1
|
Crestani C, Forde TL, Bell J, Lycett SJ, Oliveira LMA, Pinto TCA, Cobo-Ángel CG, Ceballos-Márquez A, Phuoc NN, Sirimanapong W, Chen SL, Jamrozy D, Bentley SD, Fontaine M, Zadoks RN. Genomic and functional determinants of host spectrum in Group B Streptococcus. PLoS Pathog 2024; 20:e1012400. [PMID: 39133742 PMCID: PMC11341095 DOI: 10.1371/journal.ppat.1012400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/22/2024] [Accepted: 07/08/2024] [Indexed: 08/24/2024] Open
Abstract
Group B Streptococcus (GBS) is a major human and animal pathogen that threatens public health and food security. Spill-over and spill-back between host species is possible due to adaptation and amplification of GBS in new niches but the evolutionary and functional mechanisms underpinning those phenomena are poorly known. Based on analysis of 1,254 curated genomes from all major GBS host species and six continents, we found that the global GBS population comprises host-generalist, host-adapted and host-restricted sublineages, which are found across host groups, preferentially within one host group, or exclusively within one host group, respectively, and show distinct levels of recombination. Strikingly, the association of GBS genomes with the three major host groups (humans, cattle, fish) is driven by a single accessory gene cluster per host, regardless of sublineage or the breadth of host spectrum. Moreover, those gene clusters are shared with other streptococcal species occupying the same niche and are functionally relevant for host tropism. Our findings demonstrate (1) the heterogeneity of genome plasticity within a bacterial species of public health importance, enabling the identification of high-risk clones; (2) the contribution of inter-species gene transmission to the evolution of GBS; and (3) the importance of considering the role of animal hosts, and the accessory gene pool associated with their microbiota, in the evolution of multi-host bacterial pathogens. Collectively, these phenomena may explain the adaptation and clonal expansion of GBS in animal reservoirs and the risk of spill-over and spill-back between animals and humans.
Collapse
Affiliation(s)
- Chiara Crestani
- Institute of Biodiversity, Animal Health & Comparative Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Taya L. Forde
- Institute of Biodiversity, Animal Health & Comparative Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - John Bell
- Moredun Research Institute, Penicuik, Scotland, United Kingdom
| | - Samantha J. Lycett
- The Roslin Institute, University of Edinburgh, Midlothian, Scotland, United Kingdom
| | - Laura M. A. Oliveira
- Instituto de Microbiologia Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, State of Rio de Janeiro, Brazil
| | - Tatiana C. A. Pinto
- Instituto de Microbiologia Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, State of Rio de Janeiro, Brazil
| | | | | | - Nguyen N. Phuoc
- Faculty of Fisheries, University of Agriculture and Forestry, Hue University, Hue, Vietnam
| | - Wanna Sirimanapong
- Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Swaine L. Chen
- Infectious Diseases Translational Research Programme, Department of Medicine, Division of Infectious Diseases, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Laboratory of Bacterial Genomics, Genome Institute of Singapore, Singapore
| | - Dorota Jamrozy
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, England, United Kingdom
| | - Stephen D. Bentley
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, England, United Kingdom
| | | | - Ruth N. Zadoks
- Institute of Biodiversity, Animal Health & Comparative Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom
- Moredun Research Institute, Penicuik, Scotland, United Kingdom
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Camden, NSW, Australia
| |
Collapse
|
2
|
de Oliveira LMA, Simões LC, Crestani C, Costa NS, Pantoja JCDF, Rabello RF, Teixeira LM, Khan UB, Bentley S, Jamrozy D, Pinto TDCA, Zadoks RN. Long-Term Co-Circulation of Host-Specialist and Host-Generalist Lineages of Group B Streptococcus in Brazilian Dairy Cattle with Heterogeneous Antimicrobial Resistance Profiles. Antibiotics (Basel) 2024; 13:389. [PMID: 38786118 PMCID: PMC11117364 DOI: 10.3390/antibiotics13050389] [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: 04/01/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Group B Streptococcus (GBS) is a major cause of contagious bovine mastitis (CBM) in Brazil. The GBS population is composed of host-generalist and host-specialist lineages, which may differ in antimicrobial resistance (AMR) and zoonotic potential, and the surveillance of bovine GBS is crucial to developing effective CBM control and prevention measures. Here, we investigated bovine GBS isolates (n = 156) collected in Brazil between 1987 and 2021 using phenotypic testing and whole-genome sequencing to uncover the molecular epidemiology of bovine GBS. Clonal complex (CC) 61/67 was the predominant clade in the 20th century; however, it was replaced by CC91, with which it shares a most common recent ancestor, in the 21st century, despite the higher prevalence of AMR in CC61/67 than in CC91, and high selection pressure for AMR from indiscriminate antimicrobial use in the Brazilian dairy industry. CC103 also emerged as a dominant CC in the 21st century, and a considerable proportion of herds had two or more GBS strains, suggesting poor biosecurity and within-herd evolution due to the chronic nature of CBM problems. The majority of bovine GBS belonged to serotype Ia or III, which was strongly correlated with CCs. Ninety-three isolates were resistant to tetracycline (≥8 μg/mL; tetO = 57, tetM = 34 or both = 2) and forty-four were resistant to erythromycin (2.0 to >4 μg/mL; ermA = 1, ermB = 38, mechanism unidentified n = 5). Only three isolates were non-susceptible to penicillin (≥8.0 μg/mL), providing opportunities for improved antimicrobial stewardship through the use of narrow-spectrum antimicrobials for the treatment of dairy cattle. The common bovine GBS clades detected in this study have rarely been reported in humans, suggesting limited risk of interspecies transmission of GBS in Brazil. This study provides new data to support improvements to CBM and AMR control, bovine GBS vaccine design, and the management of public health risks posed by bovine GBS in Brazil.
Collapse
Affiliation(s)
- Laura Maria Andrade de Oliveira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.C.S.); (N.S.C.); (L.M.T.); (T.d.C.A.P.)
| | - Leandro Correia Simões
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.C.S.); (N.S.C.); (L.M.T.); (T.d.C.A.P.)
| | | | - Natália Silva Costa
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.C.S.); (N.S.C.); (L.M.T.); (T.d.C.A.P.)
| | | | | | - Lucia Martins Teixeira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.C.S.); (N.S.C.); (L.M.T.); (T.d.C.A.P.)
| | - Uzma Basit Khan
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK; (U.B.K.); (S.B.); (D.J.)
| | - Stephen Bentley
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK; (U.B.K.); (S.B.); (D.J.)
| | - Dorota Jamrozy
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK; (U.B.K.); (S.B.); (D.J.)
| | - Tatiana de Castro Abreu Pinto
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.C.S.); (N.S.C.); (L.M.T.); (T.d.C.A.P.)
| | - Ruth N. Zadoks
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Camden, NSW 2570, Australia
| |
Collapse
|
3
|
Bernabeu M, Cabello-Yeves E, Flores E, Samarra A, Kimberley Summers J, Marina A, Collado MC. Role of vertical and horizontal microbial transmission of antimicrobial resistance genes in early life: insights from maternal-infant dyads. Curr Opin Microbiol 2024; 77:102424. [PMID: 38237429 DOI: 10.1016/j.mib.2023.102424] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 02/12/2024]
Abstract
Early life represents a critical window for metabolic, cognitive and immune system development, which is influenced by the maternal microbiome as well as the infant gut microbiome. Antibiotic exposure, mode of delivery and breastfeeding practices modulate the gut microbiome and the reservoir of antibiotic resistance genes (ARGs). Vertical and horizontal microbial gene transfer during early life and the mechanisms behind these transfers are being uncovered. In this review, we aim to provide an overview of the current knowledge on the transfer of antibiotic resistance in the mother-infant dyad through vertical and horizontal transmission and to highlight the main gaps and challenges in this area.
Collapse
Affiliation(s)
- Manuel Bernabeu
- Institute of Agrochemistry and Food Technology - National Research Council (IATA-CSIC), 46980 Valencia, Spain.
| | - Elena Cabello-Yeves
- Instituto de Biomedicina de Valencia-Consejo de Investigaciones Científicas (IBV-CSIC), CIBER de Enfermedades Raras (CIBERER), 46010 Valencia, Spain.
| | - Eduard Flores
- Institute of Agrochemistry and Food Technology - National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - Anna Samarra
- Institute of Agrochemistry and Food Technology - National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - Joanna Kimberley Summers
- Wellington Lab, School of Life Sciences, University of Warwick, CV4 7AL Coventry, United Kingdom
| | - Alberto Marina
- Instituto de Biomedicina de Valencia-Consejo de Investigaciones Científicas (IBV-CSIC), CIBER de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - M Carmen Collado
- Institute of Agrochemistry and Food Technology - National Research Council (IATA-CSIC), 46980 Valencia, Spain
| |
Collapse
|
4
|
Enany S, Tartor YH, Kishk RM, Gadallah AM, Ahmed E, Magdeldin S. Proteomics and metabolomics analyses of Streptococcus agalactiae isolates from human and animal sources. Sci Rep 2023; 13:20980. [PMID: 38017083 PMCID: PMC10684508 DOI: 10.1038/s41598-023-47976-y] [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: 05/21/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
Streptococcus agalactiae (S. agalactiae), group B Streptococcus (GBS), a major cause of infection in a wide variety of diseases, have been compared in different human and animal sources. We aimed to compare the bacterial proteome and metabolome profiles of human and animal S. agalactiae strains to delineate biological interactions relevant to infection. With the innovative advancement in mass spectrometry, a comparative result between both strains provided a solid impression of different responses to the host. For instance, stress-related proteins (Asp23/Gls24 family envelope stress response protein and heat shock protein 70), which play a role in the survival of GBS under extreme environmental conditions or during treatment, are highly expressed in human and animal strains. One human strain contains ꞵ-lactamase (serine hydrolase) and biofilm regulatory protein (lytR), which are important virulence regulators and potential targets for the design of novel antimicrobials. Another human strain contains the aminoglycosides-resistance bifunctional AAC/APH (A0A0U2QMQ5) protein, which confers resistance to almost all clinically used aminoglycosides. Fifteen different metabolites were annotated between the two groups. L-aspartic acid, ureidopropionic acid, adenosine monophosphate, L-tryptophan, and guanosine monophosphate were annotated at higher levels in human strains. Butyric acid, fumaric acid, isoleucine, leucine, and hippuric acid have been found in both human and animal strains. Certain metabolites were uniquely expressed in animal strains, with fold changes greater than 2. For example, putrescine modulates biofilm formation. Overall, this study provides biological insights into the substantial possible bacterial response reflected in its macromolecular production, either at the proteomic or metabolomic level.
