1
|
Xu Z, Zhang Y, Wang Y, Wu A, Meng C, Li W, Yi J, Chen C. Evaluation of the safety and immune protection of OMPAC, PAPF, and EBPSs recombinant subunit vaccines Developed for Escherichia coli, Staphylococcus aureus, and Streptococcus agalactiae in mice. Int Immunopharmacol 2025; 148:114151. [PMID: 39874844 DOI: 10.1016/j.intimp.2025.114151] [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: 07/12/2024] [Revised: 10/30/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
Bacterial mastitis in dairy cow is often caused by a combination of bacterial infections, such as Escherichia coli, Staphylococcus aureus, and Streptococcus agalactiae. Currently, there is no effective vaccine against the disease. Therefore, we constructed a recombinant subunit vaccine by fusing gene fragments of E. coli OMPA and OMPC, S. aureus EBPS, and S. agalactiae PGK, AP1, AP2, and FBSA. These gene fragments were combined into three fusion proteins: OMPAC, EBPSs, and PAPF. Mice were immunized with the three fusion proteins either alone or in combination. The test results showed that immunization with OMPAC, EBPSs, and PAPF individually or in combination could induce high titers of antibodies in the mice. Additionally, 21 days post-immunization, IFN-γ levels were significantly increased in all groups of mice, suggesting that immunization with OMPAC, EBPSs, and PAPF, whether alone or in combination, was effective in inducing antibody production. This indicates that OMPAC, EBPSs, and PAPF were effective in inducing both humoral and cellular immunity in mice. Furthermore, immunization with OMPAC, EBPSs, and PAPF individually or in combination were effective in protecting mice from E. coli, S. aureus, and S. agalactiae infections. Importantly, a mixture of the three fusion proteins was relatively safe for pregnant female mice. In conclusion, we successfully constructed and expressed recombinant subunit vaccines of OMPAC, EBPSs and PAPF and verified that these vaccines rapidly induced high levels of specific antibodies while reducing bacterial loads in the organs of mice. This lays the theoretical foundation and data support for the development of novel subunit vaccines against mastitis in dairy cows.
Collapse
Affiliation(s)
- Zhenyu Xu
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China
| | - Yuchen Zhang
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China
| | - Yueli Wang
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China
| | - Aodi Wu
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China
| | - Chuang Meng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Wei Li
- Xinjiang Center for Animal Disease Control and Prevention, Urumqi, China
| | - Jihai Yi
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China.
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China.
| |
Collapse
|
2
|
Ntozini B, Walaza S, Metcalf B, Hazelhurst S, de Gouveia L, Meiring S, Mogale D, Mtshali S, Ismail A, Ndlangisa K, Du Plessis M, Quan V, Chochua S, McGee L, von Gottberg A, Wolter N. Molecular epidemiology of invasive group B Streptococcus in South Africa, 2019-2020. J Infect Dis 2024:jiae633. [PMID: 39737783 DOI: 10.1093/infdis/jiae633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Group B Streptococcus (GBS) is a leading cause of neonatal meningitis and sepsis and an important cause of disease in adults. Capsular polysaccharide and protein-based GBS vaccines are currently under development. METHODS Through national laboratory-based surveillance, invasive GBS isolates were collected from patients of all ages between 2019 and 2020. Phenotypic serotyping and antimicrobial susceptibility testing were conducted, followed by whole-genome sequencing for analysis of population structure and surface protein and resistance genes. RESULTS 1748 invasive GBS cases were reported. Of these, 661 isolates underwent characterization, with 658 yielding both phenotypic and genotypic results. Isolates (n=658) belonged to five clonal complexes (CC1, CC8/10, CC17, CC19, and CC23) and six serotypes were detected: III (42.8%), Ia (27.9%), V (11.9%), II (8.4%), Ib (6.7%), and IV (2.3%). Phenotypically, only one isolate exhibited reduced penicillin susceptibility (MIC 0.25ug/ml). Phenotypic resistance to erythromycin, clindamycin, and tetracycline was observed in 16.1%, 3.8%, and 91.5% of isolates, respectively. ermTR (34.9%) and mefA/E (30.1%) genes were most common among erythromycin-resistant isolates, while ermB predominated clindamycin-resistant isolates (32.0%). tetM accounted for 95.8% of tetracycline resistance. All isolates carried at least one of the three pilus gene clusters, one of the four homologous alpha/Rib family determinants, and 98% harbored one of the serine-rich repeat protein genes. hvgA was found exclusively in CC17 isolates. CONCLUSION In our setting, β-lactam antibiotics remain appropriate for GBS treatment and polysaccharide and protein-based vaccines under development are expected to provide good coverage.
Collapse
Affiliation(s)
- Buhle Ntozini
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sibongile Walaza
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Benjamin Metcalf
- Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Scott Hazelhurst
- School of Electrical & Information Engineering, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg. South Africa
| | - Linda de Gouveia
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Susan Meiring
- Division of Public Health Surveillance and Response, National Institute for Communicable Diseases a division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Dineo Mogale
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Senzo Mtshali
- Sequencing Core Facility, National Institute for Communicable Diseases a division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Arshad Ismail
- Sequencing Core Facility, National Institute for Communicable Diseases a division of the National Health Laboratory Service, Johannesburg, South Africa
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Thohoyandou, South Africa
- Institute for Water and Wastewater Technology, Durban University of Technology, Durban, South Africa
| | - Kedibone Ndlangisa
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mignon Du Plessis
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Vanessa Quan
- Division of Public Health Surveillance and Response, National Institute for Communicable Diseases a division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Sopio Chochua
- Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Lesley McGee
- Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Anne von Gottberg
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nicole Wolter
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
3
|
Gent V, Lu YJ, Lukhele S, Dhar N, Dangor Z, Hosken N, Malley R, Madhi SA, Kwatra G. Surface protein distribution in Group B Streptococcus isolates from South Africa and identifying vaccine targets through in silico analysis. Sci Rep 2024; 14:22665. [PMID: 39349584 PMCID: PMC11442663 DOI: 10.1038/s41598-024-73175-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Group B Streptococcus (GBS) is a major cause of pneumonia, sepsis, and meningitis in infants younger than 3 months of age. Furthermore, GBS infection in pregnant women is associated with stillbirths and pre-term delivery. It also causes disease in immunocompromised adults and the elderly, but the highest incidence of the disease occurs in neonates and young infants. At this time, there are no licensed vaccines against GBS. Complete GBS genome sequencing has helped identify genetically conserved and immunogenic proteins, which could serve as vaccine immunogens. In this study, in silico reverse vaccinology method were used to evaluate the prevalence and conservation of GBS proteins in invasive and colonizing isolates from South African infants and women, respectively. Furthermore, this study aimed to predict potential GBS vaccine targets by evaluating metrics such as antigenicity, physico-chemical properties, subcellular localization, secondary and tertiary structures, and epitope prediction and conservation. A total of 648 invasive and 603 colonizing GBS isolate sequences were screened against a panel of 89 candidate GBS proteins. Ten of the 89 proteins were highly genetically conserved in invasive and colonizing GBS isolates, nine of which were computationally inferred proteins (gbs2106, SAN_1577, SAN_0356, SAN_1808, SAN_1685, SAN_0413, SAN_0990, SAN_1040, SAN_0226) and one was the surface Immunogenic Protein (SIP). Additionally, the nine proteins were predicted to be more antigenic than the SIP protein (antigenicity score of > 0.6498), highlighting their potential as GBS vaccine antigen targets.
Collapse
Affiliation(s)
- Vicky Gent
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ying-Jie Lu
- Division of Infectious Diseases, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sindiswa Lukhele
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nisha Dhar
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ziyaad Dangor
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nancy Hosken
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Richard Malley
- Division of Infectious Diseases, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shabir A Madhi
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gaurav Kwatra
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Department of Clinical Microbiology, Christian Medical College, Vellore, India.
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
4
|
Rajack F, Medford S, Ramadan A, Naab T. Emerging infection: streptococcal toxic shock-like syndrome caused by group B Streptococcus (GBS), Streptococcus agalactiae. Autops Case Rep 2024; 14:e2024497. [PMID: 39021470 PMCID: PMC11253910 DOI: 10.4322/acr.2024.497] [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: 09/26/2023] [Accepted: 04/25/2024] [Indexed: 07/20/2024]
Abstract
Streptococcus agalactiae or Group B Streptococcus (GBS) infections are commonly associated with infections in neonates and pregnant women. However, there has been a rising incidence in nonpregnant adults. The risk of GBS infection in nonpregnant adults is increased for patients of advanced age and those with underlying medical conditions such as diabetes mellitus and cancer. We present a 77-year-old female with type-2 diabetes mellitus, hypertension, and bilateral foot ulcers that presented in probable septic shock with necrotic foot ulcers and necrotizing fasciitis and underwent bilateral lower limb amputations. The patient fulfilled the Streptococcal Toxic Shock Syndrome (STSS) criteria as defined by The Working Group on Severe Streptococcal Infections. These criteria were created for group A Streptococcus (Streptococcus pyogenes). Our patient fulfilled the Working Group's criteria, except that the blood culture was positive for group B Streptococcus (Streptococcus agalactiae). Numerous studies demonstrate the importance of early detection and antibiotic treatment for GBS infections in general and early surgical management for necrotizing soft tissue infections (NSTIs) such as necrotizing fasciitis.
Collapse
Affiliation(s)
- Fareed Rajack
- Howard University Hospital, Department of Pathology and Laboratory Medicine, Washington, D.C., United States of America
| | - Shawn Medford
- Howard University College of Medicine, Washington, D.C., United States of America
| | - Ali Ramadan
- Howard University Hospital, Department of Pathology and Laboratory Medicine, Washington, D.C., United States of America
| | - Tammey Naab
- Howard University Hospital, Department of Pathology and Laboratory Medicine, Washington, D.C., United States of America
| |
Collapse
|
5
|
Goh KGK, Desai D, Thapa R, Prince D, Acharya D, Sullivan MJ, Ulett GC. An opportunistic pathogen under stress: how Group B Streptococcus responds to cytotoxic reactive species and conditions of metal ion imbalance to survive. FEMS Microbiol Rev 2024; 48:fuae009. [PMID: 38678005 PMCID: PMC11098048 DOI: 10.1093/femsre/fuae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024] Open
Abstract
Group B Streptococcus (GBS; also known as Streptococcus agalactiae) is an opportunistic bacterial pathogen that causes sepsis, meningitis, pneumonia, and skin and soft tissue infections in neonates and healthy or immunocompromised adults. GBS is well-adapted to survive in humans due to a plethora of virulence mechanisms that afford responses to support bacterial survival in dynamic host environments. These mechanisms and responses include counteraction of cell death from exposure to excess metal ions that can cause mismetallation and cytotoxicity, and strategies to combat molecules such as reactive oxygen and nitrogen species that are generated as part of innate host defence. Cytotoxicity from reactive molecules can stem from damage to proteins, DNA, and membrane lipids, potentially leading to bacterial cell death inside phagocytic cells or within extracellular spaces within the host. Deciphering the ways in which GBS responds to the stress of cytotoxic reactive molecules within the host will benefit the development of novel therapeutic and preventative strategies to manage the burden of GBS disease. This review summarizes knowledge of GBS carriage in humans and the mechanisms used by the bacteria to circumvent killing by these important elements of host immune defence: oxidative stress, nitrosative stress, and stress from metal ion intoxication/mismetallation.
Collapse
Affiliation(s)
- Kelvin G K Goh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Devika Desai
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Ruby Thapa
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Darren Prince
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Dhruba Acharya
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Matthew J Sullivan
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Glen C Ulett
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| |
Collapse
|
6
|
Huang J, Dai X, Wu Z, Hu X, Sun J, Tang Y, Zhang W, Han P, Zhao J, Liu G, Wang X, Mao S, Wang Y, Call DR, Liu J, Wang L. Conjugative transfer of streptococcal prophages harboring antibiotic resistance and virulence genes. THE ISME JOURNAL 2023; 17:1467-1481. [PMID: 37369704 PMCID: PMC10432423 DOI: 10.1038/s41396-023-01463-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Prophages play important roles in the transduction of various functional traits, including virulence factors, but remain debatable in harboring and transmitting antimicrobial resistance genes (ARGs). Herein we characterize a prevalent family of prophages in Streptococcus, designated SMphages, which harbor twenty-five ARGs that collectively confer resistance to ten antimicrobial classes, including vanG-type vancomycin resistance locus and oxazolidinone resistance gene optrA. SMphages integrate into four chromosome attachment sites by utilizing three types of integration modules and undergo excision in response to phage induction. Moreover, we characterize four subtypes of Alp-related surface proteins within SMphages, the lethal effects of which are extensively validated in cell and animal models. SMphages transfer via high-frequency conjugation that is facilitated by integrative and conjugative elements from either donors or recipients. Our findings explain the widespread of SMphages and the rapid dissemination of ARGs observed in members of the Streptococcus genus.
Collapse
Affiliation(s)
- Jinhu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xingyang Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zuowei Wu
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, 50011, USA
| | - Xiao Hu
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, 50011, USA
| | - Junjie Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yijun Tang
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wanqiu Zhang
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Peizhao Han
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jiaqi Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Guangjin Liu
- Key Lab of Animal Bacteriology, Ministry of Agriculture, OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaoming Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Shengyong Mao
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yang Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Douglas R Call
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, 99164, USA
| | - Jinxin Liu
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
7
|
Yanni M, Stark M, Francis L, Francis JR, McMillan M, Baird R, Heath PT, Gordon A, Riccardione J, Wilson A, Lee R, Chooi K, Quinn OP, Marshall HS. Neonatal Group B Streptococcal Infection in Australia: A Case-control Study. Pediatr Infect Dis J 2023; 42:429-435. [PMID: 36929884 DOI: 10.1097/inf.0000000000003881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
BACKGROUND To determine maternal and neonatal risk factors for, and incidence of, neonatal early-onset group B streptococcus (EOGBS) and late-onset (LOGBS) infection in South Australia (SA) and the Northern Territory (NT). METHODS A case-control study with 2:1 matched controls to cases. The study included tertiary hospitals in South Australia and the Northern Territory, Australia. Retrospective data were collected from a 16-year epoch (2000-2015). RESULTS Of a total of 188 clinically suspected or confirmed cases, 139 were confirmed, of which 56.1% (n = 78) were EOGBS and 43.9% (n = 61) were LOGBS. The incidence of clinically suspected and confirmed cases of EOGBS was 0.26/1000 live births in SA and 0.73/1000 live births in the NT, and the incidence of confirmed cases was 0.19/1000 for SA and 0.36/1000 for the NT. The incidence of clinically suspected or confirmed LOGBS was 0.18/1000 live births in SA and 0.16/1000 for the NT. The majority of infants with GBS presented with sepsis, pneumonia, or meningitis. Developmental delay was the most commonly recorded long-term complication at 1 year old. Risk factors for EOGBS included maternal GBS carriage, previous fetal death, identifying as Aboriginal and/or Torres Strait Islander, and maternal fever in labor/chorioamnionitis. CONCLUSIONS GBS remains a leading cause of neonatal morbidity and mortality. Adding previous fetal death to GBS screening guidelines would improve GBS prevention. The introduction of maternal GBS vaccination programs should be guided by country-specific disease epidemiology.
