typical and atypical strains, respectively, based on the presence or absence of the AggR regulon, suggesting a higher virulence for typical EAEC. This study aims to evaluate in the Galleria mellonella model if there are differences in the virulence profiles among clinical strains of typical and atypical EAEC, prototype strains EAEC C1096, 042 and its aggR mutant. The clinical EAEC strains (n = 20) were analyzed for the presence of 22 putative virulence factors of EAEC or extraintestinal E. coli by PCR, as well as phenotypic characteristics of virulence (enzymes, siderophore, and biofilm). The survival of the larvae was analyzed after inoculation of 104-107 CFU/larva; the monitoring of bacterial growth in vivo and hemocyte quantification was determined after inoculation of the prototype strains (105 CFU/larva) at different periods after infection. The strains of typical and atypical EAEC presented the same virulence profile for the larva, regardless of the amount or type of genes and phenotypic aspects of virulence analyzed. In addition, the EAEC 042 aggR mutant strain showed a significant reduction in the mortality of the inoculated larvae compared to the wild-type strain. In conclusion, the results obtained herein demonstrate that the virulence of EAEC seems to be related to the AggR regulon, but not exclusively, and atypical EAEC strains may be as virulent as typical ones in vivo in the G. mellonella model.
Collapse
Affiliation(s)
- Caroline Gastaldi Guerrieri
- Laboratory of Virology and Infectious Gastroenteritis, Department of Pathology, Health Sciences Center, Federal University of Espírito Santo, Vitória, Brazil
| | - Monalessa Fábia Pereira
- Laboratory of Virology and Infectious Gastroenteritis, Department of Pathology, Health Sciences Center, Federal University of Espírito Santo, Vitória, Brazil
| | - Anna Clara Milesi Galdino
- Laboratory of Advanced Studies of Emerging and Resistant Microorganisms, Department of General Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Luis Souza Dos Santos
- Laboratory of Advanced Studies of Emerging and Resistant Microorganisms, Department of General Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ricardo Pinto Schuenck
- Laboratory of Virology and Infectious Gastroenteritis, Department of Pathology, Health Sciences Center, Federal University of Espírito Santo, Vitória, Brazil
| | - Liliana Cruz Spano
- Laboratory of Virology and Infectious Gastroenteritis, Department of Pathology, Health Sciences Center, Federal University of Espírito Santo, Vitória, Brazil
| |
Collapse
|