Collapse
Affiliation(s)
- Shymaa Enany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
- Biomedical Research Department, Armed Force College of Medicine, Cairo, Egypt.
| | - Yasmine H Tartor
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Rania M Kishk
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Ahmed M Gadallah
- Department of Obstetrics and Gynecology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Eman Ahmed
- Proteomics and Metabolomics Unit, Department of Basic Research, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Unit, Department of Basic Research, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
5
|
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
|
6
|
How GBS Got Its Hump: Genomic Analysis of Group B Streptococcus from Camels Identifies Host Restriction as well as Mobile Genetic Elements Shared across Hosts and Pathogens. Pathogens 2022; 11:pathogens11091025. [PMID: 36145457 PMCID: PMC9504112 DOI: 10.3390/pathogens11091025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/23/2022] Open
Abstract
Group B Streptococcus (GBS) literature largely focuses on humans and neonatal disease, but GBS also affects numerous animals, with significant impacts on health and productivity. Spill-over events occur between humans and animals and may be followed by amplification and evolutionary adaptation in the new niche, including changes in the core or accessory genome content. Here, we describe GBS from one-humped camels (Camelus dromedarius), a relatively poorly studied GBS host of increasing importance for food security in arid regions. Genomic analysis shows that virtually all GBS from camels in East Africa belong to a monophyletic clade, sublineage (SL)609. Capsular types IV and VI, including a new variant of type IV, were over-represented compared to other host species. Two genomic islands with signatures of mobile elements contained most camel-associated genes, including genes for metal and carbohydrate utilisation. Lactose fermentation genes were associated with milk isolates, albeit at lower prevalence in camel than bovine GBS. The presence of a phage with high identity to Streptococcus pneumoniae and Streptococcus suis suggests lateral gene transfer between GBS and bacterial species that have not been described in camels. The evolution of camel GBS appears to combine host restriction with the sharing of accessory genome content across pathogen and host species.
Collapse
|
7
|
Zhou K, Xie L, Xu X, Sun J. Comparative Genomic Analysis of Type VII Secretion System in Streptococcus agalactiae Indicates Its Possible Sequence Type-Dependent Diversity. Front Cell Infect Microbiol 2022; 12:880943. [PMID: 35663471 PMCID: PMC9160427 DOI: 10.3389/fcimb.2022.880943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus agalactiae causes sepsis and meningitis in neonates, presenting substantial clinical challenges. Type VII secretion system (T7SS), an important secretion system identified in Mycobacterium sp. and Gram-positive bacteria, was recently characterized in S. agalactiae and considered to contribute to its virulence and pathogenesis. In the present study, 128 complete genomic sequences of S. agalactiae were retrieved from GenBank to build a public dataset, and their sequences, capsular types, and clonal complexes were determined. Polymerase chain reaction (PCR) screening and genomic sequencing were conducted in an additional clinical dataset. STs and capsular types were determined using PCR. Eleven different types of T7SS were detected with similarities in gene order but differences in gene content. Strains with incomplete T7SS or lack of T7SS were also identified. Deletion, insertion, and segmentation of T7SS might be related to insertion sequences. The genetic environment of T7SS in S. agalactiae was also investigated and different patterns were identified downstream the T7SS, which were related to the diversity of T7SS putative effectors. The T7SS demonstrated possible sequence type (ST)-dependent diversity in both datasets. This work elucidated detailed genetic characteristics of T7SS and its genetic environment in S. agalactiae and further identified its possible ST-dependent diversity, which gave a clue of its mode of transmission. Further investigations are required to elucidate the underlying mechanisms and its functions.
Collapse
Affiliation(s)
- Kaixin Zhou
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
| | - Lianyan Xie
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Clinical Microbiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaogang Xu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Xiaogang Xu, ; Jingyong Sun,
| | - Jingyong Sun
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Clinical Microbiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Xiaogang Xu, ; Jingyong Sun,
| |
Collapse
|
8
|
Han G, Zhang B, Luo Z, Lu B, Luo Z, Zhang J, Wang Y, Luo Y, Yang Z, Shen L, Yu S, Cao S, Yao X. Molecular typing and prevalence of antibiotic resistance and virulence genes in Streptococcus agalactiae isolated from Chinese dairy cows with clinical mastitis. PLoS One 2022; 17:e0268262. [PMID: 35522690 PMCID: PMC9075616 DOI: 10.1371/journal.pone.0268262] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/25/2022] [Indexed: 11/19/2022] Open
Abstract
Bovine mastitis is a common disease occurring in dairy farms and can be caused by more than 150 species of pathogenic bacteria. One of the most common causative organisms is Streptococcus agalactiae, which is also potentially harmful to humans and aquatic animals. At present, research on S. agalactiae in China is mostly concentrated in the northern region, with limited research in the southeastern and southwestern regions. In this study, a total of 313 clinical mastitis samples from large-scale dairy farms in five regions of Sichuan were collected for isolation of S. agalactiae. The epidemiological distribution of S. agalactiae was inferred by serotyping isolates with multiplex polymerase chain reaction. Susceptibility testing and drug resistance genes were detected to guide the clinical use of antibiotics. Virulence genes were also detected to deduce the pathogenicity of S. agalactiae in Sichuan Province. One hundred and five strains of S. agalactiae (33.6%) were isolated according to phenotypic features, biochemical characteristics, and 16S rRNA sequencing. Serotype multiplex polymerase chain reaction analysis showed that all isolates were of type Ia. The isolates were up to 100% sensitive to aminoglycosides (kanamycin, gentamicin, neomycin, and tobramycin), and the resistance rate to β-lactams (penicillin, amoxicillin, ceftazidime, and piperacillin) was up to 98.1%. The TEM gene (β-lactam-resistant) was detected in all isolates, which was in accordance with a drug-resistant phenotype. Analysis of virulence genes showed that all isolates harbored the cfb, cylE, fbsA, fbsB, hylB, and α-enolase genes and none harbored bac or lmb. These data could aid in the prevention and control of mastitis and improve our understanding of epidemiological trends in dairy cows infected with S. agalactiae in Sichuan Province.
Collapse
Affiliation(s)
- Guangli Han
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Baohai Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Zidan Luo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Biao Lu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Zhengzhong Luo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Jieru Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Yin Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Yan Luo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Zexiao Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Liuhong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Shumin Yu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Suizhong Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- * E-mail: (SC); (XY)
| | - Xueping Yao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- * E-mail: (SC); (XY)
| |
Collapse
|
9
|
Qi M, Geng H, Geng N, Cui Y, Qi C, Cheng G, Song K, Hu L, Liu Y, Liu J, Han B. Streptococcus agalactiae-induced autophagy of bovine mammary epithelial cell via PI3K/AKT/mTOR pathway. J DAIRY RES 2022; 89:1-7. [PMID: 35388773 DOI: 10.1017/s0022029922000243] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Streptococcus agalactiae (S. agalactiae) infection is a significant cause of mastitis, resulting in loss of cellular homeostasis and tissue damage. Autophagy plays an essential function in cell survival, defense, and the preservation of cellular homeostasis, and is often part of the response to pathogenic challenge. However, the effect of autophagy induced by S. agalactiae in bovine mammary epithelial cells (bMECs) is mainly unknown. So in this study, an intracellular S. agalactiae infection model was established. Through evaluating the autophagy-related indicators, we observed that after S. agalactiae infection, a significant quantity of LC3-I was converted to LC3-II, p62 was degraded, and levels of Beclin1 and Bcl2 increased significantly in bMECs, indicating that S. agalactiae induced autophagy. The increase in levels of LAMP2 and LysoTracker Deep Red fluorescent spots indicated that lysosomes had participated in the degradation of autophagic contents. After autophagy was activated by rapamycin (Rapa), the amount of p-Akt and p-mTOR decreased significantly, whilst the amount of intracellular S. agalactiae increased significantly. Whereas the autophagy was inhibited by 3-methyladenine (3MA), the number of intracellular pathogens decreased. In conclusion, the results demonstrated that S. agalactiae could induce autophagy through PI3K/Akt/mTOR pathway and utilize autophagy to survive in bMECs.