Collapse
Affiliation(s)
- Marianne Yanni
- From the Department of Paediatrics, Women's and Children's Health Network, Adelaide, South Australia, Australia
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Michael Stark
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Neonatology, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Laura Francis
- Department of Paediatrics, Royal Darwin Hospital, Tiwi, Northern Territory, Australia
| | - Joshua R Francis
- Department of Paediatrics, Royal Darwin Hospital, Tiwi, Northern Territory, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Casuarina, Northern Territory, Australia
| | - Mark McMillan
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Rob Baird
- Territory Pathology, Royal Darwin Hospital, Tiwi, Northern Territory, Australia
| | - Paul T Heath
- Paediatric Infectious Diseases Research Group & Vaccine Institute St George's, University of London, London, United Kingdom
| | - Alex Gordon
- From the Department of Paediatrics, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - James Riccardione
- Territory Pathology, Royal Darwin Hospital, Tiwi, Northern Territory, Australia
| | - Angela Wilson
- Territory Pathology, Royal Darwin Hospital, Tiwi, Northern Territory, Australia
| | - Rebecca Lee
- Department of Paediatrics, Royal Darwin Hospital, Tiwi, Northern Territory, Australia
| | - Kathrina Chooi
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Olivia-Paris Quinn
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Helen S Marshall
- From the Department of Paediatrics, Women's and Children's Health Network, Adelaide, South Australia, Australia
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Health Network, Adelaide, South Australia, Australia
| |
Collapse
|
8
|
Dangor Z, Kwatra G, Pawlowski A, Fisher PB, Izu A, Lala SG, Johansson-Lindbom B, Madhi SA. Association of infant Rib and Alp1 surface protein N-terminal domain immunoglobulin G and invasive Group B Streptococcal disease in young infants. Vaccine 2023; 41:1679-1683. [PMID: 36754766 PMCID: PMC9996286 DOI: 10.1016/j.vaccine.2023.01.071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND Vaccine development for Group B Streptococcus (GBS), a common cause of invasive disease in early-infancy and adverse pregnancy outcomes, include exploring widely-expressed GBS surface proteins as vaccine epitopes. We investigated the association between natural infant serum IgG against the RibN and Alp1N domains and risk of invasive GBS disease caused by isolates expressing these proteins. METHODS We analyzed maternal and infant serum samples from GBS disease cases and infants born to GBS-colonized women controls. Bayesian modelling was used to calculate the GBS homotypic IgG concentration associated with risk reduction of invasive disease in the infant. RESULTS PCR-based typing of 85 GBS invasive isolates showed 46 and 24 possessing the gene for Rib and Alp1, respectively. These were matched to 46 and 36 infant controls whose mothers were colonized with GBS expressing Rib and Alp1, respectively. RibN IgG geometric mean concentrations (GMC) were lower in cases than controls among infants (0.01; 95 %CI: 0.01-0.02 vs 0.04; 95 %CI: 0.03-0.06; p < 0.001), no significant difference was found between maternal RibN IgG GMC in cases compared to controls. Alp1N IgG GMC was also lower in infant cases (0.02; 95 %CI: 0.01-0.03) than controls (0.05; 95 %CI: 0.04-0.07; p < 0.001); albeit not so in mothers. An infant IgG threshold ≥ 0.428 and ≥ 0.112 µg/mL was associated with 90 % risk reduction of invasive GBS disease due to Rib and Alp1 expressing strains, respectively. DISCUSSION Lower serum RibN and Alp1N IgG GMC were evident in infants with invasive GBS disease compared with controls born to women colonized with GBS expressing the homotypic protein. These data support the evaluation of Alp family proteins as potential vaccine candidates against invasive GBS disease.
Collapse
Affiliation(s)
- Ziyaad Dangor
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Unit, University of the Witwatersrand, South Africa; Department of Paediatrics & Child Health, Faculty of Health Sciences, University of the Witwatersrand, South Africa.
| | - Gaurav Kwatra
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Unit, University of the Witwatersrand, South Africa; Department of Clinical Microbiology, Christian Medical College, Vellore, India
| | | | | | - Alane Izu
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Unit, University of the Witwatersrand, South Africa
| | - Sanjay G Lala
- Department of Paediatrics & Child Health, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - Bengt Johansson-Lindbom
- Immunology Section, BMC D14, Lund University, Lund, Sweden; MinervaX ApS, DK-2200 Copenhagen N, Denmark
| | - Shabir A Madhi
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Unit, University of the Witwatersrand, South Africa; African Leadership in Vaccinology Expertise University of the Witwatersrand, South Africa.
| |
Collapse
|
9
|
Antimicrobial Resistance and Virulence Genes of Streptococcus Agalactiae Isolated from Mastitis Milk Samples in China. J Vet Res 2022; 66:581-590. [PMID: 36846045 PMCID: PMC9944998 DOI: 10.2478/jvetres-2022-0069] [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: 05/07/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Streptococcus agalactiae is an important zoonotic pathogen that affects milk production and quality and poses a threat to public health. Treatment of infections with this bacterium exploits antimicrobials, to which the resistance of S. agalactiae is a growing problem. Addressing the possibility of a correlation between this pathogen's genetic factors for antimicrobial resistance and virulence, this study attempted to identify the relevant genes. Material and Methods Antimicrobial resistance of S. agalactiae isolated from 497 Chinese bovine mastitic milk samples was detected by the broth microdilution method. Eight drug resistance genes and eleven virulence genes were detected using PCR. Results Streptococcus agalactiae was 100% susceptible to rifampicin and vancomycin, 93.33% susceptible to sulfisoxazole and sulfamethoxazole, but 100% resistant to ≥3 of the 16 antimicrobial agents, thereby being multidrug resistant, with resistance to oxacillin, tetracycline, erythromycin, clindamycin, and gentamicin being common. The ermB, ermA and lnuA genes were carried by 73.33%, 66.67% and 60.00% of the strains, respectively. The carriage rates of the glnA, clyE, hylB, bibA, iagA, and fbsA virulence genes were greater than 40%, lmb and bac were not observed in any strain, and glnA+hylB+bibA+iagA+fbsA+clyE combined virulence gene patterns were the most commonly detected. Conclusion Antimicrobial resistance of S. agalactiae is still a great concern for cattle health in China, and multidrug resistance coupled with the high positive rates of this bacterium's strains for virulence genes indicates the importance of S. agalactiae surveillance and susceptibility tests.
Collapse
|
10
|
ICU Management of Invasive β-Hemolytic Streptococcal Infections. Infect Dis Clin North Am 2022; 36:861-887. [DOI: 10.1016/j.idc.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Virulence Profiles and Antimicrobial Resistance of Streptococcus agalactiae Infective and Colonizing Strains from Argentina. Curr Microbiol 2022; 79:392. [DOI: 10.1007/s00284-022-03050-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
|
12
|
The long and the short of Periscope Proteins. Biochem Soc Trans 2022; 50:1293-1302. [PMID: 36196877 DOI: 10.1042/bst20220194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022]
Abstract
Bacteria sense, interact with, and modify their environmental niche by deploying a molecular ensemble at the cell surface. The changeability of this exposed interface, combined with extreme changes in the functional repertoire associated with lifestyle switches from planktonic to adherent and biofilm states necessitate dynamic variability. Dynamic surface changes include chemical modifications to the cell wall; export of diverse extracellular biofilm components; and modulation of expression of cell surface proteins for adhesion, co-aggregation and virulence. Local enrichment for highly repetitive proteins with high tandem repeat identity has been an enigmatic phenomenon observed in diverse bacterial species. Preliminary observations over decades of research suggested these repeat regions were hypervariable, as highly related strains appeared to express homologues with diverse molecular mass. Long-read sequencing data have been interrogated to reveal variation in repeat number; in combination with structural, biophysical and molecular dynamics approaches, the Periscope Protein class has been defined for cell surface attached proteins that dynamically expand and contract tandem repeat tracts at the population level. Here, I review the diverse high-stability protein folds and coherent interdomain linkages culminating in the formation of highly anisotropic linear repeat arrays, so-called rod-like protein 'stalks', supporting roles in bacterial adhesion, biofilm formation, cell surface spatial competition, and immune system modulation. An understanding of the functional impacts of dynamic changes in repeat arrays and broader characterisation of the unusual protein folds underpinning this variability will help with the design of immunisation strategies, and contribute to synthetic biology approaches including protein engineering and microbial consortia construction.
Collapse
|
13
|
Tsai IA, Su Y, Wang YH, Chu C. Alterations in Genes rib, scpB and Pilus Island Decrease the Prevalence of Predominant Serotype V, Not III and VI, of Streptococcus agalactiae from 2008 to 2012. Pathogens 2022; 11:pathogens11101145. [PMID: 36297202 PMCID: PMC9611264 DOI: 10.3390/pathogens11101145] [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: 08/10/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
Streptococcus agalactiae (GBS) can infect newborns, pregnant women and immunocompromised or elderly people. This study aimed to investigate differences in three pilus genes and virulence genes pavA, cfb, rib and scpB and changes in predominant serotypes III, V and VI from 2008 to 2012. The susceptibilities to penicillin, ceftriaxone, azithromycin, erythromycin, clindamycin, levofloxacin and moxifloxacin of 145 GBS strains of serotype III, V and VI strains from 2008 and 2012 were determined using disc diffusion method. PCR identification of ST-17, the pilus genes and virulence genes; multilocus sequence typing (MLST); and conserved domain and phylogenetic analysis of scpB-1 and scpB-2 proteins were performed. A dramatic number reduction was observed in serotype V, not III and V, from 2008 to 2012. The rate of resistance to azithromycin, clindamycin and erythromycin was the highest in serotype V. ST-17 was only found in serotype III with pilus genes PI-1+PI-2b. The major pilus genotype was PI-1+PI-2a. Serotype V without the rib gene was reduced in number between two studied years. Compared to scpB-1, scpB-2 had a 128-bp deletion in a PA C5a-like peptidase domain and putative integrin-binding motif RGD. In conclusion, reduction in serotype V may be due to presence of scpB-2 or lack of genes scpB and rib.
Collapse
Affiliation(s)
- I-An Tsai
- Ph.D. Program of Agriculture Science, College of Agriculture, National Chiayi University, Chiayi City 600, Taiwan
| | - Yaochi Su
- Ph.D. Program of Agriculture Science, College of Agriculture, National Chiayi University, Chiayi City 600, Taiwan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chiayi University, Chiayi City 600, Taiwan
| | - Ying-Hsiang Wang
- Department of Pediatrics, Chang Gung Memorial Hospital, Puzi City 613, Taiwan
| | - Chishih Chu
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi City 600, Taiwan
- Correspondence: ; Tel.: +886-5-2717898
| |
Collapse
|
14
|
Keith MF, Gopalakrishna KP, Bhavana VH, Hillebrand GH, Elder JL, Megli CJ, Sadovsky Y, Hooven TA. Nitric Oxide Production and Effects in Group B Streptococcus Chorioamnionitis. Pathogens 2022; 11:1115. [PMID: 36297171 PMCID: PMC9608865 DOI: 10.3390/pathogens11101115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Intrauterine infection, or chorioamnionitis, due to group B Streptococcus (GBS) is a common cause of miscarriage and preterm birth. To cause chorioamnionitis, GBS must bypass maternal-fetal innate immune defenses including nitric oxide (NO), a microbicidal gas produced by nitric oxide synthases (NOS). This study examined placental NO production and its role in host-pathogen interactions in GBS chorioamnionitis. In a murine model of ascending GBS chorioamnionitis, placental NOS isoform expression quantified by RT-qPCR revealed a four-fold expression increase in inducible NOS, no significant change in expression of endothelial NOS, and decreased expression of neuronal NOS. These NOS expression results were recapitulated ex vivo in freshly collected human placental samples that were co-incubated with GBS. Immunohistochemistry of wild type C57BL/6 murine placentas with GBS chorioamnionitis demonstrated diffuse inducible NOS expression with high-expression foci in the junctional zone and areas of abscess. Pregnancy outcomes between wild type and inducible NOS-deficient mice did not differ significantly although wild type dams had a trend toward more frequent preterm delivery. We also identified possible molecular mechanisms that GBS uses to survive in a NO-rich environment. In vitro exposure of GBS to NO resulted in dose-dependent growth inhibition that varied by serovar. RNA-seq on two GBS strains with distinct NO resistance phenotypes revealed that both GBS strains shared several detoxification pathways that were differentially expressed during NO exposure. These results demonstrate that the placental immune response to GBS chorioamnionitis includes induced NO production and indicate that GBS activates conserved stress pathways in response to NO exposure.