Collapse
Affiliation(s)
- Mengzhu Qi
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Hao Geng
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Na Geng
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Yukun Cui
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Changxi Qi
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Guodong Cheng
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Kaimin Song
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Liping Hu
- Shandong Provincial Center for Animal Disease Control and Prevention, Jinan, Shandong251000, China
| | - Yongxia Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong271018, China
| | - Bo Han
- College of Veterinary Medicine, China Agricultural University, Beijing100193, China
| |
Collapse
|
10
|
Abdel-Glil MY, Fischer U, Steinhagen D, McCarthy U, Neubauer H, Sprague LD. Phylogenetic Relatedness and Genome Structure of Yersinia ruckeri Revealed by Whole Genome Sequencing and a Comparative Analysis. Front Microbiol 2021; 12:782415. [PMID: 34867924 PMCID: PMC8640586 DOI: 10.3389/fmicb.2021.782415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Yersinia ruckeri is the causative agent of enteric redmouth disease (ERM), a serious infection that affects global aquaculture with high economic impact. The present study used whole genome sequences to perform a comparative analysis on 10 Y. ruckeri strains and to explore their genetic relatedness to other members of the genus. Y. ruckeri, Yersinia entomophaga, and Yersinia nurmii formed a species complex that constitutes the most basal lineage of the genus. The results showed that the taxonomy of Y. ruckeri strains is better defined by using a core genome alignment and phylogenetic analysis. The distribution of accessory genes in all Yersinia species revealed the presence of 303 distinctive genes in Y. ruckeri. Of these, 169 genes were distributed in 17 genomic islands potentially involved in the pathogenesis of ERM via (1) encoding virulence factors such as Afp18, Yrp1, phage proteins and (2) improving the metabolic capabilities by enhancing utilization and metabolism of iron, amino acids (specifically, arginine and histidine), and carbohydrates. The genome of Y. ruckeri is highly conserved regarding gene structure, gene layout and functional categorization of genes. It contains various components of mobile genetic elements but lacks the CRISPR-Cas system and possesses a stable set of virulence genes possibly playing a critical role in pathogenicity. Distinct virulence plasmids were exclusively restricted to a specific clonal group of Y. ruckeri (CG4), possibly indicating a selective advantage. Phylogenetic analysis of Y. ruckeri genomes revealed the co-presence of multiple genetically distant lineages of Y. ruckeri strains circulating in Germany. Our results also suggest a possible dissemination of a specific group of strains in the United States, Peru, Germany, and Denmark. In conclusion, this study provides new insights into the taxonomy and evolution of Y. ruckeri and contributes to a better understanding of the pathogenicity of ERM in aquaculture. The genomic analysis presented here offers a framework for the development of more efficient control strategies for this pathogen.
Collapse
Affiliation(s)
- Mostafa Y Abdel-Glil
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses (IBIZ), Jena, Germany
| | - Uwe Fischer
- Friedrich-Loeffler-Institut, Institute of Infectiology, Greifswald-Insel Riems, Germany
| | - Dieter Steinhagen
- Fish Disease Research Unit, Institute for Parasitology, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Una McCarthy
- Marine Laboratory, Marine Scotland, Aberdeen, United Kingdom
| | - Heinrich Neubauer
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses (IBIZ), Jena, Germany
| | - Lisa D Sprague
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses (IBIZ), Jena, Germany
| |
Collapse
|
11
|
Whole genome sequencing reveals possible host species adaptation of Streptococcus dysgalactiae. Sci Rep 2021; 11:17350. [PMID: 34462475 PMCID: PMC8405622 DOI: 10.1038/s41598-021-96710-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/13/2021] [Indexed: 11/08/2022] Open
Abstract
Streptococcus dysgalactiae (SD) is an emerging pathogen in human and veterinary medicine, and is associated with several host species, disease phenotypes and virulence mechanisms. SD has traditionally been divided into the subspecies dysgalactiae (SDSD) and subsp. equisimilis (SDSE), but recent molecular studies have indicated that the phylogenetic relationships are more complex. Moreover, the genetic basis for the niche versatility of SD has not been extensively investigated. To expand the knowledge about virulence factors, phylogenetic relationships and host-adaptation strategies of SD, we analyzed 78 SDSD genomes from cows and sheep, and 78 SDSE genomes from other host species. Sixty SDSD and 40 SDSE genomes were newly sequenced in this study. Phylogenetic analysis supported SDSD as a distinct taxonomic entity, presenting a mean value of the average nucleotide identity of 99%. Bovine and ovine associated SDSD isolates clustered separately on pangenome analysis, but no single gene or genetic region was uniquely associated with host species. In contrast, SDSE isolates were more heterogenous and could be delineated in accordance with host. Although phylogenetic clustering suggestive of cross species transmission was observed, we predominantly detected a host restricted distribution of the SD-lineages. Furthermore, lineage specific virulence factors were detected, several of them located in proximity to hotspots for integration of mobile genetic elements. Our study indicates that SD has evolved to adapt to several different host species and infers a potential role of horizontal genetic transfer in niche specialization.
Collapse
|
12
|
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: 25] [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
|
13
|
Ma Z, Yin X, Wu P, Hu R, Wang Y, Yi J, Wang Z, Chen C. The Recombinant Expression Proteins FnBP and ClfA From Staphylococcus aureus in Addition to GapC and Sip From Streptococcus agalactiae Can Protect BALB/c Mice From Bacterial Infection. Front Vet Sci 2021; 8:666098. [PMID: 34250059 PMCID: PMC8263938 DOI: 10.3389/fvets.2021.666098] [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: 02/09/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Dairy cow mastitis is a serious disease that is mainly caused by intramammary infection with Staphylococcus aureus and Streptococcus agalactiae [group B streptococcus (GBS)]. FnBP and ClfA are the virulence factors of S. aureus, while GapC is the respective factor for S. agalactiae. Sip is a highly immunogenic protein, and it is conserved in all GBS serotypes. In this study, we analyzed the abovementioned four genes prepared a FnBP+ClfA chimeric protein (FC), a GapC+Sip chimeric protein (GS), and a FnBP+ClfA+GapC+Sip chimeric protein (FCGS) based on the antigenic sites to evaluate their use in vaccine development. After expression and purification of the recombinant proteins in Escherichia coli, BALB/c mice were immunized with them to examine resistance effects. The total lethal and half lethal doses of S. aureus and S. agalactiae were then measured, and the immunoprotective effects of the fusion proteins were evaluated. The FC and FCGS chimeric proteins could induce mice to produce high levels of antibodies, and bacterial loads were significantly reduced in the spleens and livers after challenge. After immunization with FCGS, the recipients resisted the attacks of both S. aureus and S. agalactiae, indicating the potential of the fusion protein as a mastitis vaccine.
Collapse
Affiliation(s)
- Zhongchen Ma
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.,Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China.,College of Life Sciences, Shihezi University, Shihezi, China
| | - Xinyue Yin
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.,Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China.,College of Life Sciences, Shihezi University, Shihezi, China
| | - Peng Wu
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Ruirui Hu
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Yong Wang
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.,Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China.,College of Life Sciences, Shihezi University, Shihezi, China
| | - Jihai Yi
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.,Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China.,College of Life Sciences, Shihezi University, Shihezi, China
| | - Zhen Wang
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.,Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China.,College of Life Sciences, Shihezi University, Shihezi, China
| | - Chuangfu Chen
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.,Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China.,College of Life Sciences, Shihezi University, Shihezi, China
| |
Collapse
|
14
|
Abd El-Aziz NK, Ammar AM, El-Naenaeey ESYM, El Damaty HM, Elazazy AA, Hefny AA, Shaker A, Eldesoukey IE. Antimicrobial and antibiofilm potentials of cinnamon oil and silver nanoparticles against Streptococcus agalactiae isolated from bovine mastitis: new avenues for countering resistance. BMC Vet Res 2021; 17:136. [PMID: 33789637 PMCID: PMC8010958 DOI: 10.1186/s12917-021-02842-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 03/18/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Streptococcus agalactiae (S. agalactiae) is a contagious pathogen of bovine mastitis. It has financial implications for the dairy cattle industry in certain areas of the world. Since antimicrobial resistance increases in dairy farms, natural antimicrobials from herbal origins and nanoparticles have been given more attention as an alternative therapy. Hence, this study reported the antimicrobial and antibiofilm potentials of cinnamon oil, silver nanoparticles (AgNPs), and their combination against multidrug-resistant (MDR) S. agalactiae recovered from clinical bovine mastitis in Egypt. RESULTS Our findings revealed that 73% (146/200) of the examined milk samples collected from dairy cows with clinical mastitis were infected with Streptococci species. Of these, 9.59% (14/146) were identified as S. agalactiae and categorized as MDR. S. agalactiae isolates expressed four virulence genes (Hyl, cylE, scpB, and lmb) and demonstrated an ability to produce biofilms. Cinnamon oil showed high antimicrobial (MICs ≤0.063 μg /mL) and antibiofilm (MBIC50 = 4 μg/mL) potentials against planktonic and biofilms of S. agalactiae isolates, respectively. However, AgNPs showed reasonable antimicrobial (MICs ≤16 μg/mL) and relatively low antibiofilm (MBIC50 = 64 μg/mL) activities against screened isolates. Synergistic antimicrobial or additive antibiofilm interactions of cinnamon oil combined with AgNPs were reported for the first time. Scanning electron microscope (SEM) analysis revealed that biofilms of S. agalactiae isolates treated with cinnamon oil were more seriously damaged than observed in AgNPs cinnamon oil combination. Moreover, reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) showed that cinnamon oil exerted a remarkable down-regulation of pili biosynthesis genes (pilA and pilB) and their regulator (rogB) against S. agalactiae biofilms, meanwhile the AgNPs cinnamon oil combination demonstrated a lower efficacy. CONCLUSIONS This is an in vitro preliminary approach that documented the antibiofilm potential of cinnamon oil and the inhibitory activity of cinnamon oil and its combination with AgNPs against MDR S. agalactiae recovered from clinical mastitis. Further in vivo studies should be carried out in animal models to provide evidence of concept for implementing these alternative candidates in the treatment of dairy farms infected by streptococcal mastitis in the future.
Collapse
Affiliation(s)
- Norhan K Abd El-Aziz
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia, 44511, Egypt.
| | - Ahmed M Ammar
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia, 44511, Egypt
| | - El-Sayed Y M El-Naenaeey
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia, 44511, Egypt
| | - Hend M El Damaty
- Department of Animal Medicine, Infectious Diseases, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia, Egypt
| | - Asmaa A Elazazy
- Abou Hamad Veterinary Organizations, Ministry of Agriculture, Abou Hamad, Sharkia, Egypt
| | - Ahmed A Hefny
- Veterinary Hospital, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia, Egypt
| | - Asmaa Shaker
- Department of Microbiology, Veterinary Hospital, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Ibrahim E Eldesoukey
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| |
Collapse
|
15
|
Alves-Barroco C, Rivas-García L, Fernandes AR, Baptista PV. Tackling Multidrug Resistance in Streptococci - From Novel Biotherapeutic Strategies to Nanomedicines. Front Microbiol 2020; 11:579916. [PMID: 33123110 PMCID: PMC7573253 DOI: 10.3389/fmicb.2020.579916] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023] Open
Abstract
The pyogenic streptococci group includes pathogenic species for humans and other animals and has been associated with enduring morbidity and high mortality. The main reason for the treatment failure of streptococcal infections is the increased resistance to antibiotics. In recent years, infectious diseases caused by pyogenic streptococci resistant to multiple antibiotics have been raising with a significant impact to public health and veterinary industry. The rise of antibiotic-resistant streptococci has been associated to diverse mechanisms, such as efflux pumps and modifications of the antimicrobial target. Among streptococci, antibiotic resistance emerges from previously sensitive populations as result of horizontal gene transfer or chromosomal point mutations due to excessive use of antimicrobials. Streptococci strains are also recognized as biofilm producers. The increased resistance of biofilms to antibiotics among streptococci promote persistent infection, which comprise circa 80% of microbial infections in humans. Therefore, to overcome drug resistance, new strategies, including new antibacterial and antibiofilm agents, have been studied. Interestingly, the use of systems based on nanoparticles have been applied to tackle infection and reduce the emergence of drug resistance. Herein, we present a synopsis of mechanisms associated to drug resistance in (pyogenic) streptococci and discuss some innovative strategies as alternative to conventional antibiotics, such as bacteriocins, bacteriophage, and phage lysins, and metal nanoparticles. We shall provide focused discussion on the advantages and limitations of agents considering application, efficacy and safety in the context of impact to the host and evolution of bacterial resistance.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Lorenzo Rivas-García
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal.,Biomedical Research Centre, University of Granada, Granada, Spain
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro Viana Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
16
|
Tong J, Sun M, Zhang H, Yang D, Zhang Y, Xiong B, Jiang L. Proteomic analysis of bovine mammary epithelial cells after in vitro incubation with S. agalactiae: potential biomarkers. Vet Res 2020; 51:98. [PMID: 32746898 PMCID: PMC7398202 DOI: 10.1186/s13567-020-00808-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 05/19/2020] [Indexed: 12/21/2022] Open
Abstract
Streptococcus agalactiae is one of the causative agents of subclinical mastitis, a common disease of dairy cows that causes great economic losses in the industry worldwide. It is thought that pathology is mainly due to inflammatory damage of bovine mammary epithelial cells (bMECs); however, the mechanism by which S. agalactiae damages the bMECs is not clear. The aim of this study was to evaluate the inflammatory effects of S. agalactiae on bMECs and the resulting changes in protein profiles. The bMECs were incubated with S. agalactiae for different times and assayed for cell viability by MTT assay, apoptosis by annexin V and propidium iodide dual staining, and morphological and ultrastructural changes by scanning and transmission electron microscopy. Quantitative real-time PCR was used to determine the effect of S. agalactiae on expression of mRNA of inflammatory factors in bMECs and protein levels were quantitated by liquid chromatography/mass spectrometry. Exposure to S. agalactiae significantly decreased the cell viability and triggered apoptosis, as well as up-regulating TNF-α, IL-1β and IL-6 mRNA, and inhibiting IL-8 expression. S. agalactiae also induced morphological and ultrastructural changes. Furthermore, we identified 325 up-regulated and 704 down-regulated proteins in the treated vs control group. All significant differentially expressed proteins (DSEPs) were classified into three major areas by function: biological processes, cellular components and molecular functions. These differentially expressed proteins included enzymes and proteins associated with various metabolic processes and cellular immunity. Pathway enrichment analysis showed that eight down-regulated signaling pathways were significantly enriched. Exposure to even subclinical levels of S. agalactiae can lead to inflammation and bMEC damage. Our data suggest some possible molecular mechanisms for the harmful effects of subclinical mastitis in dairy cows.
Collapse
Affiliation(s)
- Jinjin Tong
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing, 102206, People's Republic of China
| | - Mingwei Sun
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing, 102206, People's Republic of China
| | - Hua Zhang
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing, 102206, People's Republic of China
| | - Delian Yang
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing, 102206, People's Republic of China
| | - Yonghong Zhang
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing, 102206, People's Republic of China
| | - Benhai Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China.
| | - Linshu Jiang
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing, 102206, People's Republic of China.
| |
Collapse
|
17
|
Pan-GWAS of Streptococcus agalactiae Highlights Lineage-Specific Genes Associated with Virulence and Niche Adaptation. mBio 2020; 11:mBio.00728-20. [PMID: 32518186 PMCID: PMC7373188 DOI: 10.1128/mbio.00728-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
GBS is a leading cause of mortality in newborn babies in high- and low-income countries worldwide. Different strains of GBS are characterized by different degrees of virulence, where some are harmlessly carried by humans or animals and others are much more likely to cause disease.
The genome sequences of almost 2,000 GBS samples isolated from both animals and humans in high- and low- income countries were analyzed using a pan-genome-wide association study approach. This allowed us to identify 279 genes which are associated with different lineages of GBS, characterized by a different virulence and preferred host. Additionally, we propose that the GBS now carried in humans may have first evolved in animals before expanding clonally once adapted to the human host.
These findings are essential to help understand what is causing GBS disease and how the bacteria have evolved and are transmitted. Streptococcus agalactiae (group B streptococcus; GBS) is a colonizer of the gastrointestinal and urogenital tracts, and an opportunistic pathogen of infants and adults. The worldwide population of GBS is characterized by clonal complexes (CCs) with different invasive potentials. CC17, for example, is a hypervirulent lineage commonly associated with neonatal sepsis and meningitis, while CC1 is less invasive in neonates and more commonly causes invasive disease in adults with comorbidities. The genetic basis of GBS virulence and the extent to which different CCs have adapted to different host environments remain uncertain. We have therefore applied a pan-genome-wide association study (GWAS) approach to 1,988 GBS strains isolated from different hosts and countries. Our analysis identified 279 CC-specific genes associated with virulence, disease, metabolism, and regulation of cellular mechanisms that may explain the differential virulence potential of particular CCs. In CC17 and CC23, for example, we have identified genes encoding pilus, quorum-sensing proteins, and proteins for the uptake of ions and micronutrients which are absent in less invasive lineages. Moreover, in CC17, carriage and disease strains were distinguished by the allelic variants of 21 of these CC-specific genes. Together our data highlight the lineage-specific basis of GBS niche adaptation and virulence. The genome sequences of almost 2,000 GBS samples isolated from both animals and humans in high- and low- income countries were analyzed using a pan-genome-wide association study approach. This allowed us to identify 279 genes which are associated with different lineages of GBS, characterized by a different virulence and preferred host. Additionally, we propose that the GBS now carried in humans may have first evolved in animals before expanding clonally once adapted to the human host. These findings are essential to help understand what is causing GBS disease and how the bacteria have evolved and are transmitted.