Collapse
Affiliation(s)
- Mary Frances Keith
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | - Gideon Hayden Hillebrand
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Jordan Lynn Elder
- Manual Hematology and Coagulation Department, The Cleveland Clinic, Cleveland, OH 44195, USA
| | - Christina Joann Megli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- UPMC Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Yoel Sadovsky
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- UPMC Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Thomas Alexander Hooven
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
- UPMC Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- UPMC Children’s Hospital of Pittsburgh Richard King Mellon Institute for Pediatric Research, Pittsburgh, PA 15224, USA
- UPMC Children’s Hospital of Pittsburgh, 4401 Penn Ave. Rangos Research Building #8128, Pittsburgh, PA 15224, USA
| |
Collapse
|
15
|
Zhang K, Lu M, Zhu X, Wang K, Jie X, Li T, Dong H, Li R, Zhang F, Gu L. Antibiotic resistance and pathogenicity assessment of various Gardnerella sp. strains in local China. Front Microbiol 2022; 13:1009798. [PMID: 36225381 PMCID: PMC9549249 DOI: 10.3389/fmicb.2022.1009798] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022] Open
Abstract
Gardnerella overgrowth is the primary cause of bacterial vaginosis (BV), a common vaginal infection with incidences as high as 23-29% worldwide. Here, we studied the pathogenicity, drug resistance, and prevalence of varying Gardnerella spp. We isolated 20 Gardnerella strains from vaginal samples of 31 women in local China. Ten strains were then selected via phylogenetic analysis of cpn60 and vly gene sequences to carry out genome sequencing and comparative genomic analysis. Biofilm-formation, sialidase, and antibiotic resistance activities of the strains were characterized. All strains showed striking heterogeneity in genomic structure, biofilm formation and drug resistance. Two of the ten strains, JNFY3 and JNFY15, were classified as Gardnerella swidsinskii and Gardnerella piotii, respectively, according to their phenotypic characteristics and genome sequences. In particular, seven out of the ten strains exhibited super resistance (≥ 128 μg/mL) to metronidazole, which is the first line of treatment for BV in China. Based on the biochemical and genomic results of the strains, we proposed a treatment protocol of prevalent Gardnerella strains in local China, which provides the basis for accurate diagnosis and therapy.
Collapse
Affiliation(s)
- Kundi Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Mengyao Lu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiaoxuan Zhu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Kun Wang
- Jinan Key Laboratory of Female Reproductive Tract Infection, Jinan Genital Tract Microecological Clinical Laboratory, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xuemei Jie
- Jinan Key Laboratory of Female Reproductive Tract Infection, Jinan Genital Tract Microecological Clinical Laboratory, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tan Li
- Faculty of Health Sciences, Cumming School of Medicine, Calgary, AB, Canada
| | - Hongjie Dong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Rongguo Li
- Jinan Key Laboratory of Female Reproductive Tract Infection, Jinan Genital Tract Microecological Clinical Laboratory, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fengyu Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Lichuan Gu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
16
|
Marzhoseyni Z, Shayestehpour M, Salimian M, Esmaeili D, Saffari M, Fathizadeh H. Designing a novel fusion protein from Streptococcus agalactiae with apoptosis induction effects on cervical cancer cells. Microb Pathog 2022; 169:105670. [PMID: 35809755 DOI: 10.1016/j.micpath.2022.105670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022]
Abstract
Cervical cancer remains life-threatening cancer in women around the world. Due to the limitations of conventional treatment approaches, there is an urgent need to develop novel and more efficient strategies against cervical cancer. Therefore, the researchers attend to the alternative anti-cancer compounds like bacterial products. Rib and α are known as surface proteins of Streptococcus agalactiae with immunologic effects. In the present study, we designed a new anti-cancer fusion protein (Rib-α) originating from S. agalactiae with in silico methods, and then, the recombinant gene was cloned in the pET-22 (+) expression vector. The recombinant protein was expressed in E. coli BL21. To purify the expressed protein, we applied the Ni-NTA column. The molecular mechanism by which Rib-α is cytotoxic to cancer cells has been discussed based on MTT, flow cytometry, and real-time PCR methods. The engineered fusion protein suppressed the proliferation of the cancer cells at 180 μg/ml. Cytotoxic assessment and morphological changes, augmentation of apoptotic-related genes, upregulation of caspase-3 mRNA, and flow cytometric analysis confirmed that apoptosis might be the principal mechanism of cell death. According to our findings, Rib-α fusion protein motivated the intrinsic apoptosis pathway. Therefore, it can be an exciting candidate to discover a new class of antineoplastic agents.
Collapse
Affiliation(s)
- Zeynab Marzhoseyni
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Shayestehpour
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Morteza Salimian
- Anatomical Science Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| | - Davoud Esmaeili
- Department of Microbiology and Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mahmood Saffari
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Hadis Fathizadeh
- Student Research Committee, Sirjan School of Medical Sciences, Sirjan, Iran; Department of Laboratory Sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| |
Collapse
|
17
|
Jones S, Newton P, Payne M, Furfaro L. Epidemiology, Antimicrobial Resistance, and Virulence Determinants of Group B Streptococcus in an Australian Setting. Front Microbiol 2022; 13:839079. [PMID: 35774462 PMCID: PMC9238357 DOI: 10.3389/fmicb.2022.839079] [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: 12/19/2021] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus agalactiae [group B Streptococcus (GBS)] is a major neonatal pathogen and also causes invasive disease in non-pregnant adults. One hundred GBS isolates (n = 50 invasive disease and n = 50 colonizing pregnant women) were characterized using capsular serotyping by latex agglutination, antimicrobial susceptibility testing, and whole genome sequencing (WGS). All isolates were susceptible to penicillin, 32% were resistant to clindamycin. Of these, two isolates had reduced susceptibility to ceftriaxone (MIC 0.75 mg/L) and were found to have unique alleles at pbp2X and pbp1A. Capsular serotypes Ia (18%), III (18%), Ib (14%), V (12%), and VI (11%) were most common and comparison of latex agglutination and capsular genotyping by WGS showed 71% agreement. Less common capsular genotypes VI-VIII represented 15% of isolates, indicating that a significant proportion may not be targeted by the proposed pentavalent or hexavalent vaccines under development. WGS is a useful aid in GBS surveillance and shows correlation to phenotypic serotyping and antimicrobial susceptibility data.
Collapse
Affiliation(s)
- Sandra Jones
- Microbiology, NSW Health Pathology, Wollongong Hospital, Wollongong, NSW, Australia
| | - Peter Newton
- Microbiology, NSW Health Pathology, Wollongong Hospital, Wollongong, NSW, Australia
- Graduate Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Matthew Payne
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Perth, WA, Australia
- Women and Infants Research Foundation, Subiaco, WA, Australia
| | - Lucy Furfaro
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
18
|
Brokaw A, Nguyen S, Quach P, Orvis A, Furuta A, Johansson-Lindbom B, Fischer PB, Rajagopal L. A Recombinant Alpha-Like Protein Subunit Vaccine (GBS-NN) Provides Protection in Murine Models of Group B Streptococcus Infection. J Infect Dis 2022; 226:177-187. [PMID: 35429401 PMCID: PMC9890916 DOI: 10.1093/infdis/jiac148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Group B Streptococcus (GBS) transmission during pregnancy causes preterm labor, stillbirths, fetal injury, or neonatal infections. Rates of adult infections are also rising. The GBS-NN vaccine, engineered by fusing N-terminal domains of GBS Alpha C and Rib proteins, is safe in healthy, nonpregnant women, but further assessment is needed for use during pregnancy. Here, we tested GBS-NN vaccine efficacy using mouse models that recapitulate human GBS infection outcomes. METHODS Following administration of GBS-NN vaccine or adjuvant, antibody profiles were compared by ELISA. Vaccine efficacy was examined by comparing infection outcomes in GBS-NN vaccinated versus adjuvant controls during systemic and pregnancy-associated infections, and during intranasal infection of neonatal mice following maternal vaccination. RESULTS Vaccinated mice had higher GBS-NN-specific IgG titers versus controls. These antibodies bound alpha C and Rib on GBS clinical isolates. Fewer GBS were recovered from systemically challenged vaccinated mice versus controls. Although vaccination did not eliminate GBS during ascending infection in pregnancy, vaccinated dams experienced fewer in utero fetal deaths. Additionally, maternal vaccination prolonged neonatal survival following intranasal GBS challenge. CONCLUSIONS These findings demonstrate GBS-NN vaccine efficacy in murine systemic and perinatal GBS infections and suggest that maternal vaccination facilitates the transfer of protective antibodies to neonates.
Collapse
Affiliation(s)
- Alyssa Brokaw
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Shayla Nguyen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Phoenicia Quach
- Present affiliation: Phoenicia Quach, Universal Cells, Seattle 98121, Washington
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Anna Furuta
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Global Health, University of Washington, Seattle, Washington, USA
| | | | | | - Lakshmi Rajagopal
- Correspondence: L. Rajagopal, PhD, Department of Pediatrics, University of Washington, Seattle Children’s Hospital Research Institute, 307 Westlake Ave N, Seattle, WA 98109 ()
| |
Collapse
|
19
|
The Copper Resistome of Group B Streptococcus Reveals Insight into the Genetic Basis of Cellular Survival during Metal Ion Stress. J Bacteriol 2022; 204:e0006822. [PMID: 35404113 DOI: 10.1128/jb.00068-22] [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] [Indexed: 12/16/2022] Open
Abstract
In bacteria, copper (Cu) can support metabolic processes as an enzymatic cofactor but can also cause cell damage if present in excess, leading to intoxication. In group B Streptococcus (GBS), a system for control of Cu efflux based on the prototypical cop operon supports survival during Cu stress. In some other bacteria, genetic systems additional to the cop operon are engaged during Cu stress and also contribute to the management of cellular Cu homeostasis. Here, we examined genetic systems beyond the cop operon in GBS for regions that contribute to survival of GBS in Cu stress using a forward genetic screen and probe of the entire bacterial genome. A high-density mutant library, generated using pGh9-ISS1, was used to expose GBS to Cu stress and compare it to nonexposed controls en masse. Eight genes were identified as essential for GBS survival in Cu stress, whereas five genes constrained GBS growth in Cu stress. The genes encode varied factors including enzymes for metabolism, cell wall synthesis, transporters, and cell signaling factors. Targeted mutation of the genes validated their roles in GBS resistance to Cu stress. Excepting copA, the genes identified are new to the area of bacterial metal ion intoxication. We conclude that a discrete and limited suite of genes beyond the cop operon in GBS contributes to a repertoire of mechanisms used to survive Cu stress in vitro and achieve cellular homeostasis. IMPORTANCE Genetic systems for copper (Cu) homeostasis in bacteria, including streptococci, are vital to survive metal ion stress. Genetic systems that underpin survival of GBS during Cu stress, beyond the archetypal cop operon for Cu management, are undefined. We show that Streptococcus resists Cu intoxication by utilizing a discrete and limited suite of genes beyond the cop operon, including several genes that are new to the area of bacterial cell metal ion homeostasis. The Cu resistome of GBS defined here enhances our understanding of metal ion homeostasis in GBS.
Collapse
|
20
|
Dobrut A, Ochońska D, Brzozowska E, Górska S, Kaszuba-Zwoinska J, Gołda-Cępa M, Gamian A, Brzychczy-Wloch M. Molecular Characteristic, Antibiotic Resistance, and Detection of Highly Immunoreactive Proteins of Group B Streptococcus Strains Isolated From Urinary Tract Infections in Polish Adults. Front Microbiol 2022; 13:809724. [PMID: 35391726 PMCID: PMC8981152 DOI: 10.3389/fmicb.2022.809724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/21/2022] [Indexed: 12/02/2022] Open
Abstract
Group B streptococcus (GBS) is one of the uropathogens that causes urinary tract infections (UTIs). The aims of this article were molecular characterization, an analysis of antimicrobial susceptibility profiles, adherence to bladder endothelial cells, and the detection of immunoreactive proteins of 94 clinical strains of GBS isolated from adult Polish patients with UTI. Antibiotic susceptibilities were determined by disk diffusion. Serotyping and Alp family genes detection were studied using multiplex PCR. Genetic profiles were determined by pulsed-field gel electrophoresis. The adherence ability of the studied strains was estimated by incubation on human bladder microvascular endothelial cell line. Immunoreactive proteins were studied by immunoblotting. Antibiotic susceptibility investigation revealed that 22% of GBS strains were resistant to erythromycin, whereas 18% demonstrated resistance to clindamycin. cMLSB was present in 76% of the resistant strains, M phenotype was detected in 14%, whereas iMLSB was present for 10%. The most common serotype was serotype III (31%), followed by serotype V (27%), and serotype Ia (17%). The genes that dominated among other Alp genes were: epsilon (29%), alp2 (27%), and rib (23%). The most common co-occurring serotypes and Alp genes were: Ia and epsilon, III and rib, III and alp2, V and alp2, and V and alp3 (p < 0.001). The PFGE method showed high clonality for serotype V and cMLSB (p < 001). The PFGE method showed high clonality for serotype V. Furthermore, this serotype was significantly associated with the cMLSB phenotype (p < 0.001). The most common immunoreactive proteins demonstrated masses of 50 kDa and 45–47 kDa. Although examined GBS isolates showed high genetic diversity, immunoreactive proteins were common for most of the studied GBS isolates, which may indicate their conservation, and allows to consider them as potential immunodiagnostic markers. Although the examined GBS isolates showed high genetic diversity, immunoreactive proteins were shared by most of the studied GBS isolates. It may indicate their conservation, thus allowing to consider them as potential immunodiagnostic markers.
Collapse
Affiliation(s)
- Anna Dobrut
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Dorota Ochońska
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Ewa Brzozowska
- Laboratory of Medical Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Jolanta Kaszuba-Zwoinska
- Chair of Pathophysiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | | | - Andrzej Gamian
- Laboratory of Medical Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Monika Brzychczy-Wloch
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
21
|
Kaushik S, Yadav J, Das S, Karthikeyan D, Chug R, Jyoti A, Srivastava VK, Jain A, Kumar S, Sharma V. Identification of Protein Drug Targets of Biofilm Formation and Quorum
Sensing in Multidrug Resistant Enterococcus faecalis. Curr Protein Pept Sci 2022; 23:248-263. [DOI: 10.2174/1389203723666220526155644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/16/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Enterococcus faecalis (E. faecalis) is an opportunistic multidrug-resistant (MDR) pathogen
found in the guts of humans and farmed animals. Due to the occurrence of (MDR) strain there is an
urgent need to look for an alternative treatment approach. E. faecalis is a Gram-positive bacterium,
which is among the most prevalent multidrug resistant hospital pathogens. Its ability to develop quorum
sensing (QS) mediated biofilm formation further exacerbates the pathogenicity and triggers lifethreatening
infections. Therefore, developing a suitable remedy for curing E. faecalis mediated enterococcal
infections is an arduous task. Several putative virulence factors and proteins are involved in the
development of biofilms in E. faecalis. Such proteins often play important roles in virulence, disease,
and colonization by pathogens. The elucidation of the structure-function relationship of such protein
drug targets and the interacting compounds could provide an attractive paradigm towards developing
structure-based drugs against E. faecalis. This review provides a comprehensive overview of the current
status, enigmas that warrant further studies, and the prospects toward alleviating the antibiotic resistance
in E. faecalis. Specifically, the role of biofilm and quorum sensing (QS) in the emergence of
MDR strains had been elaborated along with the importance of the protein drug targets involved in both
the processes.