Collapse
|
18
|
LMB-1 producing Citrobacter freundii from Argentina, a novel player in the field of MBLs. Int J Antimicrob Agents 2020; 55:105857. [DOI: 10.1016/j.ijantimicag.2019.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/18/2019] [Accepted: 11/24/2019] [Indexed: 01/02/2023]
|
19
|
Khazaal S, Al Safadi R, Osman D, Hiron A, Gilot P. Dual and divergent transcriptional impact of IS1548 insertion upstream of the peptidoglycan biosynthesis gene murB of Streptococcus agalactiae. Gene 2019; 720:144094. [PMID: 31476407 DOI: 10.1016/j.gene.2019.144094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/01/2019] [Accepted: 08/28/2019] [Indexed: 11/28/2022]
Abstract
Fourteen different insertion sequences belonging to seven families were identified in the genome of Streptococcus agalactiae. Among them, IS1548, a mobile element of the ISAs1 family, was linked to clonal complex (CC) 19 strains associated with neonatal meningitis and endocarditis. IS1548 impacts S. agalactiae in two reported ways: i) inactivation of virulence genes by insertion in an open reading frame (e.g. hylB or cpsD), ii) positive modulation of the expression of a downstream gene by insertion in an intergenic region (e.g. lmb). We previously identified an unknown integration site of IS1548 in the intergenic region between the folK and the murB genes involved in folate and peptidoglycan biosynthesis, respectively. In this work, we analyzed the prevalence of IS1548 in a large collection of nine hundred and eleven S. agalactiae strains. IS1548 positive strains belong to twenty-nine different sequence types and to ten CCs. The majority of them were, however, clustered within sequence type 19 and sequence type 22, belonging to CC19 and CC22, respectively. In contrast, IS1548 targets the folK-murB intergenic region exclusively in CC19 strains. We evaluated the impact of the insertion of IS1548 on the expression of murB by locating transcriptional promoters influencing its expression in the presence or absence of IS1548 and by comparative β-galactosidase transcriptional fusion assays. We found that in the absence of IS1548, genes involved in folate biosynthesis are co-transcribed with murB. As it was postulated that a folic acid mediated reaction may be involved in cell wall synthesis, this co-transcription could be necessary to synchronize these two processes. The insertion of IS1548 in the folK-murB intergenic region disrupt this co-transcription. Interestingly, we located a promoter at the right end of IS1548 that is able to initiate additional transcripts of murB. The insertion of IS1548 in this region has thus a dual and divergent impact on the expression of murB. By comparative β-galactosidase transcriptional fusion assays, we showed that, consequently, the overall impact of the insertion of IS1548 results in a minor decrease of murB gene transcription. This study provides new insights into gene expression effects mediated by IS1548 in S. agalactiae.
Collapse
Affiliation(s)
- Sarah Khazaal
- ISP, Bactéries et Risque Materno-Foetal, Université de Tours, INRA, 37032 Tours, France; Azm Center for Research in Biotechnology and its Applications, LBA3B, EDST, Lebanese University, Tripoli 1300, Lebanon
| | - Rim Al Safadi
- Azm Center for Research in Biotechnology and its Applications, LBA3B, EDST, Lebanese University, Tripoli 1300, Lebanon
| | - Dani Osman
- Azm Center for Research in Biotechnology and its Applications, LBA3B, EDST, Lebanese University, Tripoli 1300, Lebanon
| | - Aurélia Hiron
- ISP, Bactéries et Risque Materno-Foetal, Université de Tours, INRA, 37032 Tours, France
| | - Philippe Gilot
- ISP, Bactéries et Risque Materno-Foetal, Université de Tours, INRA, 37032 Tours, France.
| |
Collapse
|
20
|
Richards VP, Velsko IM, Alam MT, Zadoks RN, Manning SD, Pavinski Bitar PD, Hassler HB, Crestani C, Springer GH, Probert BM, Town CD, Stanhope MJ. Population Gene Introgression and High Genome Plasticity for the Zoonotic Pathogen Streptococcus agalactiae. Mol Biol Evol 2019; 36:2572-2590. [PMID: 31350563 PMCID: PMC6805230 DOI: 10.1093/molbev/msz169] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/04/2019] [Accepted: 07/18/2019] [Indexed: 01/06/2023] Open
Abstract
The influence that bacterial adaptation (or niche partitioning) within species has on gene spillover and transmission among bacterial populations occupying different niches is not well understood. Streptococcus agalactiae is an important bacterial pathogen that has a taxonomically diverse host range making it an excellent model system to study these processes. Here, we analyze a global set of 901 genome sequences from nine diverse host species to advance our understanding of these processes. Bayesian clustering analysis delineated 12 major populations that closely aligned with niches. Comparative genomics revealed extensive gene gain/loss among populations and a large pan genome of 9,527 genes, which remained open and was strongly partitioned among niches. As a result, the biochemical characteristics of 11 populations were highly distinctive (significantly enriched). Positive selection was detected and biochemical characteristics of the dispensable genes under selection were enriched in ten populations. Despite the strong gene partitioning, phylogenomics detected gene spillover. In particular, tetracycline resistance (which likely evolved in the human-associated population) from humans to bovine, canines, seals, and fish, demonstrating how a gene selected in one host can ultimately be transmitted into another, and biased transmission from humans to bovines was confirmed with a Bayesian migration analysis. Our findings show high bacterial genome plasticity acting in balance with selection pressure from distinct functional requirements of niches that is associated with an extensive and highly partitioned dispensable genome, likely facilitating continued and expansive adaptation.
Collapse
Affiliation(s)
- Vincent P Richards
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
- Department of Biological Sciences, College of Science, Clemson University, Clemson, SC
| | - Irina M Velsko
- Department of Biological Sciences, College of Science, Clemson University, Clemson, SC
- Department of Archaeogenetics, Max Planck Institute for the Science of Human History, Jena, Germany
| | - Md Tauqeer Alam
- Department of Biological Sciences, College of Science, Clemson University, Clemson, SC
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL
| | - Ruth N Zadoks
- Pentlands Science Park, Moredun Research Institute, Penicuik, United Kingdom
- Institute for Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Shannon D Manning
- Department of Microbiology and Molecular Genetics, Michigan State University, E. Lansing, MI
| | - Paulina D Pavinski Bitar
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Hayley B Hassler
- Department of Biological Sciences, College of Science, Clemson University, Clemson, SC
| | - Chiara Crestani
- Pentlands Science Park, Moredun Research Institute, Penicuik, United Kingdom
| | - Garrett H Springer
- Department of Biological Sciences, College of Science, Clemson University, Clemson, SC
| | - Brett M Probert
- Department of Biological Sciences, College of Science, Clemson University, Clemson, SC
| | | | - Michael J Stanhope
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| |
Collapse
|
21
|
Cobo-Angel CG, Jaramillo-Jaramillo AS, Palacio-Aguilera M, Jurado-Vargas L, Calvo-Villegas EA, Ospina-Loaiza DA, Rodriguez-Lecompte JC, Sanchez J, Zadoks R, Ceballos-Marquez A. Potential group B Streptococcus interspecies transmission between cattle and people in Colombian dairy farms. Sci Rep 2019. [PMID: 31575879 DOI: 10.1038/s41598–019–50225–w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Group B Streptococcus (GBS), is a leading cause of neonatal death and an emerging pathogen in adults. Additionally, GBS is a bovine pathogen causing intramammary infections. The likelihood of GBS interspecies transmission is largely unknown. We explored the potential transmission of GBS between cattle and people on dairy farms in Colombia and compared the antimicrobial resistance (AMR) profiles of isolates from both host species. Across 33 farms, throat swabs and rectal swabs were collected from 191 people, and rectal swabs and composite milk samples from 2092 cattle, yielding 60 human isolates and 301 bovine isolates. The majority (64%) of isolates belonged to shared sequence types (ST). Sequence type (ST) 1 was the most common strain in both host species, suggesting that interspecies transmission may be possible. Two members of the bovine-specific clonal complex 61/67 were detected in human samples (ST718 and ST1175), providing evidence for the lack of genuine species barriers. Apparent prevalence of penicillin resistance was surprisingly high in human and bovine isolates. Further investigation of this phenomenon is needed and could lead to modification of standard testing and treatment recommendations in human and veterinary medicine.
Collapse
Affiliation(s)
- Claudia G Cobo-Angel
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia
| | - Ana S Jaramillo-Jaramillo
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia
| | | | | | | | - Diego A Ospina-Loaiza
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia
| | - Juan C Rodriguez-Lecompte
- Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | - Javier Sanchez
- Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | - Ruth Zadoks
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Sydney School of Veterinary Science, University of Sydney, Camden, NSW, Australia
| | - Alejandro Ceballos-Marquez
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia.
| |
Collapse
|
22
|
Potential group B Streptococcus interspecies transmission between cattle and people in Colombian dairy farms. Sci Rep 2019; 9:14025. [PMID: 31575879 PMCID: PMC6773701 DOI: 10.1038/s41598-019-50225-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 08/27/2019] [Indexed: 01/10/2023] Open
Abstract
Group B Streptococcus (GBS), is a leading cause of neonatal death and an emerging pathogen in adults. Additionally, GBS is a bovine pathogen causing intramammary infections. The likelihood of GBS interspecies transmission is largely unknown. We explored the potential transmission of GBS between cattle and people on dairy farms in Colombia and compared the antimicrobial resistance (AMR) profiles of isolates from both host species. Across 33 farms, throat swabs and rectal swabs were collected from 191 people, and rectal swabs and composite milk samples from 2092 cattle, yielding 60 human isolates and 301 bovine isolates. The majority (64%) of isolates belonged to shared sequence types (ST). Sequence type (ST) 1 was the most common strain in both host species, suggesting that interspecies transmission may be possible. Two members of the bovine-specific clonal complex 61/67 were detected in human samples (ST718 and ST1175), providing evidence for the lack of genuine species barriers. Apparent prevalence of penicillin resistance was surprisingly high in human and bovine isolates. Further investigation of this phenomenon is needed and could lead to modification of standard testing and treatment recommendations in human and veterinary medicine.