Collapse
Affiliation(s)
- Sanket Kaushik
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Jyoti Yadav
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Satyajeet Das
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
- Structural Biology Lab, CSIR-Institute of Microbial Technology, Chandigarh, India
| | | | - Ravneet Chug
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Anupam Jyoti
- Department of Biotechnology, University Institute of Biotechnology,
Chandigarh University, Chandigarh, India
| | | | - Ajay Jain
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Sanjit Kumar
- Centre for Bioseparation Technology, VIT
University, Vellore-632014, Tamil Nadu, India
| | - Vinay Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| |
Collapse
|
22
|
Xu X, Marffy ALL, Keightley A, McCarthy AJ, Geisbrecht BV. Group B Streptococcus Surface Protein β: Structural Characterization of a Complement Factor H-Binding Motif and Its Contribution to Immune Evasion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1232-1247. [PMID: 35110419 PMCID: PMC8881398 DOI: 10.4049/jimmunol.2101078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 11/19/2022]
Abstract
The β protein from group B Streptococcus (GBS) is a ∼132-kDa, cell-surface exposed molecule that binds to multiple host-derived ligands, including complement factor H (FH). Many details regarding this interaction and its significance to immune evasion by GBS remain unclear. In this study, we identified a three-helix bundle domain within the C-terminal half of the B75KN region of β as the major FH-binding determinant and determined its crystal structure at 2.5 Å resolution. Analysis of this structure suggested a role in FH binding for a loop region connecting helices α1 and α2, which we confirmed by mutagenesis and direct binding studies. Using a combination of protein cross-linking and mass spectrometry, we observed that B75KN bound to complement control protein (CCP)3 and CCP4 domains of FH. Although this binding site lies within a complement regulatory region of FH, we determined that FH bound by β retained its decay acceleration and cofactor activities. Heterologous expression of β by Lactococcus lactis resulted in recruitment of FH to the bacterial surface and a significant reduction of C3b deposition following exposure to human serum. Surprisingly, we found that FH binding by β was not required for bacterial resistance to phagocytosis by neutrophils or killing of bacteria by whole human blood. However, loss of the B75KN region significantly diminished bacterial survival in both assays. Although our results show that FH recruited to the bacterial surface through a high-affinity interaction maintains key complement-regulatory functions, they raise questions about the importance of FH binding to immune evasion by GBS as a whole.
Collapse
Affiliation(s)
- Xin Xu
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS U.S.A
| | - Alexander L. Lewis Marffy
- Department of Infectious Diseases, Section of Molecular Microbiology, MRC Centre for Molecular Bacteriology & Infection, Imperial College London; London, U.K
| | - Andrew Keightley
- Department of Opthamology, School of Medicine, University of Missouri-Kansas City; Kansas City, MO U.S.A
| | - Alex J. McCarthy
- Department of Infectious Diseases, Section of Molecular Microbiology, MRC Centre for Molecular Bacteriology & Infection, Imperial College London; London, U.K
| | - Brian V. Geisbrecht
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS U.S.A.,To whom correspondence should be addressed: Brian V. Geisbrecht, Ph.D., Kansas State University, 141 Chalmers Hall, 1711 Claflin Road, Manhattan, KS 66506, PH: 785.532.3154,
| |
Collapse
|
23
|
Peng J, Lu Q, Liu X, Deng Y, Shang T, Yuan L, Zhang H, Zeng Q. Antibacterial effect of synthetic ultra-short lipopeptide on Streptococcus agalactiae and its active on bacterial mastitis in mice. Biochem Biophys Res Commun 2022; 601:153-159. [PMID: 35247769 DOI: 10.1016/j.bbrc.2022.02.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 01/01/2023]
Abstract
Streptococcus agalactiae mastitis is one of the significant threats to the milk industry. The traditional antibiotic treatment method is easy to cause the emergence of resistant strains, and the problem of drug residue is increasingly severe. In this study, we designed and synthesized five lipopeptides. The antibacterial activity of different molecular structure lipopeptides against Streptococcus agalactiae was detected. Furthermore, the mouse mastitis model was established using Streptococcus agalactiae. The lipopeptides with better antibacterial effect were selected for the treatment experiment to evaluate the application value in the treatment of mastitis. The results showed that 4 of the synthesized lipopeptides had specific antibacterial activity. SLP3 and SLP4 have an excellent antibacterial effect and can treat murine mastitis caused by Streptococcus agalactiae infection within the safe concentration range. The results of this study can provide an excellent experimental basis for new antibiotics and clinical application in the treatment of dairy cow mastitis.
Collapse
Affiliation(s)
- Jie Peng
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, PR China.
| | - Qiangsheng Lu
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, PR China
| | - Xuming Liu
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, PR China
| | - Yuanjie Deng
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, PR China
| | - Tiantian Shang
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, PR China
| | - Lvfeng Yuan
- Lanzhou Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Gansu, PR China
| | - Hecheng Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, PR China
| | - Qiaoying Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, PR China
| |
Collapse
|
24
|
Dobrut A, Brzychczy-Włoch M. Immunogenic Proteins of Group B Streptococcus-Potential Antigens in Immunodiagnostic Assay for GBS Detection. Pathogens 2021; 11:43. [PMID: 35055991 PMCID: PMC8778278 DOI: 10.3390/pathogens11010043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/29/2021] [Accepted: 12/29/2021] [Indexed: 12/21/2022] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) is an opportunistic pathogen, which asymptomatically colonizes the gastrointestinal and genitourinary tract of up to one third of healthy adults. Nevertheless, GBS carriage in pregnant women may lead to several health issues in newborns causing life threatening infection, such as sepsis, pneumonia or meningitis. Recommended GBS screening in pregnant women significantly reduced morbidity and mortality in infants. Nevertheless, intrapartum antibiotic prophylaxis, recommended following the detection of carriage or in case of lack of a carriage test result for pregnant women who demonstrate certain risk factors, led to the expansion of the adverse phenomenon of bacterial resistance to antibiotics. In our paper, we reviewed some immunogenic GBS proteins, i.e., Alp family proteins, β protein, Lmb, Sip, BibA, FsbA, ScpB, enolase, elongation factor Tu, IMPDH, and GroEL, which possess features characteristic of good candidates for immunodiagnostic assays for GBS carriage detection, such as immunoreactivity and specificity. We assume that they can be used as an alternative diagnostic method to the presently recommended bacteriological cultivation and MALDI.
Collapse
Affiliation(s)
| | - Monika Brzychczy-Włoch
- Department of Molecular Medical Microbiology, Faculty of Medicine, Medical College, Jagiellonian University, 31-121 Krakow, Poland;
| |
Collapse
|
25
|
Awwad E, Srour M, Hasan S, Khatib S. Molecular determination, serotyping, antibiotic profile and virulence factors of group B Streptococcus isolated from invasive patients at Arabcare Hospital Laboratory, Palestine. Am J Infect Control 2021; 50:934-940. [PMID: 34963647 DOI: 10.1016/j.ajic.2021.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Streptococcus agalactiae (group B Streptococcus) is beta-hemolytic, catalase negative, gram-positive cocci, recognized as main bacterial pathogen causing infections in newborns, infants, adults, and elderly people around the world. The aim of this study is to investigate group B Streptococcus samples recovered from invasive patients and determine serotype, virulent genes, and antibiotic-resistant profile of Streptococcus agalactiae in Palestine. METHODS A total of 95 group B Streptococcus strains were isolated from neonates, infants, pregnant and non-pregnant women and males at Arabcare Hospital Laboratory, Palestine, between the period of June 2018 and September 2020. Species identification was carried out through cultivation and conventional biochemical tests. A conventional Polymerase Chain Reaction (cPCR) was used to determine the 5 serotypes and virulent genes of the Streptococcus agalactiae strains. The antibiotic resistance test of group B Streptococcus was evaluated using Kirby-Bauer disk susceptibility. Sequencing and BLAST analysis were used to determine the relationship of the isolates in this study to worldwide isolates. RESULTS Serotype III (35%) was the major group B Streptococcus strains serotype causing invasive infections in neonates, infants, pregnant and nonpregnant women, and males, followed by serotypes V (19%), Ia, and II (15%), Ib (6%), respectively. All our isolates encoding for surface protein virulent factors, including a highly virulent gene (HvgA) were mostly found in strains isolated from pregnant women (12%). These group B Streptococcus strains exhibited a high rate of resistance to clindamycin (26%). The overall percentage of levofloxacin resistance was 11%, while vancomycin and ampicillin showed higher resistance, at 14.7 and 16% respectively. In addition, the phylogenetic relationship dendrogram illustrates that Streptococcus agalactiae isolated from an invasive patient (newborn) in Palestine was similar to strains found in China and Japan. CONCLUSIONS The outcomes of this study demonstrate that resistant group B Streptococcus strains are common in Palestine, therefore, evidence-based infection prevention and antibiotic stewardship efforts are necessary.
Collapse
Affiliation(s)
| | - Mahmoud Srour
- Biology and Biochemistry Department, Faculty of Science, Birzeit University, Palestine
| | - Shadi Hasan
- Biology and Biochemistry Department, Faculty of Science, Birzeit University, Palestine
| | | |
Collapse
|
26
|
Shabayek S, Ferrieri P, Spellerberg B. Group B Streptococcal Colonization in African Countries: Prevalence, Capsular Serotypes, and Molecular Sequence Types. Pathogens 2021; 10:pathogens10121606. [PMID: 34959562 PMCID: PMC8706430 DOI: 10.3390/pathogens10121606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 11/20/2022] Open
Abstract
Streptococcus agalactiae or group B streptococcus (GBS) is a commensal of the gastrointestinal and genitourinary tracts of healthy women and an important cause of neonatal invasive infections worldwide. Transmission of bacteria to the newborn occurs at birth and can be prevented by intrapartum antibiotic prophylaxis. However, this not available in resource limited settings in Africa, which carries a particular high burden of disease. Serotype based vaccines are in development and present a suitable alternative to prevent neonatal infections. To be able to assess vaccine efficacy, knowledge and surveillance of GBS epidemiological data are required. This review summarizes investigations about the serotype distribution and the multi-locus sequence types (MLST) found in different African countries. While most serotypes and MLST data are comparable to findings from other continents, some specific differences exist. Serotype V is predominant among colonizing maternal strains in many different African countries. Serotypes that are rarely detected in western industrialized nations, such as serotypes VI, VII and IX, are prevalent in studies from Ghana and Egypt. Moreover, some specific MLST sequence types that seem to be more or less unique to Africa have been detected. However, overall, the data confirm that a hexavalent vaccine can provide broad coverage for the African continent and that a protein vaccine could represent a promising alternative.
Collapse
Affiliation(s)
- Sarah Shabayek
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| | - Patricia Ferrieri
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, 89081 Ulm, Germany
- Correspondence:
| |
Collapse
|
27
|
Kwofie SK, Broni E, Yunus FU, Nsoh J, Adoboe D, Miller WA, Wilson MD. Molecular Docking Simulation Studies Identifies Potential Natural Product Derived-Antiwolbachial Compounds as Filaricides against Onchocerciasis. Biomedicines 2021; 9:biomedicines9111682. [PMID: 34829911 PMCID: PMC8615632 DOI: 10.3390/biomedicines9111682] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
Onchocerciasis is the leading cause of blindness and severe skin lesions which remain a major public health problem, especially in tropical areas. The widespread use of antibiotics and the long duration required for effective treatment continues to add to the increasing global menace of multi-resistant pathogens. Onchocerca volvulus harbors the endosymbiont bacteria Wolbachia, essential for the normal development of embryos, larvae and long-term survival of the adult worm, O. volvulus. We report here results of using structure-based drug design (SBDD) approach aimed at identifying potential novel Wolbachia inhibitors from natural products against the Wolbachia surface protein (WSP). The protein sequence of the WSP with UniProtKB identifier Q0RAI4 was used to model the three-dimensional (3D) structure via homology modelling techniques using three different structure-building algorithms implemented in Modeller, I-TASSER and Robetta. Out of the 15 generated models of WSP, one was selected as the most reasonable quality model which had 82, 15.5, 1.9 and 0.5% of the amino acid residues in the most favored regions, additionally allowed regions, generously allowed regions and disallowed regions, respectively, based on the Ramachandran plot. High throughput virtual screening was performed via Autodock Vina with a library comprising 42,883 natural products from African and Chinese databases, including 23 identified anti-Onchocerca inhibitors. The top six compounds comprising ZINC000095913861, ZINC000095486235, ZINC000035941652, NANPDB4566, acetylaleuritolic acid and rhemannic acid had binding energies of −12.7, −11.1, −11.0, −11, −10.3 and −9.5 kcal/mol, respectively. Molecular dynamics simulations including molecular mechanics Poisson-Boltzmann (MMPBSA) calculations reinforced the stability of the ligand-WSP complexes and plausible binding mechanisms. The residues Arg45, Tyr135, Tyr148 and Phe195 were predicted as potential novel critical residues required for ligand binding in pocket 1. Acetylaleuritolic acid and rhemannic acid (lantedene A) have previously been shown to possess anti-onchocercal activity. This warrants the need to evaluate the anti-WSP activity of the identified molecules. The study suggests the exploitation of compounds which target both pockets 1 and 2, by investigating their potential for effective depletion of Wolbachia. These compounds were predicted to possess reasonably good pharmacological profiles with insignificant toxicity and as drug-like. The compounds were computed to possess biological activity including antibacterial, antiparasitic, anthelmintic and anti-rickettsials. The six natural products are potential novel antiwolbachial agents with insignificant toxicities which can be explored further as filaricides for onchocerciasis.
Collapse
Affiliation(s)
- Samuel K. Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic and Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra LG 77, Ghana; (E.B.); (F.U.Y.); (J.N.); (D.A.)
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
- Correspondence: ; Tel.: +233-203-797922
| | - Emmanuel Broni
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic and Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra LG 77, Ghana; (E.B.); (F.U.Y.); (J.N.); (D.A.)
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
- Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, P.O. Box LG 581, Legon, Accra LG 581, Ghana;
| | - Faruk U. Yunus
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic and Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra LG 77, Ghana; (E.B.); (F.U.Y.); (J.N.); (D.A.)
| | - John Nsoh
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic and Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra LG 77, Ghana; (E.B.); (F.U.Y.); (J.N.); (D.A.)
| | - Dela Adoboe
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic and Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra LG 77, Ghana; (E.B.); (F.U.Y.); (J.N.); (D.A.)
| | - Whelton A. Miller
- Department of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA;
- Department of Molecular Pharmacology and Neuroscience, Loyola University Medical Center, Maywood, IL 60153, USA
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, IL 19104, USA
| | - Michael D. Wilson
- Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, P.O. Box LG 581, Legon, Accra LG 581, Ghana;
- Department of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA;
| |
Collapse
|
28
|
Sulovari A, Ninomiya MJ, Beck CA, Ricciardi BF, Ketonis C, Mesfin A, Kaplan NB, Soin SP, McDowell SM, Mahmood B, Daiss JL, Schwarz EM, Oh I. Clinical utilization of species-specific immunoassays for identification of Staphylococcus aureus and Streptococcus agalactiae in orthopedic infections. J Orthop Res 2021; 39:2141-2150. [PMID: 33274775 PMCID: PMC8175449 DOI: 10.1002/jor.24935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/09/2020] [Accepted: 12/01/2020] [Indexed: 02/04/2023]
Abstract
Staphylococcus aureus and Streptococcus agalactiae (Group B streptococcus, GBS) are common causes of deep musculoskeletal infections (MSKI) and result in significant patient morbidity and cost to the healthcare system. One of the major challenges with MSKI is the lack of faithful diagnostics to correctly identify the primary pathogen, as standard culture-based assays are prone to false positives in the case of polymicrobial infections, and false negatives due to limitations in sample acquisition and antibiotic use before presentation. To improve upon our current diagnostic methods for MSKI, we developed a multiplex immunoassay for antigen-specific IgGs in serum (Luminex), and medium enriched for newly synthesized antibodies (MENSA) for anti-S. aureus and GBS generated from cultured peripheral blood mononuclear cells (PBMCs) of orthopedic infection patients undergoing surgical treatment. Samples were obtained from 110 MSKI patients: 80 diabetic foot ulcer, 21 periprosthetic joint infection, 5 septic arthritis, 2 spine, 1 hand, and 1 fracture-related infection (FRI). Anti-S. aureus and anti-GBS antibody titers were compared to culture results to assess their concordance in identifying the pathogens. Immunoassay, particularly MENSA, showed high diagnostic potential for monomicrobial S. aureus and GBS orthopedic infections (AUC > 0.95). MENSA also demonstrated diagnostic potential for GBS polymicrobial orthopedic infection and for GBS DFU (AUC > 0.83 for both). Serum showed high diagnostic potential for S. aureus PJI (AUC > 0.95). Taken together, these findings support the development of species-specific immunoassays for the identification of causal pathogens in active MSKI, especially in conjunction with standard culture.
Collapse
Affiliation(s)
- Aron Sulovari
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Mark J. Ninomiya
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher A. Beck
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Benjamin F. Ricciardi
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Constantinos Ketonis
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Addisu Mesfin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Nathan B. Kaplan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Sandeep P. Soin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Susan M. McDowell
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Bilal Mahmood
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - John L. Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Irvin Oh
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
29
|
Shen J, Wu X, Yang Y, Lv Y, Li X, Ding X, Wang S, Yan Z, Yan Y, Yang F, Li H. Antimicrobial Resistance and Virulence Factor of Streptococcus dysgalactiae Isolated from Clinical Bovine Mastitis Cases in Northwest China. Infect Drug Resist 2021; 14:3519-3530. [PMID: 34511943 PMCID: PMC8418370 DOI: 10.2147/idr.s327924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/24/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Streptococcus dysgalactiae is a major pathogen in bovine mastitis. The purpose of this study was to survey the prevalence, antimicrobial resistance, as well as the spread of resistance and virulence-associated gene of S. dysgalactiae. Methods A total of 60 S. dysgalactiae strains were obtained from 830 milk samples from Holstein cows with clinical mastitis. Antimicrobial resistance was examined by the disk diffusion method. Antimicrobial resistance and virulence genes were investigated by PCR, agarose gel electrophoresis and 16S rRNA gene sequencing. Results All isolates were resistant to tetracycline and showed a high level of resistance to aminoglycoside antibiotics, where 81.67% of the strains were multi-resistant to these ten sorts of antibiotics. In addition, the most prevalent resistance gene in S. dysgalactiae was aphA-1 (98.33%), followed by blaTEM (96.67%), ermB (83.3%), aadA1/aadA2 (78.33%) and tetL (73.33%). Totally, seven virulence genes with 25 combination patterns were detected in these isolates, and each isolates harbored at least one virulence gene. 21.67% of the isolates carried three or more virulence genes, while one strain with seven virulence-related genes and belonged to cfb+lmb+eno+napr+bca+scpB+cyl. Conclusion These findings indicate that S. dysgalactiae isolated from clinical bovine mastitis cases in Northwest China show a variety of molecular ecology and are highly resistant to antibiotics commonly used in dairy farms. This research will help investigators better understand the pathophysiology S. dysgalactiae in bovine mastitis and choose the appropriate antibiotics to treat mastitis.
Collapse
Affiliation(s)
- Jirao Shen
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xiaohu Wu
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Yayuan Yang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Yanan Lv
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xinpu Li
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xuezhi Ding
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Shengyi Wang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Zuoting Yan
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Yong Yan
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Feng Yang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Hongsheng Li
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| |
Collapse
|
30
|
Mazzuoli MV, Daunesse M, Varet H, Rosinski-Chupin I, Legendre R, Sismeiro O, Gominet M, Kaminski PA, Glaser P, Chica C, Trieu-Cuot P, Firon A. The CovR regulatory network drives the evolution of Group B Streptococcus virulence. PLoS Genet 2021; 17:e1009761. [PMID: 34491998 PMCID: PMC8448333 DOI: 10.1371/journal.pgen.1009761] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/17/2021] [Accepted: 08/09/2021] [Indexed: 01/31/2023] Open
Abstract
Virulence of the neonatal pathogen Group B Streptococcus is under the control of the master regulator CovR. Inactivation of CovR is associated with large-scale transcriptome remodeling and impairs almost every step of the interaction between the pathogen and the host. However, transcriptome analyses suggested a plasticity of the CovR signaling pathway in clinical isolates leading to phenotypic heterogeneity in the bacterial population. In this study, we characterized the CovR regulatory network in a strain representative of the CC-17 hypervirulent lineage responsible of the majority of neonatal meningitis. Transcriptome and genome-wide binding analysis reveal the architecture of the CovR network characterized by the direct repression of a large array of virulence-associated genes and the extent of co-regulation at specific loci. Comparative functional analysis of the signaling network links strain-specificities to the regulation of the pan-genome, including the two specific hypervirulent adhesins and horizontally acquired genes, to mutations in CovR-regulated promoters, and to variability in CovR activation by phosphorylation. This regulatory adaptation occurs at the level of genes, promoters, and of CovR itself, and allows to globally reshape the expression of virulence genes. Overall, our results reveal the direct, coordinated, and strain-specific regulation of virulence genes by the master regulator CovR and suggest that the intra-species evolution of the signaling network is as important as the expression of specific virulence factors in the emergence of clone associated with specific diseases. Streptococcus agalactiae, commonly known as the Group B Streptococcus (GBS), is a commensal bacterium of the intestinal and vaginal tracts found in approximately 30% of healthy adults. However, GBS is also an opportunistic pathogen and the leading cause of neonatal invasive infections. Epidemiologic data have identified a particular GBS clone, designated the CC-17 hypervirulent clonal complex, as responsible for the overwhelming majority of neonatal meningitis. The hypervirulence of CC-17 has been linked to the expression of two specific surface proteins increasing their abilities to cross epithelial and endothelial barriers. In this study, we characterized the role of the major regulator of virulence gene expression, the CovR response regulator, in a representative hypervirulent strain. Transcriptome and genome-wide binding analysis reveal the architecture of the CovR signaling network characterized by the direct repression of a large array of virulence-associated genes, including the specific hypervirulent adhesins. Comparative analysis in a non-CC-17 wild type strain demonstrates a high level of plasticity of the regulatory network, allowing to globally reshape pathogen-host interaction. Overall, our results suggest that the intra-species evolution of the regulatory network is an important factor in the emergence of GBS clones associated with specific pathologies.
Collapse
Affiliation(s)
- Maria-Vittoria Mazzuoli
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
- Sorbonne Paris Cité, Université de Paris, Paris, France
| | - Maëlle Daunesse
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, Paris, France
| | - Hugo Varet
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, Paris, France
- Plate-forme Technologique Biomics—Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, Paris, France
| | - Isabelle Rosinski-Chupin
- Unité Écologie et Évolution de la Résistance aux Antibiotiques, CNRS UMR3525, Institut Pasteur, Paris, France
| | - Rachel Legendre
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, Paris, France
- Plate-forme Technologique Biomics—Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, Paris, France
| | - Odile Sismeiro
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
- Plate-forme Technologique Biomics—Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, Paris, France
| | - Myriam Gominet
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
| | - Pierre Alexandre Kaminski
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
| | - Philippe Glaser
- Unité Écologie et Évolution de la Résistance aux Antibiotiques, CNRS UMR3525, Institut Pasteur, Paris, France
| | - Claudia Chica
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, Paris, France
| | - Patrick Trieu-Cuot
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
| | - Arnaud Firon
- Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, Paris, France
- * E-mail:
| |
Collapse
|
31
|
C-protein α-antigen modulates the lantibiotic thusin resistance in Streptococcus agalactiae. Antonie van Leeuwenhoek 2021; 114:1595-1607. [PMID: 34319449 DOI: 10.1007/s10482-021-01626-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
Screening for producers of potent antimicrobial peptides, resulted in the isolation of Bacillus cereus BGNM1 with strong antimicrobial activity against Listeria monocytogenes. Genome sequence analysis revealed that BGNM1 contains the gene cluster associated with the production of the lantibiotic, thusin, previously identified in B. thuringiensis. Purification of the antimicrobial activity confirmed that strain BGMN1 produces thusin. Both thusin sensitive and resistant strains were detected among clinical isolates of Streptococcus agalactiae. Random mutagenesis of a thusin sensitive strain, S. agalactiae B782, was performed in an attempt to identify the receptor protein for thusin. Three independent thusin resistant mutants were selected and their complete genomes sequenced. Comparative sequence analysis of these mutants with the WT strain revealed that duplication of a region encoding a 79 amino acids repeat in a C-protein α-antigen was a common difference, suggesting it to be responsible for increased resistance to thusin. Since induced thusin resistant mutants showed higher level of resistance than the naturally resistant B761 strain, complete genome sequencing of strain B761 was performed to check the integrity of the C-protein α-antigen-encoding gene. This analysis revealed that this gene is deleted in B761, providing further evidence that this protein promotes interaction of the thusin with receptor.
Collapse
|
32
|
Copper intoxication in group B Streptococcus triggers transcriptional activation of the cop operon that contributes to enhanced virulence during acute infection. J Bacteriol 2021; 203:e0031521. [PMID: 34251869 DOI: 10.1128/jb.00315-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacteria can utilize Copper (Cu) as a trace element to support cellular processes; however, excess Cu can intoxicate bacteria. Here, we characterize the cop operon in group B streptococcus (GBS), and establish its role in evasion of Cu intoxication and the response to Cu stress on virulence. Growth of GBS mutants deficient in either the copA Cu exporter, or the copY repressor, were severely compromised in Cu-stress conditions. GBS survival of Cu stress reflected a mechanism of CopY de-repression of the CopA efflux system. However, neither mutant was attenuated for intracellular survival in macrophages. Analysis of global transcriptional responses to Cu by RNA-sequencing revealed a stress signature encompassing homeostasis of multiple metals. Genes induced by Cu stress included putative metal transporters for manganese import, whereas a system for iron export was repressed. In addition, copA promoted the ability of GBS to colonize the blood, liver and spleen of mice following disseminated infection. Together, these findings show that GBS copA mediates resistance to Cu intoxication, via regulation by the Cu-sensing transcriptional repressor, copY. Cu stress responses in GBS reflect a transcriptional signature that heightens virulence and represents an important part of the bacteria's ability to survive in different environments. Importance Understanding how bacteria manage cellular levels of metal ions, such as copper, helps to explain how microbial cells can survive in different stressful environments. We show how the opportunistic pathogen group B Streptococcus (GBS) achieves homeostasis of intracellular copper through the activities of the genes that comprise the cop operon, and describe how this helps GBS survive in stressful environments, including in the mammalian host during systemic disseminated infection.
Collapse
|
33
|
Rainard P, Gilbert FB, Germon P, Foucras G. Invited review: A critical appraisal of mastitis vaccines for dairy cows. J Dairy Sci 2021; 104:10427-10448. [PMID: 34218921 DOI: 10.3168/jds.2021-20434] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/23/2021] [Indexed: 11/19/2022]
Abstract
Infections of the mammary gland remain a frequent disease of dairy ruminants that negatively affect animal welfare, milk quality, farmer serenity, and farming profitability and cause an increase in use of antimicrobials. There is a need for efficacious vaccines to alleviate the burden of mastitis in dairy farming, but this need has not been satisfactorily fulfilled despite decades of research. A careful appraisal of past and current research on mastitis vaccines reveals the peculiarities but also the commonalities among mammary gland infections associated with the major mastitis pathogens Escherichia coli, Staphylococcus aureus, Streptococcus uberis, Streptococcus agalactiae, or Streptococcus dysgalactiae. A major pitfall is that the immune mechanisms of effective protection have not been fully identified. Until now, vaccine development has been directed toward the generation of antibodies. In this review, we drew up an inventory of the main approaches used to design vaccines that aim at the major pathogens for the mammary gland, and we critically appraised the current and tentative vaccines. In particular, we sought to relate efficacy to vaccine-induced defense mechanisms to shed light on some possible reasons for current vaccine shortcomings. Based on the lessons learned from past attempts and the recent results of current research, the design of effective vaccines may take a new turn in the years to come.