Collapse
|
23
|
Chen SL. Genomic Insights Into the Distribution and Evolution of Group B Streptococcus. Front Microbiol 2019; 10:1447. [PMID: 31316488 PMCID: PMC6611187 DOI: 10.3389/fmicb.2019.01447] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 06/11/2019] [Indexed: 01/31/2023] Open
Abstract
Streptococcus agalactiae, also known as Group B Streptococcus (GBS), is a bacteria with truly protean biology. It infects a variety of hosts, among which the most commonly studied are humans, cattle, and fish. GBS holds a singular position in the history of bacterial genomics, as it was the substrate used to describe one of the first major conceptual advances of comparative genomics, the idea of the pan-genome. In this review, I describe a brief history of GBS and the major contributions of genomics to understanding its genome plasticity and evolution as well as its molecular epidemiology, focusing on the three hosts mentioned above. I also discuss one of the major recent paradigm shifts in our understanding of GBS evolution and disease burden: foodborne GBS can cause invasive infections in humans.
Collapse
Affiliation(s)
- Swaine L Chen
- Division of Infectious Diseases, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Infectious Diseases Group, Genome Institute of Singapore, Singapore, Singapore
| |
Collapse
|
24
|
Florindo C, Barroco CA, Silvestre I, Damião V, Gomes JP, Spellerberg B, Santos-Sanches I, Borrego MJ. Capsular Type, Sequence Type and Microbial Resistance Factors Impact on DNase Activity of Streptococcus agalactiae Strains from Human and Bovine Origin. Eur J Microbiol Immunol (Bp) 2018; 8:149-154. [PMID: 30719332 PMCID: PMC6348702 DOI: 10.1556/1886.2018.00026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/13/2018] [Indexed: 12/20/2022] Open
Abstract
Extracellular deoxyribonucleases (DNases) contribute to the spread of pathogenic bacteria through the evasion from host innate immunity. Our main objective was to evaluate the production of extracellular DNases by human and bovine Streptococcus agalactiae clinical strains and perform a correlation of genetic lineages and DNase activity with capsular type, genetic determinants, clinical origin (colonization and infection), and host (human or bovine). DNase activity was evaluated by qualitative and quantitative assays for a collection of 406 human (n = 285) and bovine (n = 121) strains. All (121/121) bovine were isolated from mastitis and revealed to be DNase (+), indicating a putative pathogenic role in this clinical scenario. From the human S. agalactiae strains, 86% (245/285) showed DNase activity, among which all strains belonging to capsular types, namely, Ia, Ib, III-2, and IV. All CC17 strains (n = 58) and 56/96 (58.3%) of the CC19 displayed DNase activity. DNase (-) strains belonged to the CC19 group. However, the subcharacterization of CC19 S. agalactiae strains through multiple-locus variable number tandem repeat analysis (MLVA), antibiotic resistance, mobile elements, and surface proteins did not provide any distinction among DNase producers and non-producers. The production of DNases by all human CC17 strains, about two-fifths of human CC19, and all bovine strains, suggest an important contribution of DNases to hypervirulence.
Collapse
Affiliation(s)
- Carlos Florindo
- Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, Departamento de Ciências da Vida, UCIBIO
- Department of Infectious Diseases, National Institute of Health, Lisbon
| | - Cinthia Alves Barroco
- Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, Departamento de Ciências da Vida, UCIBIO
| | - Inês Silvestre
- Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, Departamento de Ciências da Vida, UCIBIO
- Department of Infectious Diseases, National Institute of Health, Lisbon
| | - Vera Damião
- Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, Departamento de Ciências da Vida, UCIBIO
- Department of Infectious Diseases, National Institute of Health, Lisbon
| | - João Paulo Gomes
- Department of Infectious Diseases, National Institute of Health, Lisbon
| | | | - Ilda Santos-Sanches
- Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, Departamento de Ciências da Vida, UCIBIO
| | | |
Collapse
|
25
|
Cobo-Ángel C, Jaramillo-Jaramillo AS, Lasso-Rojas LM, Aguilar-Marin SB, Sanchez J, Rodriguez-Lecompte JC, Ceballos-Márquez A, Zadoks RN. Streptococcus agalactiae is not always an obligate intramammary pathogen: Molecular epidemiology of GBS from milk, feces and environment in Colombian dairy herds. PLoS One 2018; 13:e0208990. [PMID: 30532177 PMCID: PMC6287850 DOI: 10.1371/journal.pone.0208990] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/27/2018] [Indexed: 01/01/2023] Open
Abstract
For many years Streptococcus agalactiae has been considered an obligate intramammary and strictly contagious pathogen in dairy cattle. However, recent reports of S. agalactiae isolation from extramammary sources have contradicted that premise. To gain further insight into the epidemiology of S. agalactiae infection in cattle, we examined its distribution and heterogeneity of strains in bovine milk, bovine feces, and the environment in Colombian dairy farms. First, a longitudinal study was conducted at herd level in 152 dairy herds. Bulk tank milk samples from each herd where collected twice a month for six months. A follow-up study with a cross sectional design at the cow level was conducted in a subset of 25 farms positive for S. agalactiae. Cow-level milk samples from 1712 lactatting cows and 1545 rectal samples were collected, as well as 120 environmental samples. Samples were used for S. agalactiae detection and genotyping using Multi Locus Sequence Typing. Results showed sporadic rather than repeated isolation of S. agalactiae from bulk tank milk in 40% of the positive herds, challenging the idea that S. agalactiae is a highly contagious pathogen causing chronic infections. S. agalactiae was isolated from rectal or environmental samples in 32% and 12% of cross-sectional study farms, respectively, demonstrating that the bacteria can survive in extramammary sources and that S. agalactiae is not an obligate intramammary pathogen. The same strain was isolated from rectal and bulk tank milk samples in eight farms, suggesting that fecal shedding is frequent, and contributes to the presence of S. agalactiae in bulk tank. High within-herd heterogeneity of strains was found, which is distinct from the situation in developed dairy industries. These new epidemiological findings should be considered to adjust surveillance and control recommendations for S. agalactiae.
Collapse
Affiliation(s)
- Claudia Cobo-Ángel
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia
- * E-mail:
| | - Ana S. Jaramillo-Jaramillo
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia
| | - Laura M. Lasso-Rojas
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia
| | - Sandra B. Aguilar-Marin
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia
| | - Javier Sanchez
- Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | - Juan C. Rodriguez-Lecompte
- Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | - Alejandro Ceballos-Márquez
- Research Group in Milk Quality and Veterinary Epidemiology, Faculty of Agricultural Sciences, Universidad de Caldas, Manizales, Colombia
| | - Ruth N. Zadoks
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Moredun Research Institute, Penicuik, United Kingdom
| |
Collapse
|
26
|
Wang R, Li L, Huang T, Huang Y, Huang W, Yang X, Lei A, Chen M. Phylogenetic, comparative genomic and structural analyses of human Streptococcus agalactiae ST485 in China. BMC Genomics 2018; 19:716. [PMID: 30261834 PMCID: PMC6161333 DOI: 10.1186/s12864-018-5084-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/17/2018] [Indexed: 12/24/2022] Open
Abstract
Background Streptococcus agalactiae (Group B Streptococcus, GBS) is a common bacteria species infecting both human and bovine. Previous studies have shown that the GBS isolated from human and bovine are mostly unrelated and belong to separate populations. However, recently, the bovine GBS CC103 has become the dominant epidemic strain and frequently isolated from human patients. In particular, the ST485 GBS, a member of CC103, has become the new dominant ST in China and exhibited very high pathogenicity. This phenomenon is not consistent with the established understanding about the relationship between bovine and human GBS, which needs to be re-investigated. Results The genome-based phylogenetic analysis showed that the human and bovine GBS CC103 strains had very close genetic relationship and they were alternately distributed on the evolutionary tree. CC103 strains evolved into several branches, including the ST485, which exhibited high pathogenicity and specifically infected human. Compared to other CC103 strains, the ST485 lacked Lac.2 gene structure and acquired the CadDX gene structure in their genomes. Conclusions Our results indicate that GBS CC103 could propagate across human and bovine, and GBS ST485 might evolve from the ST103 that could infect both human and bovine. Moreover, the recombination of Lac.2 and CadDX gene structures might play an important role in the formation of highly pathogenic ST485 in China. Electronic supplementary material The online version of this article (10.1186/s12864-018-5084-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Wang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, Guangxi, 530021, People's Republic of China.,Institute of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi, China
| | - Liping Li
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, Guangxi, 530021, People's Republic of China
| | - Ting Huang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, Guangxi, 530021, People's Republic of China
| | - Yan Huang
- Guangxi Center for Disease Control and Prevention, Nanning, 530021, Guangxi, China
| | - Weiyi Huang
- Institute of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi, China
| | - Xiuying Yang
- National Medical College of Right Rivers, Baise, 533000, Guangxi, China
| | - Aiying Lei
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, Guangxi, 530021, People's Republic of China
| | - Ming Chen
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, Guangxi, 530021, People's Republic of China. .,Guangxi Center for Disease Control and Prevention, Nanning, 530021, Guangxi, China.
| |
Collapse
|
27
|
Microevolution of Streptococcus agalactiae ST-261 from Australia Indicates Dissemination via Imported Tilapia and Ongoing Adaptation to Marine Hosts or Environment. Appl Environ Microbiol 2018; 84:AEM.00859-18. [PMID: 29915111 DOI: 10.1128/aem.00859-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/12/2018] [Indexed: 11/20/2022] Open
Abstract
Streptococcus agalactiae (group B Streptococcus [GBS]) causes disease in a wide range of animals. The serotype Ib lineage is highly adapted to aquatic hosts, exhibiting substantial genome reduction compared with terrestrial conspecifics. Here, we sequence genomes from 40 GBS isolates, including 25 isolates from wild fish and captive stingrays in Australia, six local veterinary or human clinical isolates, and nine isolates from farmed tilapia in Honduras, and compared them with 42 genomes from public databases. Phylogenetic analysis based on nonrecombinant core-genome single nucleotide polymorphisms (SNPs) indicated that aquatic serotype Ib isolates from Queensland were distantly related to local veterinary and human clinical isolates. In contrast, Australian aquatic isolates are most closely related to a tilapia isolate from Israel, differing by only 63 core-genome SNPs. A consensus minimum spanning tree based on core-genome SNPs indicates the dissemination of sequence type 261 (ST-261) from an ancestral tilapia strain, which is congruent with several introductions of tilapia into Australia from Israel during the 1970s and 1980s. Pangenome analysis identified 1,440 genes as core, with the majority being dispensable or strain specific, with non-protein-coding intergenic regions (IGRs) divided among core and strain-specific genes. Aquatic serotype Ib strains have lost many virulence factors during adaptation, but six adhesins were well conserved across the aquatic isolates and might be critical for virulence in fish and for targets in vaccine development. The close relationship among recent ST-261 isolates from Ghana, the United States, and China with the Israeli tilapia isolate from 1988 implicates the global trade in tilapia seed for aquaculture in the widespread dissemination of serotype Ib fish-adapted GBS.IMPORTANCEStreptococcus agalactiae (GBS) is a significant pathogen of humans and animals. Some lineages have become adapted to particular hosts, and serotype Ib is highly specialized to fish. Here, we show that this lineage is likely to have been distributed widely by the global trade in tilapia for aquaculture, with probable introduction into Australia in the 1970s and subsequent dissemination in wild fish populations. We report here the variability in the polysaccharide capsule among this lineage but identify a cohort of common surface proteins that may be a focus of future vaccine development to reduce the biosecurity risk in international fish trade.
Collapse
|
28
|
Tomazi T, de Souza Filho AF, Heinemann MB, dos Santos MV. Molecular characterization and antimicrobial susceptibility pattern of Streptococcus agalactiae isolated from clinical mastitis in dairy cattle. PLoS One 2018; 13:e0199561. [PMID: 29928042 PMCID: PMC6013152 DOI: 10.1371/journal.pone.0199561] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/08/2018] [Indexed: 12/12/2022] Open
Abstract
The objectives of this study were to: (a) genotypically characterize Streptococcus agalactiae isolates recovered from clinical mastitis (CM) cases in dairy cows and, (b) determine the association of antimicrobial susceptibility (AMS) and genotypes of Strep. agalactiae clustered according to the genetic similarity. A total of 89 Strep. agalactiae isolates recovered from bovine CM were genotyped using random amplified polymorphic DNA (RAPD) analysis. In addition, the AMS of the isolates was determined using a commercial broth microdilution test composed of 10 antimicrobials (penicillin, ampicillin, oxacillin, cephalothin, ceftiofur, penicillin/novobiocin, erythromycin, pirlimycin, tetracycline, and sulfadimethoxine). Descriptive analysis was used to report the frequency of RAPD-types and genotypic clusters within herd, housing system, season and CM severity scores. The minimal antimicrobial concentrations that inhibited 50% (MIC50) and 90% (MIC90) of the isolates were calculated and survival analysis was completed to verify the differences of AMS among genotypic clusters. Results of RAPD showed a great genotypic diversity of Strep. agalactiae (45 RAPD-types) and three clusters (Ia, Ib and II) were created based on the genetic similarity among genotypes. After clustering, a high genetic similarity was observed within and between herds. Overall, Strep. agalactiae showed high susceptibility to most antimicrobials, except to tetracycline and erythromycin. Differences in the AMS among clusters were observed for ampicillin, ceftiofur, erythromycin, pirlimycin, sulfadimethoxine and tetracycline. In conclusion, Strep. agalactiae is still highly susceptible to most antimicrobials, although differences in susceptibility to certain antimicrobials were observed among genotypic clusters.
Collapse
Affiliation(s)
- Tiago Tomazi
- Department of Animal Production and Nutrition, Milk Quality Research Laboratory (Qualileite), University of São Paulo, Pirassununga, Brazil
| | - Antonio Francisco de Souza Filho
- Department of Preventive Veterinary Medicine and Animal Health, Laboratory of Bacterial Zoonosis, University of São Paulo, São Paulo, Brazil
| | - Marcos Bryan Heinemann
- Department of Preventive Veterinary Medicine and Animal Health, Laboratory of Bacterial Zoonosis, University of São Paulo, São Paulo, Brazil
| | - Marcos Veiga dos Santos
- Department of Animal Production and Nutrition, Milk Quality Research Laboratory (Qualileite), University of São Paulo, Pirassununga, Brazil
- * E-mail:
| |
Collapse
|
29
|
CTX-M-15-Producing Shewanella Species Clinical Isolate Expressing OXA-535, a Chromosome-Encoded OXA-48 Variant, Putative Progenitor of the Plasmid-Encoded OXA-436. Antimicrob Agents Chemother 2017; 62:AAC.01879-17. [PMID: 29038283 DOI: 10.1128/aac.01879-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/11/2017] [Indexed: 11/20/2022] Open
Abstract
Shewanella spp. constitute a reservoir of antibiotic resistance determinants. In a bile sample, we identified three extended-spectrum-β-lactamase (ESBL)-producing bacteria (Escherichia coli, Klebsiella pneumoniae, and Shewanella sp. strain JAB-1) isolated from a child suffering from cholangitis. Our objectives were to characterize the genome and the resistome of the first ESBL-producing isolate of the genus Shewanella and determine whether plasmidic exchange occurred between the three bacterial species. Bacterial isolates were characterized using matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS), standard biochemical tools, and antimicrobial susceptibility testing. Shewanella sp. JAB-1 and ESBL gene-encoding plasmids were characterized using PacBio and Illumina whole-genome sequencing, respectively. The Shewanella sp. JAB-1 chromosome-encoded OXA-48 variant was cloned and functionally characterized. Whole-genome sequencing (WGS) of the Shewanella sp. clinical isolate JAB-1 revealed the presence of a 193-kb plasmid belonging to the IncA/C incompatibility group and harboring two ESBL genes, blaCTX-M-15 and blaSHV-2ablaCTX-M-15 gene-carrying plasmids belonging to the IncY and IncR incompatibility groups were also found in the E. coli and K. pneumoniae isolates from the same patient, respectively. A comparison of the blaCTX-M-15 genetic environment indicated the independent origin of these plasmids and dismissed in vivo transfers. Furthermore, characterization of the resistome of Shewanella sp. JAB-1 revealed the presence of a chromosome-carried blaOXA-535 gene, likely the progenitor of the plasmid-carried blaOXA-436 gene, a novel blaOXA-48-like gene. The expression of blaOXA-535 in E. coli showed the carbapenem-hydrolyzing activity of OXA-535. The production of OXA-535 in Shewanella sp. JAB-1 could be evidenced using molecular and immunoenzymatic tests, but not with biochemical tests that monitor carbapenem hydrolysis. In this study, we have identified a CTX-M-15-producing Shewanella species that was responsible for a hepatobiliary infection and that is likely the progenitor of OXA-436, a novel plasmid-encoded OXA-48-like class D carbapenemase.
Collapse
|
30
|
Pang M, Sun L, He T, Bao H, Zhang L, Zhou Y, Zhang H, Wei R, Liu Y, Wang R. Molecular and virulence characterization of highly prevalent Streptococcus agalactiae circulated in bovine dairy herds. Vet Res 2017; 48:65. [PMID: 29037262 PMCID: PMC5644065 DOI: 10.1186/s13567-017-0461-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/11/2017] [Indexed: 11/23/2022] Open
Abstract
Bovine mastitis caused by Streptococcus agalactiae continues to be one of the major veterinary and economic issues in certain areas of the world. The more prevalent S. agalactiae strains that cause bovine mastitis in China dairy farms belong to a number of bovine-adapted sequence types (STs) ST67, ST103 and ST568. However, it is unknown why these STs can emerge as highly prevalent clones in bovine dairy farms. Here, to determine if a variety of virulence characteristics were associated with these highly prevalent STs, the molecular and virulence characterization of 116 strains isolated from bovine, human, fish and environment were analyzed. Our data showed that all bovine-adapted strains could be assigned to capsular genotype Ia or II, and carried pilus island 2b, and lactose operon. Importantly, we demonstrated that the growth ability in milk, biofilm formation ability and adhesion ability to bovine mammary epithelial cells (BMECs) were significantly higher for all bovine-adapted strains compared to strains from other origins. Additionally, ST103 and ST568 strains exhibited significantly higher hemolytic activity and cytotoxicity than ST67 strains. In conclusion, our study provides substantial evidence for the hypothesis that the virulence characteristics including efficient growth in milk, elevated biofilm formation ability, together with strong adhesion ability might have favored the high prevalence of the STs in the bovine environment, whereas the hemolytic activity and cytotoxicity were not the crucial characteristics.