Collapse
Affiliation(s)
- Pascal Rainard
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Tours, Infectiologie et Santé Publique, 37380 Nouzilly, France.
| | - Florence B Gilbert
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Tours, Infectiologie et Santé Publique, 37380 Nouzilly, France
| | - Pierre Germon
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Tours, Infectiologie et Santé Publique, 37380 Nouzilly, France
| | - Gilles Foucras
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Toulouse, École Nationale Vétérinaire de Toulouse, Interactions Hôtes-Agents Pathogènes, 31076 Toulouse, France
| |
Collapse
|
34
|
Safety and immunogenicity of a prototype recombinant alpha-like protein subunit vaccine (GBS-NN) against Group B Streptococcus in a randomised placebo-controlled double-blind phase 1 trial in healthy adult women. Vaccine 2021; 39:4489-4499. [PMID: 34215454 DOI: 10.1016/j.vaccine.2021.06.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/24/2021] [Accepted: 06/18/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Group B Streptococcus (GBS) is the leading cause of life-threatening infections in new-borns and may cause invasive disease, stillbirth and preterm delivery during pregnancy. While no licensed vaccine exists, maternal immunization might protect against neonatal disease and adverse pregnancy outcomes. We assessed the safety and immunogenicity of a prototype vaccine consisting of the fused N-terminal domains of the AlphaC and Rib surface proteins of GBS (GBS-NN). METHODS GBS-NN was tested in a randomised, double-blind, placebo-controlled, parallel group, phase I study, in healthy non-pregnant women. A dose-escalation phase, with two doses, four weeks apart, of 10, 50 or 250 µg, administered with or without aluminium hydroxide, was initially assessed (n = 60). This was followed by a dose-confirmation study, where one dose of 100 µg adjuvanted GBS-NN was compared with two doses of either 50 or 100 µg adjuvanted GBS-NN, again administered with four weeks interval between the doses (n = 180). Safety and immunogenicity were monitored for one year. RESULTS GBS-NN was well tolerated with some, mostly mild, injection site reactions observed. Adjuvant significantly increased antibody concentrations and the response was boosted by a second dose. The IgG GMCs remained strongly elevated during the whole one-year duration of the study. Maximal responses occurred after two 50 µg doses, resulting in IgG GMC of 16.9 µg/ml at the primary immunological endpoint, twelve weeks after the first dose. For this regimen, 100% and 89% of the subjects achieved antibody levels above the arbitrary thresholds of 1 and 4 µg/ml, respectively. The added beneficial effect of a second dose was most pronounced for subjects with pre-existing IgG levels below the median of the entire cohort. CONCLUSION The prototype GBS-NN vaccine was found to be well tolerated and highly immunogenic with an optimal regimen of two doses of 50 µg in the presence of adjuvant. Further development of a maternal vaccine based on the N-terminal domains of the alpha-like protein family of GBS is warranted (NCT02459262).
Collapse
|
35
|
Ding Y, Wu Q, Guo Y, Li M, Li P, Ma Y, Liu W. Effects of in vitro-induced drug resistance on the virulence of Streptococcus. Vet Med Sci 2021; 7:935-943. [PMID: 33314727 PMCID: PMC8136945 DOI: 10.1002/vms3.404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/25/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
This study aimed to evaluate the effects of in vitro-induced drug resistance on the virulence of Streptococcus. Micro-dilution method was used to determine the minimal inhibitory concentration (MIC). In vitro-induced drug resistance was conducted for S. agalactiae (CVCC1886) and S. dysgalactiae (CVCC3701) by gradually increasing the antimicrobial concentration (strains were from IVDC, China). PCR was used to detect the resistance and virulence genes of the strains before and after resistance induction. Colony morphology was observed to compare the physiological and biochemical properties of the strains. A total of 88 clean-grade Kunming mice (obtained from Inner Mongolia University, Hohhot, China) were used in half of the lethal dose (LD50) test for detecting the changes in virulence of strains. The results showed that S. agalactiae (CVCC1886) and S. dysgalactiae (CVCC3701) developed resistance against seven kinds of antibiotics, respectively. Resistance and virulence genes of CVCC3701 were changed when treated by the Penicillin-inducing. The growth of the CVCC3701-PEN was decreased compared to the CVCC3701. Virulence test in mice indicated that the LD50 of CVCC3701 before induction and CVCC3701-PEN after induction were 5.45 × 106 and 5.82 × 108 CFU/ml, respectively. Compared with the untreated bacteria, the bacterial virulence was reduced 1.1 × 102 times after resistance induction. In conclusion, S. dysgalactiae (CVCC3701) is a susceptible strain of drug resistance to antibiotics, in vitro-induced drug resistance reduced the virulence of CVCC3701, but the virulence is still existing and also could result in the death of mice. For public health safety, it must be alert to the emergence of drug resistance of Streptococcus in animal production.
Collapse
Affiliation(s)
- Yue‐Xia Ding
- College of Coastal Agricultural SciencesGuangdong Ocean UniversityZhanjiangPR China
- Department of Veterinary Pharmacology & ToxicologyCollege of Veterinary MedicineInner Mongolia Agricultural UniversityHohhotPR China
- Laboratory of Clinical Diagnosis and Treatment Techniques for Animal DiseaseMinistry of AgricultureHohhotPR China
| | - Qun Wu
- Research Institute of Agricultural MachineryChinese Academy of Tropical Agricultural SciencesZhanjiangPR China
| | - Yan Guo
- College of Coastal Agricultural SciencesGuangdong Ocean UniversityZhanjiangPR China
| | - Man Li
- Department of Veterinary Pharmacology & ToxicologyCollege of Veterinary MedicineInner Mongolia Agricultural UniversityHohhotPR China
- Laboratory of Clinical Diagnosis and Treatment Techniques for Animal DiseaseMinistry of AgricultureHohhotPR China
| | - Pei‐Feng Li
- Department of Veterinary Pharmacology & ToxicologyCollege of Veterinary MedicineInner Mongolia Agricultural UniversityHohhotPR China
- Laboratory of Clinical Diagnosis and Treatment Techniques for Animal DiseaseMinistry of AgricultureHohhotPR China
| | - Yi Ma
- College of Coastal Agricultural SciencesGuangdong Ocean UniversityZhanjiangPR China
- Maoming BranchGuangdong Laboratory for Lingnan Modern AgricultureMaomingPR China
| | - Wen‐Chao Liu
- College of Coastal Agricultural SciencesGuangdong Ocean UniversityZhanjiangPR China
| |
Collapse
|
36
|
Hernandez L, Bottini E, Cadona J, Cacciato C, Monteavaro C, Bustamante A, Sanso AM. Multidrug Resistance and Molecular Characterization of Streptococcus agalactiae Isolates From Dairy Cattle With Mastitis. Front Cell Infect Microbiol 2021; 11:647324. [PMID: 33996629 PMCID: PMC8120232 DOI: 10.3389/fcimb.2021.647324] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/09/2021] [Indexed: 11/28/2022] Open
Abstract
Streptococcus agalactiae is a pathogen-associated to bovine mastitis, a health disorder responsible for significant economic losses in the dairy industry. Antimicrobial therapy remains the main strategy for the control of this bacterium in dairy herds and human In order to get insight on molecular characteristics of S. agalactiae strains circulating among Argentinean cattle with mastitis, we received 1500 samples from 56 dairy farms between 2016 and 2019. We recovered 56 S. agalactiae isolates and characterized them in relation to serotypes, virulence genes, and antimicrobial susceptibility. Serotypes III and II were the most prevalent ones (46% and 41%, respectively), followed by Ia (7%). In relation to the 13 virulence genes screened in this study, the genes spb1, hylB, cylE, and PI-2b were present in all the isolates, meanwhile, bca, cpsA, and rib were detected in different frequencies, 36%, 96%, and 59%, respectively. On the other hand, bac, hvgA, lmb, PI-1, PI-2a, and scpB genes could not be detected in any of the isolates. Disk diffusion method against a panel of eight antimicrobial agents showed an important number of strains resistant simultaneously to five antibiotics. We also detected several resistance-encoding genes, tet(M), tet(O), ermB, aphA3, and lnu(B) (9%, 50%, 32%, 32%, and 5%, respectively). The results here presented are the first molecular data on S. agalactiae isolates causing bovine mastitis in Argentina and provide a foundation for the development of diagnostic, prophylactic, and therapeutic methods, including the perspective of a vaccine.
Collapse
Affiliation(s)
- Luciana Hernandez
- Laboratorio de Inmunoquímica y Biotecnología, CIVETAN (CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Argentina
| | - Enriqueta Bottini
- Laboratorio de Microbiología Clínica y Experimental, CIVETAN (CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Argentina
| | - Jimena Cadona
- Laboratorio de Inmunoquímica y Biotecnología, CIVETAN (CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Argentina
| | - Claudio Cacciato
- Laboratorio de Microbiología Clínica y Experimental, CIVETAN (CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Argentina
| | - Cristina Monteavaro
- Laboratorio de Microbiología Clínica y Experimental, CIVETAN (CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Argentina
| | - Ana Bustamante
- Laboratorio de Inmunoquímica y Biotecnología, CIVETAN (CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Argentina
| | - Andrea Mariel Sanso
- Laboratorio de Inmunoquímica y Biotecnología, CIVETAN (CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Argentina
| |
Collapse
|
37
|
van Sorge NM, Bonsor DA, Deng L, Lindahl E, Schmitt V, Lyndin M, Schmidt A, Nilsson OR, Brizuela J, Boero E, Sundberg EJ, van Strijp JAG, Doran KS, Singer BB, Lindahl G, McCarthy AJ. Bacterial protein domains with a novel Ig-like fold target human CEACAM receptors. EMBO J 2021; 40:e106103. [PMID: 33522633 PMCID: PMC8013792 DOI: 10.15252/embj.2020106103] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 01/19/2023] Open
Abstract
Streptococcus agalactiae, also known as group B Streptococcus (GBS), is the major cause of neonatal sepsis in humans. A critical step to infection is adhesion of bacteria to epithelial surfaces. GBS adhesins have been identified to bind extracellular matrix components and cellular receptors. However, several putative adhesins have no host binding partner characterised. We report here that surface-expressed β protein of GBS binds to human CEACAM1 and CEACAM5 receptors. A crystal structure of the complex showed that an IgSF domain in β represents a novel Ig-fold subtype called IgI3, in which unique features allow binding to CEACAM1. Bioinformatic assessment revealed that this newly identified IgI3 fold is not exclusively present in GBS but is predicted to be present in adhesins from other clinically important human pathogens. In agreement with this prediction, we found that CEACAM1 binds to an IgI3 domain found in an adhesin from a different streptococcal species. Overall, our results indicate that the IgI3 fold could provide a broadly applied mechanism for bacteria to target CEACAMs.
Collapse
Affiliation(s)
- Nina M van Sorge
- Department of Medical MicrobiologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
- Present address:
Department of Medical Microbiology,Infection Prevention and Netherlands Reference Laboratory for Bacterial MeningitisAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Daniel A Bonsor
- Institute of Human VirologyUniversity of Maryland School of MedicineUniversity of MarylandBaltimoreMDUSA
| | - Liwen Deng
- Department of Immunology & MicrobiologyUniversity of Colorado Anschutz Medical CampusAuroraCOUSA
| | - Erik Lindahl
- Department of Biochemistry and BiophysicsScience for Life LaboratoryStockholm UniversityStockholmSweden
| | - Verena Schmitt
- Institute of AnatomyMedical Faculty, University Duisburg‐EssenEssenGermany
| | - Mykola Lyndin
- Institute of AnatomyMedical Faculty, University Duisburg‐EssenEssenGermany
- Department of PathologySumy State UniversitySumyUkraine
| | - Alexej Schmidt
- Department of Medical BiosciencesUmeå UniversityPathology, UmeåSweden
| | - Olof R Nilsson
- Department of Laboratory MedicineDivision of Medical MicrobiologyLund UniversityLundSweden
| | - Jaime Brizuela
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology & InfectionImperial College LondonLondonUK
| | - Elena Boero
- Department of Medical MicrobiologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Eric J Sundberg
- Institute of Human VirologyUniversity of Maryland School of MedicineUniversity of MarylandBaltimoreMDUSA
- Department of BiochemistryEmory University School of MedicineAtlantaGAUSA
| | - Jos A G van Strijp
- Department of Medical MicrobiologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Kelly S Doran
- Department of Immunology & MicrobiologyUniversity of Colorado Anschutz Medical CampusAuroraCOUSA
| | - Bernhard B Singer
- Institute of AnatomyMedical Faculty, University Duisburg‐EssenEssenGermany
| | - Gunnar Lindahl
- Department of Laboratory MedicineDivision of Medical MicrobiologyLund UniversityLundSweden
- Department of ChemistryDivision of Applied MicrobiologyLund UniversityLundSweden
| | - Alex J McCarthy
- Department of Medical MicrobiologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology & InfectionImperial College LondonLondonUK
| |
Collapse
|
38
|
Atopobium vaginae and Prevotella bivia Are Able to Incorporate and Influence Gene Expression in a Pre-Formed Gardnerella vaginalis Biofilm. Pathogens 2021; 10:pathogens10020247. [PMID: 33672647 PMCID: PMC7924186 DOI: 10.3390/pathogens10020247] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Bacterial vaginosis (BV) is associated with a highly structured polymicrobial biofilm on the vaginal epithelium where Gardnerella species presumably play a pivotal role. Gardnerella vaginalis, Atopobium vaginae, and Prevotella bivia are vaginal pathogens detected during the early stages of incident BV. Herein, we aimed to analyze the impact of A. vaginae and P. bivia on a pre-established G. vaginalis biofilm using a novel in vitro triple-species biofilm model. Total biofilm biomass was determined by the crystal violet method. We also discriminated the bacterial populations in the biofilm and in its planktonic fraction by using PNA FISH. We further analyzed the influence of A. vaginae and P. bivia on the expression of key virulence genes of G. vaginalis by quantitative PCR. In our tested conditions, A. vaginae and P. bivia were able to incorporate into pre-established G. vaginalis biofilms but did not induce an increase in total biofilm biomass, when compared with 48-h G. vaginalis biofilms. However, they were able to significantly influence the expression of HMPREF0424_0821, a gene suggested to be associated with biofilm maintenance in G. vaginalis. This study suggests that microbial relationships between co-infecting bacteria can deeply affect the G. vaginalis biofilm, a crucial marker of BV.