Collapse
Affiliation(s)
- Maoda Pang
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China
| | - Lichang Sun
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China
| | - Tao He
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China
| | - Hongdu Bao
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China
| | - Lili Zhang
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China
| | - Yan Zhou
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China
| | - Hui Zhang
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China
| | - Ruicheng Wei
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China
| | - Yongjie Liu
- College of Veterinary Medicine, Nanjing Agricultural University, No. 1 Weigang, Nanjing, 210095, China
| | - Ran Wang
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Nanjing, 210014, China.
| |
Collapse
|
31
|
Wang R, Li L, Huang Y, Huang T, Tang J, Xie T, Lei A, Luo F, Li J, Huang Y, Shi Y, Wang D, Chen M, Mi Q, Huang W. Pathogenicity of Human ST23 Streptococcus agalactiae to Fish and Genomic Comparison of Pathogenic and Non-pathogenic Isolates. Front Microbiol 2017; 8:1933. [PMID: 29056932 PMCID: PMC5635047 DOI: 10.3389/fmicb.2017.01933] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/21/2017] [Indexed: 12/14/2022] Open
Abstract
Streptococcus agalactiae, or Group B Streptococcus (GBS), is a major pathogen causing neonatal sepsis and meningitis, bovine mastitis, and fish meningoencephalitis. CC23, including its namesake ST23, is not only the predominant GBS strain derived from human and cattle, but also can infect a variety of homeothermic and poikilothermic species. However, it has never been characterized in fish. This study aimed to determine the pathogenicity of ST23 GBS to fish and explore the mechanisms causing the difference in the pathogenicity of ST23 GBS based on the genome analysis. Infection of tilapia with 10 human-derived ST23 GBS isolates caused tissue damage and the distribution of pathogens within tissues. The mortality rate of infection was ranged from 76 to 100%, and it was shown that the mortality rate caused by only three human isolates had statistically significant difference compared with fish-derived ST7 strain (P < 0.05), whereas the mortality caused by other seven human isolates did not show significant difference compared with fish-derived ST7 strain. The genome comparison and prophage analysis showed that the major genome difference between virulent and non-virulent ST23 GBS was attributed to the different prophage sequences. The prophage in the P1 region contained about 43% GC and encoded 28–39 proteins, which can mediate the acquisition of YafQ/DinJ structure for GBS by phage recombination. YafQ/DinJ belongs to one of the bacterial toxin–antitoxin (TA) systems and allows cells to cope with stress. The ST23 GBS strains carrying this prophage were not pathogenic to tilapia, but the strains without the prophage or carrying the pophage that had gene mutation or deletion, especially the deletion of YafQ/DinJ structure, were highly pathogenic to tilapia. In conclusion, human ST23 GBS is highly pathogenic to fish, which may be related to the phage recombination.
Collapse
Affiliation(s)
- Rui Wang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, China.,Institute of Animal Science and Technology, Guangxi University, Nanning, China
| | - Liping Li
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, China
| | - Yin Huang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, China
| | - Ting Huang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, China
| | - Jiayou Tang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, China
| | - Ting Xie
- Hechi Center for Animal Disease Control and Prevention, Hechi, China
| | - Aiying Lei
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, China
| | - Fuguang Luo
- Aquatic Animal Disease Pevention and Control Laboratory, Liuzhou's Aquaculture Technology Extending Station, Liuzhou, China
| | - Jian Li
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yan Huang
- Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Yunliang Shi
- Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Dongying Wang
- Institute of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ming Chen
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, China
| | - Qiang Mi
- Aquaculture Laboratory, Guangxi Aquaculture and Animal Husbandry School, Nanning, China
| | - Weiyi Huang
- Guangxi Center for Disease Control and Prevention, Nanning, China
| |
Collapse
|
32
|
Reyes J, Chaffer M, Rodriguez-Lecompte JC, Sánchez J, Zadoks RN, Robinson N, Cardona X, Ramírez N, Keefe GP. Short communication: Molecular epidemiology of Streptococcus agalactiae differs between countries. J Dairy Sci 2017; 100:9294-9297. [PMID: 28918144 DOI: 10.3168/jds.2017-13363] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/30/2017] [Indexed: 01/20/2023]
Abstract
Group B Streptococcus or Streptococcus agalactiae continue to be challenging for milk quality programs in countries with emerging dairy industries, such as Colombia, where high prevalence has been reported. Molecular typing of isolates is needed to understand the variability and epidemiology of this pathogen and to develop effective control and eradication programs. We characterized the molecular profile of Strep. agalactiae isolated from cows with subclinical mastitis in 21 Colombian dairy herds and measured diversity within and between herds using multilocus sequence typing. Isolates belonged to sequence type 248 [clonal complex (CC) 103; n = 30), ST1 (CC1; n = 6) or ST22 (CC22; n = 4)], whereas members of CC67/61, the dominant type in North America, were not detected. Presence of multiple clonally unrelated sequence type within a herd was common, which contrasts with the situation in European countries and suggests introduction from multiple sources. Our results demonstrate that conclusions from molecular epidemiological studies in 1 region cannot necessarily be extrapolated to other regions, and no single bovine-adapted CC of Strep. agalactiae exists in Colombia. Improvements in internal and external biosecurity will be needed to reduce Strep. agalactiae prevalence in Colombian dairy herds.
Collapse
Affiliation(s)
- J Reyes
- Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3; Biogenesis Research Group, Faculty of Agricultural Sciences, University of Antioquia, Calle 70 No. 52-21, C.P. 050010 Medellin, Colombia.
| | - M Chaffer
- Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3
| | - Juan Carlos Rodriguez-Lecompte
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3
| | - Javier Sánchez
- Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3
| | - Ruth N Zadoks
- Moredun Research Institute, Penicuik, Scotland, UK, EH26 0PZ; Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK, G61 1QH
| | - Natasha Robinson
- Maritime Quality Milk Laboratory, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3
| | | | - N Ramírez
- Epidemiology and Public Health, Centauro Research Group, Faculty of Agricultural Sciences, University of Antioquia, Calle 70 No. 52-21, C.P. 050010 Medellín, Colombia
| | - G P Keefe
- Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3
| |
Collapse
|
33
|
Parallel Evolution of Group B Streptococcus Hypervirulent Clonal Complex 17 Unveils New Pathoadaptive Mutations. mSystems 2017; 2:mSystems00074-17. [PMID: 28904998 PMCID: PMC5585690 DOI: 10.1128/msystems.00074-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/10/2017] [Indexed: 12/21/2022] Open
Abstract
The incidence of group B Streptococcus (GBS) neonatal disease continues to be a significant cause of concern worldwide. Strains belonging to clonal complex 17 (CC17) are the most frequently responsible for GBS infections in neonates, especially among late-onset disease cases. Therefore, we undertook the largest genomic study of GBS CC17 strains to date to decipher the genetic bases of their remarkable colonization and infection ability. We show that crucial functions involved in different steps of the colonization or infection process of GBS are distinctly mutated during the adaptation of CC17 to the human host. In particular, our results implicate the CovRS two-component regulator of virulence in the differentiation between carriage- and disease-associated isolates. Not only does this work raise important implications for the ongoing development of a vaccine against GBS but might also drive the discovery of key functions for GBS adaptation and pathogenesis that have been overlooked until now. Group B Streptococcus (GBS) is a commensal of the gastrointestinal and genitourinary tracts, while a prevailing cause of neonatal disease worldwide. Of the various clonal complexes (CCs), CC17 is overrepresented in GBS-infected newborns for reasons that are still largely unknown. Here, we report a comprehensive genomic analysis of 626 CC17 isolates collected worldwide, identifying the genetic traits behind their successful adaptation to humans and the underlying differences between carriage and clinical strains. Comparative analysis with 923 GBS genomes belonging to CC1, CC19, and CC23 revealed that the evolution of CC17 is distinct from that of other human-adapted lineages and recurrently targets functions related to nucleotide and amino acid metabolism, cell adhesion, regulation, and immune evasion. We show that the most distinctive features of disease-specific CC17 isolates were frequent mutations in the virulence-associated CovS and Stk1 kinases, underscoring the crucial role of the entire CovRS regulatory pathway in modulating the pathogenicity of GBS. Importantly, parallel and convergent evolution of major components of the bacterial cell envelope, such as the capsule biosynthesis operon, the pilus, and Rib, reflects adaptation to host immune pressures and should be taken into account in the ongoing development of a GBS vaccine. The presence of recurrent targets of evolution not previously implicated in virulence also opens the way for uncovering new functions involved in host colonization and GBS pathogenesis. IMPORTANCE The incidence of group B Streptococcus (GBS) neonatal disease continues to be a significant cause of concern worldwide. Strains belonging to clonal complex 17 (CC17) are the most frequently responsible for GBS infections in neonates, especially among late-onset disease cases. Therefore, we undertook the largest genomic study of GBS CC17 strains to date to decipher the genetic bases of their remarkable colonization and infection ability. We show that crucial functions involved in different steps of the colonization or infection process of GBS are distinctly mutated during the adaptation of CC17 to the human host. In particular, our results implicate the CovRS two-component regulator of virulence in the differentiation between carriage- and disease-associated isolates. Not only does this work raise important implications for the ongoing development of a vaccine against GBS but might also drive the discovery of key functions for GBS adaptation and pathogenesis that have been overlooked until now. Author Video: An author video summary of this article is available.
Collapse
|