Collapse
|
39
|
Mudzana R, Mavenyengwa RT, Gudza-Mugabe M. Analysis of virulence factors and antibiotic resistance genes in group B streptococcus from clinical samples. BMC Infect Dis 2021; 21:125. [PMID: 33509097 PMCID: PMC7844887 DOI: 10.1186/s12879-021-05820-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/19/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Streptococcus agalacticae (Group B Streptococcus, GBS) is one of the most important causative agents of serious infections among neonates. This study was carried out to identify antibiotic resistance and virulence genes associated with GBS isolated from pregnant women. METHODS A total of 43 GBS isolates were obtained from 420 vaginal samples collected from HIV positive and negative women who were 13-35 weeks pregnant attending Antenatal Care at Chitungwiza and Harare Central Hospitals in Zimbabwe. Identification tests of GBS isolates was done using standard bacteriological methods and molecular identification testing. Antibiotic susceptibility testing was done using the modified Kirby-Bauer method and E-test strips. The boiling method was used to extract DNA and Polymerase Chain Reaction (PCR) was used to screen for 13 genes. Data was fed into SPSS 24.0. RESULTS Nine distinct virulence gene profiles were identified and hly-scpB-bca-rib 37.2% (16/43) was common. The virulence genes identified were namely hly 97.8% (42/43), scpB 90.1% (39/43), bca 86.0% (37/43), rib 69.8% (30/43) and bac 11.6% (5/43). High resistance to tetracycline 97.7% (42/43) was reported followed by 72.1% (31/43) cefazolin, 69.8% (30/43) penicillin G, 58.1% (25/43) ampicillin, 55.8% (24/43) clindamycin, 46.5% (20/43) ceftriaxone, 34.9% (15/43) chloramphenicol, and 30.2% (13/43) for both erythromycin and vancomycin using disk diffusion. Antibiotic resistance genes among the resistant and intermediate-resistant isolates showed high frequencies for tetM 97.6% (41/42) and low frequencies for ermB 34.5% (10/29), ermTR 10.3% (3/29), mefA 3.4% (1/29), tetO 2.4% (1/42) and linB 0% (0/35). The atr housekeeping gene yielded 100% (43/43) positive results, whilst the mobile genetic element IS1548 yielded 9.3% (4/43). CONCLUSION The study showed high prevalence of hly, scpB, bca and rib virulence genes in S. agalactiae strains isolated from pregnant women. Tetracycline resistance was predominantly caused by the tetM gene, whilst macrolide resistance was predominantly due to the presence of erm methylase, with the ermB gene being more prevalent. Multi-drug resistance coupled with the recovery of resistant isolates to antimicrobial agents such as penicillins indicates the importance of GBS surveillance and susceptibility tests. It was also observed that in vitro phenotypic resistance is not always accurately predicted by resistance genotypes.
Collapse
Affiliation(s)
- Raymond Mudzana
- Department of Medical Microbiology, National Polio Laboratory, University of Zimbabwe College of Health Sciences, P. O. Box A178, Avondale, Harare, Zimbabwe
| | - Rooyen T. Mavenyengwa
- Department of Medical Microbiology, National Polio Laboratory, University of Zimbabwe College of Health Sciences, P. O. Box A178, Avondale, Harare, Zimbabwe
| | - Muchaneta Gudza-Mugabe
- Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Room No. 3.22 Falmouth Building, Anzio Road, Observatory, Cape Town, 7925 South Africa
| |
Collapse
|
40
|
Zhang H, Zhou T, Su L, Wang H, Zhang B, Su Y. Effects of srtA variation on phagocytosis resistance and immune response of Streptococcus equi. INFECTION GENETICS AND EVOLUTION 2021; 89:104732. [PMID: 33503504 DOI: 10.1016/j.meegid.2021.104732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/17/2020] [Accepted: 01/21/2021] [Indexed: 11/26/2022]
Abstract
Strangles, which is caused by Streptococcus equi subspecies equi (S. equi), is one of the most prevalent equine infectious diseases with worldwide distribution and leads to serious economic loss in the horse industry. Sortase A (srtA) is a transpeptidase that anchors multiple virulence-associated surface proteins to the cell surface of S. equi. srtA plays a major role in S. equi infection and colonization of the host cell. In this study, we aimed to investigate the effects of srtA mutation on the phagocytic activity and immunogenicity of S. equi. The point-mutated recombinant sortases, including srtA-HT1112 (I88V), srtA-5012 (R147G), and srtA-ZZM17 (control), were expressed, purified, and used to immunize the mouse models. Phagocytic activity was assessed using equine polymorphonuclear cells, whereas opsonophagocytic function and adherence inhibition were measured using the antiserum of these mutants. Mouse serum antibody, bacterial load, and weight gain were also measured. The srtA-HT1112 (I88V) mutant showed significantly enhanced antiphagocytic capability, and its antiserum exhibited increased adherence inhibition activity. In addition, the srtA-HT1112 (I88V) mutant presented the highest lung bacterial load and lowest protection rate (50%) after the challenge with S. equi ZZM17. The srtA-5012 (R147G) mutant exhibited a high IgG2a level and protection rate (62.5%-75%) and the lowest lung bacterial load. These results indicate that the I88V mutation is associated with a high antiphagocytic activity, whereas R147G mutation is associated with the decreased lung bacterial load. Our findings may be useful for the evaluation and development of vaccines.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, China
| | - Tingting Zhou
- Department of Microbiology and Immunology, College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, China
| | - Lining Su
- Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Hao Wang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, China
| | - Baojiang Zhang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, China
| | - Yan Su
- Department of Microbiology and Immunology, College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, China.
| |
Collapse
|
41
|
Bobadilla FJ, Novosak MG, Cortese IJ, Delgado OD, Laczeski ME. Prevalence, serotypes and virulence genes of Streptococcus agalactiae isolated from pregnant women with 35-37 weeks of gestation. BMC Infect Dis 2021; 21:73. [PMID: 33446117 PMCID: PMC7807883 DOI: 10.1186/s12879-020-05603-5] [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] [Received: 05/15/2020] [Accepted: 11/09/2020] [Indexed: 01/31/2023] Open
Abstract
Background In pregnant women Streptococcus agalactiae (GBS) can be transmitted to newborn causing severe infections. It is classified into 10 serotypes (Ia, Ib, II-IX). The severity of neonatal disease is determined by the capsular serotype and virulence factors such as the polysaccharide capsule, encoded by the cps gene, protein C, which includes the Cα surface proteins (bca gene), Rib (rib gene) and Cβ (bac gene); the proteins Lmb (lmb gene), FbsB (fbsB gene), FbsA (fbsA gene), the cyl operon encoding a β-hemolysin (hylB gene), the CAMP factor (cfb gene) and the C5a peptidase (scpB gene). The aim of this work was to determine the degree of GBS colonization in pregnant women, the serotypes distribution and to investigate virulence-associated genes. Methods We worked with 3480 samples of vagino-rectal swabs of women with 35–37 weeks of gestation. The identification of the strains was carried out using conventional biochemical tests and group confirmatory serology using a commercial latex particle agglutination kit. Two hundred GBS strains were selected. Their serotype was determined by agglutination tests. The monoplex PCR technique was used to investigate nine virulence-associated genes (cps, bca, rib, bac, lmb, fbsB, fbsA, hylB and scpB). Results The maternal colonization was 9.09%. The serotypes found were: Ia (33.50%), III (19.00%), Ib (15.50%), II (14.00%), V (7.00%) and IX (5.50%). 5.50% of strains were found to be non-serotypeable (NT). The nine virulence genes investigated were detected simultaneously in 36.50% of the strains. The genes that were most frequently detected were scpB (100.00%), fbsA (100.00%), fbsB (100.00%), cylB (95.00%), lmb (94.00%) and bca (87.50%). We found associations between serotype and genes bac (p = 0.003), cylB (p = 0.02), rib (p = 0.01) and lmb (p < 0.001). Conclusions The frequency of vaginal-rectal colonization, serotypes distribution and associated virulence genes, varies widely among geographical areas. Therefore, epidemiological surveillance is necessary to provide data to guide decision-making and planning of prevention and control strategies.
Collapse
Affiliation(s)
- Fernando J Bobadilla
- Facultad de Ciencias Exactas, Químicas y Naturales (FCEQyN), Departamento de Microbiología, Universidad Nacional de Misiones (UNaM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, Argentina. .,Facultad de Ciencias Exactas, Químicas y Naturales (FCEQyN), Instituto de Biotecnología de Misiones (InBioMis), Universidad Nacional de Misiones (UNaM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, Argentina.
| | - Marina G Novosak
- Facultad de Ciencias Exactas, Químicas y Naturales (FCEQyN), Departamento de Microbiología, Universidad Nacional de Misiones (UNaM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, Argentina.,Facultad de Ciencias Exactas, Químicas y Naturales (FCEQyN), Instituto de Biotecnología de Misiones (InBioMis), Universidad Nacional de Misiones (UNaM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, Argentina
| | - Iliana J Cortese
- Facultad de Ciencias Exactas, Químicas y Naturales (FCEQyN), Instituto de Biotecnología de Misiones (InBioMis), Universidad Nacional de Misiones (UNaM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, Argentina
| | - Osvaldo D Delgado
- Facultad de Ciencias Exactas y Naturales (FACEN), Universidad Nacional de Catamarca (UNCa), Centro de Investigación y Transferencia Catamarca (CITCA-CONICET), San Fernando del Valle de Catamarca, Argentina
| | - Margarita E Laczeski
- Facultad de Ciencias Exactas, Químicas y Naturales (FCEQyN), Departamento de Microbiología, Universidad Nacional de Misiones (UNaM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, Argentina.,Facultad de Ciencias Exactas, Químicas y Naturales (FCEQyN), Instituto de Biotecnología de Misiones (InBioMis), Universidad Nacional de Misiones (UNaM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Posadas, Argentina
| |
Collapse
|
42
|
Perme T, Golparian D, Bombek Ihan M, Rojnik A, Lučovnik M, Kornhauser Cerar L, Fister P, Lozar Krivec J, Grosek Š, Ihan A, Jeverica S, Unemo M. Genomic and phenotypic characterisation of invasive neonatal and colonising group B Streptococcus isolates from Slovenia, 2001-2018. BMC Infect Dis 2020; 20:958. [PMID: 33327946 PMCID: PMC7739447 DOI: 10.1186/s12879-020-05599-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 11/09/2020] [Indexed: 11/30/2022] Open
Abstract
Background Group B Streptococcus (GBS) is the leading cause of invasive neonatal disease in the industrialized world. We aimed to genomically and phenotypically characterise invasive GBS isolates in Slovenia from 2001 to 2018 and contemporary colonising GBS isolates from screening cultures in 2018. Methods GBS isolates from 101 patients (invasive isolates) and 70 pregnant women (colonising isolates) were analysed. Basic clinical characteristics of the patients were collected from medical records. Antimicrobial susceptibility and phenotypic capsular serotype were determined. Whole-genome sequencing was performed to assign multilocus sequence types (STs), clonal complexes (CCs), pathogenicity/virulence factors, including capsular genotypes, and genome-based phylogeny. Results Among invasive neonatal disease patients, 42.6% (n = 43) were females, 41.5% (n = 39/94) were from preterm deliveries (< 37 weeks gestation), and 41.6% (n = 42) had early-onset disease (EOD). All isolates were susceptible to benzylpenicillin with low minimum inhibitory concentrations (MICs; ≤0.125 mg/L). Overall, 7 serotypes were identified (Ia, Ib, II-V and VIII); serotype III being the most prevalent (59.6%). Twenty-eight MLST STs were detected that clustered into 6 CCs. CC-17 was the most common CC overall (53.2%), as well as among invasive (67.3%) and non-invasive (32.9%) isolates (p < 0.001). CC-17 was more common among patients with late-onset disease (LOD) (81.4%) compared to EOD (47.6%) (p < 0.001). The prevalence of other CCs was 12.9% (CC-23), 11.1% (CC-12), 10.5% (CC-1), 8.2% (CC-19), and 1.8% (CC-498). Of all isolates, 2.3% were singletons. Conclusions A high prevalence of hypervirulent CC-17 isolates, with low genomic diversity and characteristic profile of pathogenicity/virulence factors, was detected among invasive neonatal and colonising GBS isolates from pregnant women in Slovenia. This is the first genomic characterisation of GBS isolates in Slovenia and provides valuable microbiological and genomic baseline data regarding the invasive and colonising GBS population nationally. Continuous genomic surveillance of GBS infections is crucial to analyse the impact of IND prevention strategies on the population structure of GBS locally, nationally, and internationally. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-020-05599-y.
Collapse
Affiliation(s)
- Tina Perme
- Department of Perinatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Daniel Golparian
- WHO Collaborating Centre for Gonorrhoea and other STIs, National Reference Laboratory for STIs, Department of Laboratory Medicine, Microbiology, Örebro University, SE-70185, Örebro, Sweden
| | - Maja Bombek Ihan
- National Laboratory for Health, Environment and Food, Maribor, Slovenia
| | - Andrej Rojnik
- National Laboratory for Health, Environment and Food, Maribor, Slovenia
| | - Miha Lučovnik
- Department of Perinatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | - Petja Fister
- Department of Neonatology, Division of Pediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Jana Lozar Krivec
- Department of Neonatology, Division of Pediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Štefan Grosek
- Department of Perinatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Chair of Pediatrics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Institute for Microbiology and Immunology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Samo Jeverica
- National Laboratory for Health, Environment and Food, Maribor, Slovenia
| | - Magnus Unemo
- WHO Collaborating Centre for Gonorrhoea and other STIs, National Reference Laboratory for STIs, Department of Laboratory Medicine, Microbiology, Örebro University, SE-70185, Örebro, Sweden.
| |
Collapse
|
43
|
Benmimoun B, Papastefanaki F, Périchon B, Segklia K, Roby N, Miriagou V, Schmitt C, Dramsi S, Matsas R, Spéder P. An original infection model identifies host lipoprotein import as a route for blood-brain barrier crossing. Nat Commun 2020; 11:6106. [PMID: 33257684 PMCID: PMC7704634 DOI: 10.1038/s41467-020-19826-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Pathogens able to cross the blood-brain barrier (BBB) induce long-term neurological sequelae and death. Understanding how neurotropic pathogens bypass this strong physiological barrier is a prerequisite to devise therapeutic strategies. Here we propose an innovative model of infection in the developing Drosophila brain, combining whole brain explants with in vivo systemic infection. We find that several mammalian pathogens are able to cross the Drosophila BBB, including Group B Streptococcus (GBS). Amongst GBS surface components, lipoproteins, and in particular the B leucine-rich Blr, are important for BBB crossing and virulence in Drosophila. Further, we identify (V)LDL receptor LpR2, expressed in the BBB, as a host receptor for Blr, allowing GBS translocation through endocytosis. Finally, we show that Blr is required for BBB crossing and pathogenicity in a murine model of infection. Our results demonstrate the potential of Drosophila for studying BBB crossing by pathogens and identify a new mechanism by which pathogens exploit the machinery of host barriers to generate brain infection.
Collapse
Affiliation(s)
- Billel Benmimoun
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France
| | - Florentia Papastefanaki
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Bruno Périchon
- Unité de Biologie des Bactéries Pathogènes à Gram-positif, Institut Pasteur, CNRS, UMR 2001, Paris, France
| | - Katerina Segklia
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Nicolas Roby
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France
| | - Vivi Miriagou
- Laboratory of Bacteriology, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Christine Schmitt
- Ultrastructure UTechS Ultrastructural Bioimaging Platform, Institut Pasteur, Paris, France
| | - Shaynoor Dramsi
- Unité de Biologie des Bactéries Pathogènes à Gram-positif, Institut Pasteur, CNRS, UMR 2001, Paris, France
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Pauline Spéder
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France.
| |
Collapse
|
44
|
Subdominance in Antibody Responses: Implications for Vaccine Development. Microbiol Mol Biol Rev 2020; 85:85/1/e00078-20. [PMID: 33239435 DOI: 10.1128/mmbr.00078-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Vaccines work primarily by eliciting antibodies, even when recovery from natural infection depends on cellular immunity. Large efforts have therefore been made to identify microbial antigens that elicit protective antibodies, but these endeavors have encountered major difficulties, as witnessed by the lack of vaccines against many pathogens. This review summarizes accumulating evidence that subdominant protein regions, i.e., surface-exposed regions that elicit relatively weak antibody responses, are of particular interest for vaccine development. This concept may seem counterintuitive, but subdominance may represent an immune evasion mechanism, implying that the corresponding region potentially is a key target for protective immunity. Following a presentation of the concepts of immunodominance and subdominance, the review will present work on subdominant regions in several major human pathogens: the protozoan Plasmodium falciparum, two species of pathogenic streptococci, and the dengue and influenza viruses. Later sections are devoted to the molecular basis of subdominance, its potential role in immune evasion, and general implications for vaccine development. Special emphasis will be placed on the fact that a whole surface-exposed protein domain can be subdominant, as demonstrated for all of the pathogens described here. Overall, the available data indicate that subdominant protein regions are of much interest for vaccine development, not least in bacterial and protozoal systems, for which antibody subdominance remains largely unexplored.
Collapse
|
45
|
Identification of Zinc-Dependent Mechanisms Used by Group B Streptococcus To Overcome Calprotectin-Mediated Stress. mBio 2020; 11:mBio.02302-20. [PMID: 33173000 PMCID: PMC7667036 DOI: 10.1128/mbio.02302-20] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Group B Streptococcus (GBS) asymptomatically colonizes the female reproductive tract but is a common causative agent of meningitis. GBS meningitis is characterized by extensive infiltration of neutrophils carrying high concentrations of calprotectin, a metal chelator. To persist within inflammatory sites and cause invasive disease, GBS must circumvent host starvation attempts. Here, we identified global requirements for GBS survival during calprotectin challenge, including known and putative systems involved in metal ion transport. We characterized the role of zinc import in tolerating calprotectin stress in vitro and in a mouse model of infection. We observed that a global zinc uptake mutant was less virulent than the parental GBS strain and found calprotectin knockout mice to be equally susceptible to infection by wild-type (WT) and mutant strains. These findings suggest that calprotectin production at the site of infection results in a zinc-limited environment and reveals the importance of GBS metal homeostasis to invasive disease. Nutritional immunity is an elegant host mechanism used to starve invading pathogens of necessary nutrient metals. Calprotectin, a metal-binding protein, is produced abundantly by neutrophils and is found in high concentrations within inflammatory sites during infection. Group B Streptococcus (GBS) colonizes the gastrointestinal and female reproductive tracts and is commonly associated with severe invasive infections in newborns such as pneumonia, sepsis, and meningitis. Although GBS infections induce robust neutrophil recruitment and inflammation, the dynamics of GBS and calprotectin interactions remain unknown. Here, we demonstrate that disease and colonizing isolate strains exhibit susceptibility to metal starvation by calprotectin. We constructed a mariner transposon (Krmit) mutant library in GBS and identified 258 genes that contribute to surviving calprotectin stress. Nearly 20% of all underrepresented mutants following treatment with calprotectin are predicted metal transporters, including known zinc systems. As calprotectin binds zinc with picomolar affinity, we investigated the contribution of GBS zinc uptake to overcoming calprotectin-imposed starvation. Quantitative reverse transcriptase PCR (qRT-PCR) revealed a significant upregulation of genes encoding zinc-binding proteins, adcA, adcAII, and lmb, following calprotectin exposure, while growth in calprotectin revealed a significant defect for a global zinc acquisition mutant (ΔadcAΔadcAIIΔlmb) compared to growth of the GBS wild-type (WT) strain. Furthermore, mice challenged with the ΔadcAΔadcAIIΔlmb mutant exhibited decreased mortality and significantly reduced bacterial burden in the brain compared to mice infected with WT GBS; this difference was abrogated in calprotectin knockout mice. Collectively, these data suggest that GBS zinc transport machinery is important for combatting zinc chelation by calprotectin and establishing invasive disease.
Collapse
|
46
|
Distribution of Streptococcus agalactiae Among Iranian Women from 1992 to 2018: A Systematic Review and Meta-Analysis. Jundishapur J Microbiol 2020. [DOI: 10.5812/jjm.102314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Context: Group B Streptococcus has the capacity of being colonized in the rectovaginal organ of women and causes infections in a mother and her fetus, thereby leading to neonatal diseases. Evidence Acquisition: The aim of this review was to summarize all of the relevant articles published to highlight the prevalence of group B Streptococcus colonization or infection in different regions of Iran. A systematic literature review was conducted by searching PubMed, Scopus, Web of Science (ISI), ScienceDirect, Google Scholar, and domestic databases for papers published in English or Persian from 1992 up to July 2019, concerning the prevalence of group B Streptococcus among Iranian women. All information regarding year, location of cases, frequency, author’s name, date of publication, participants, pregnancy period, sampling, and quality assessment were recorded. Summary effects were derived using the random effects model. Results: Among 61 suitable papers, data revealed that 36,807 cases of pregnant and non-pregnant women had been tested for group B Streptococcus during 1992-2018. Overall, 11.9% of pregnant and 5.3% of non-pregnant women were positive. Further results were as follows: vaginal, recto-vaginal, rectal, and endocervical colonization rates were 12.9%, 9.7%, 18.5%, and 3.7%, respectively. Group B Streptococcus incidence was the highest in Sanandaj (61.5%), and the lowest in Tabriz (1.8%). Conclusions: Although the prevalence distribution in Iran seems to be acceptable, more investigations are needed to represent the real incidence of group B Streptococcus around the country. In addition, a program with a standard lab technique is needed to screen pregnant women for further treatment before birth.
Collapse
|
47
|
Genome-Wide Assessment of Streptococcus agalactiae Genes Required for Survival in Human Whole Blood and Plasma. Infect Immun 2020; 88:IAI.00357-20. [PMID: 32747604 DOI: 10.1128/iai.00357-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/26/2020] [Indexed: 12/17/2022] Open
Abstract
Streptococcus agalactiae (group B streptococcus, or GBS) is a common cause of bacteremia and sepsis in newborns, pregnant women, and immunocompromised patients. The molecular mechanisms used by GBS to survive and proliferate in blood are not well understood. Here, using a highly virulent GBS strain and transposon-directed insertion site sequencing (TraDIS), we performed genome-wide screens to discover novel GBS genes required for bacterial survival in human whole blood and plasma. The screen identified 85 and 41 genes that are required for GBS growth in whole blood and plasma, respectively. A common set of 29 genes was required in both whole blood and plasma. Targeted gene deletion confirmed that (i) genes encoding methionine transporter (metP) and manganese transporter (mtsA) are crucial for GBS survival in whole blood and plasma, (ii) gene W903_1820, encoding a small multidrug export family protein, contributes significantly to GBS survival in whole blood, (iii) the shikimate pathway gene aroA is essential for GBS growth in whole blood and plasma, and (iv) deletion of srr1, encoding a fibrinogen-binding adhesin, increases GBS survival in whole blood. Our findings provide new insight into the GBS-host interactions in human blood.
Collapse
|
48
|
Takahashi T, Maeda T, Lee S, Lee DH, Kim S. Clonal Distribution of Clindamycin-Resistant Erythromycin-Susceptible (CRES) Streptococcus agalactiae in Korea Based on Whole Genome Sequences. Ann Lab Med 2020; 40:370-381. [PMID: 32311850 PMCID: PMC7169627 DOI: 10.3343/alm.2020.40.5.370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/17/2020] [Accepted: 03/27/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The clindamycin-resistant erythromycin-susceptible (CRES) phenotype is rare in Streptococcus agalactiae (group B streptococci). We aimed to determine the molecular characteristics of CRES S. agalactiae using whole genome sequencing (WGS). METHODS Sixty-six S. agalactiae isolates obtained from blood (N=26), cerebrospinal fluid (N=10), urine (N=17), and vaginal discharge (N=13) between 2010 and 2017 in Korea were subjected to WGS. Based on the WGS data, we analyzed antimicrobial resistance (AMR) determinants, sequence types (STs), capsular polysaccharide (CPS) genotypes, and virulence gene profiles, and constructed a phylogenetic tree. We included the clindamycin-susceptible erythromycin-resistant (CSER) phenotype for comparison. RESULTS We identified seven CRES S. agalactiae isolates from urine (N=5) and vaginal discharge (N=2) collected between 2010 and 2011. All CRES isolates harbored AMR determinants of lnu(B), lsa(E), and aac(6')-aph(2″), revealed ST19 and CPS genotype III, and had a virulence gene profile of rib-lmb-cylE. Phylogenetic tree analysis revealed that all CRES isolates belonged to the same cluster, suggesting a clonal distribution. In contrast, seven CSER isolates showed a diverse distribution and clustered separately from the CRES isolates. CONCLUSIONS CRES isolates collected between 2010 and 2011 showed a unique cluster with ST19 and CPS genotype III in Korea. This is the first report on WGS-based characteristics of S. agalactiae in Korea.
Collapse
Affiliation(s)
- Takashi Takahashi
- Laboratory of Infectious Diseases, Graduate School of Infection Control Sciences & Kitasato Institute for Life Sciences, Kitasato University, Tokyo,
Japan
| | - Takahiro Maeda
- Laboratory of Infectious Diseases, Graduate School of Infection Control Sciences & Kitasato Institute for Life Sciences, Kitasato University, Tokyo,
Japan
| | - Seungjun Lee
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon,
Korea
| | - Dong-Hyun Lee
- Department of Laboratory Medicine, Gyeongsang National University Hospital, Jinju,
Korea
| | - Sunjoo Kim
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon,
Korea
- Department of Laboratory Medicine, Gyeongsang National University College of Medicine, Institute of Health Sciences, Jinju,
Korea
| |
Collapse
|
49
|
Quorum Sensing-Linked agrA Expression by Ethno-Synthesized Gold Nanoparticles in Tilapia Streptococcus agalactiae Biofilm Formation. BIONANOSCIENCE 2020. [DOI: 10.1007/s12668-020-00758-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
50
|
Syed S, Viazmina L, Mager R, Meri S, Haapasalo K. Streptococci and the complement system: interplay during infection, inflammation and autoimmunity. FEBS Lett 2020; 594:2570-2585. [PMID: 32594520 DOI: 10.1002/1873-3468.13872] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 11/09/2022]
Abstract
Streptococci are a broad group of Gram-positive bacteria. This genus includes various human pathogens causing significant morbidity and mortality. Two of the most important human pathogens are Streptococcus pneumoniae (pneumococcus) and Streptococcus pyogenes (group A streptococcus or GAS). Streptococcal pathogens have evolved to express virulence factors that enable them to evade complement-mediated attack. These include factor H-binding M (S. pyogenes) and pneumococcal surface protein C (PspC) (S. pneumoniae) proteins. In addition, S. pyogenes and S. pneumoniae express cytolysins (streptolysin and pneumolysin), which are able to destroy host cells. Sometimes, the interplay between streptococci, the complement, and antistreptococcal immunity may lead to an excessive inflammatory response or autoimmune disease. Understanding the fundamental role of the complement system in microbial clearance and the bacterial escape mechanisms is of paramount importance for understanding microbial virulence, in general, and, the conversion of commensals to pathogens, more specifically. Such insights may help to identify novel antibiotic and vaccine targets in bacterial pathogens to counter their growing resistance to commonly used antibiotics.
Collapse
Affiliation(s)
- Shahan Syed
- Department of Bacteriology and Immunology, University of Helsinki, Finland
| | - Larisa Viazmina
- Department of Bacteriology and Immunology, University of Helsinki, Finland
| | | | - Seppo Meri
- Department of Bacteriology and Immunology, University of Helsinki, Finland.,Humanitas University, Milano, Italy
| | - Karita Haapasalo
- Department of Bacteriology and Immunology, University of Helsinki, Finland
| |
Collapse
|