Next Article in Journal
Use of Recombinant Escherichia coli Strains in Immunofluorescence Assays for Melioidosis Diagnosis
Next Article in Special Issue
Investigation of Potential Reservoirs of Non-Tuberculous Mycobacteria in a European Sea Bass (Dicentrarchus labrax) Farm
Previous Article in Journal
Investigating the Circulation of Ebola Viruses in Bats during the Ebola Virus Disease Outbreaks in the Equateur and North Kivu Provinces of the Democratic Republic of Congo from 2018
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Advances on Tilapia Streptococcosis

1
College of Life Sciences, Beijing Normal University, Beijing 100875, China
2
National Institute of Biological Sciences, Zhongguancun Life Science Park, Beijing 102206, China
3
Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
Pathogens 2021, 10(5), 558; https://doi.org/10.3390/pathogens10050558
Submission received: 17 February 2021 / Revised: 14 March 2021 / Accepted: 17 March 2021 / Published: 6 May 2021
(This article belongs to the Special Issue New Insights into Fish Pathogens in Aquaculture)

Abstract

:
Streptococcus agalactiae, often referred to as group B streptococci (GBS), is a severe pathogen that can infect humans as well as other animals, including tilapia, which is extremely popular in commercial aquaculture. This pathogen causes enormous pecuniary loss, and typical symptoms of streptococcosis—the disease caused by S. agalactiae—include abnormal behavior, exophthalmos, and meningitis, among others. Multiple studies have examined virulence factors associated with S. agalactiae infection, and vaccines were explored, including studies of subunit vaccines. Known virulence factors include capsular polysaccharide (CPS), hemolysin, Christie-Atkins-Munch-Peterson (CAMP) factor, hyaluronidase (HAase), superoxide dismutase (SOD), and serine-threonine protein kinase (STPK), and effective vaccine antigens reported to date include GapA, Sip, OCT, PGK, FbsA, and EF-Tu. In this review, I summarize findings from several studies about the etiology, pathology, virulence factors, and vaccine prospects for S. agalactiae. I end by considering which research areas are likely to yield success in the prevention and treatment of tilapia streptococcosis.

1. Introduction

Streptococcus agalactiae (S. agalactiae), also commonly referred to as group B streptococcus (GBS), is a severe pathogen that can infect humans and a diversity of other animals, including reptiles, frogs, bovines, fish (including tilapia), and pigs, among others [1,2,3]. Streptococcosis—the disorder caused by GBS infectionis a major obstacle faced by the tilapia aquaculture industry: in 2011, streptococcosis caused the loss of $40 million in the tilapia industry in China, owing to high morbidity and mortality, which can reach up to 80% in outbreaks [4,5]. Given this potential for loss, it is unsurprising that researchers are striving to develop effective means to control tilapia streptococcosis outbreaks. There has been some progress in research regarding serotypes and virulence mechanisms, and there are some promising prospects for effective vaccines; this review considers progress over the past several years from investigations about these topics.
GBS, like many other pathogenic species, possesses many virulence factors, biomolecular factors that promote the ability of pathogens to infect and/or damage hosts [6,7,8]. Among GBS virulence factors, a large number are known to affect adherence and invasion of host cells, as well as evasion of host immunity [7]. A better understanding of these virulence factors can support the development of control and therapeutic strategies. Studies of GBS isolated from humans have split virulence factors into pore-forming toxins, factors for immune evasion, resistance to antimicrobial peptides (AMPs), host-cell adherence and invasion, and other virulence factors [7,8,9]. Pore-forming toxins—vitally important pathogenesis factors—can facilitate the entry of GBS into host cells. The most common kinds of pore-forming toxins are β-hemolysin/cytolysin (β-H/C) and Christie-Atkins-Munch-Peterson (CAMP) factor. GBS can encode several virulence factors that promote immune evasion. The adherent factors, such as fibrinogen-binding protein (FbsA/B/C), laminin-binding protein (Lmb), the immunogenic bacterial adhesin (BibA), Pili (PilA/B/C), serine-rich repeat (srr), α C protein, and others, are responsible for GBS binding to extracellular matrix (ECM) components [7,8]. As a β-hemolytic bacterium, GBS has the hemolytic property, which is essential for immune evasion and further infection [7,8]. Immune evasion factors include C5α peptidase (ScpB), hemolytic pigment, superoxide dismutase (SodA), HylB, and so on. Several classes of resistance to AMPs factors have been discovered, like D-alanylation of LTA and penicillin-binding proteins (PBPs). In addition, pili possess the ability to mediate GBS resistance to AMPs [7,8,9]. Recently, other virulence factors, cyclic di-AMP and cell wall-anchored ectonucleotidase (CdnP) have been discovered [10].
The abuse of antibiotics to control streptococcosis can cause many problems, such as the resistance of strains [11,12]. Moreover, the application regimes for antibiotics can be inaccurate, and antibiotics can deleteriously affect food quality and safety. Vaccines—which induce adaptive immune responses—can overcome some of these challenges. Given their potent efficacy, the development of vaccines for preventing GBS outbreaks has been widely investigated, including inactivated vaccines, attenuated vaccines, subunit vaccines, and DNA vaccines.

2. The Etiology of Tilapia Streptococcosis

Several studies have sought to identify the pathogen responsible for tilapia streptococcosis. Although this topic has been controversial, it is now acknowledged that the major pathogen for tilapia streptococcosis affecting aquaculture is Streptococcus agalactiae (GBS) [13]. In addition, there is evidence that Streptococcus iniae can cause similar disease symptoms, and it is now clear that many aspects contribute to outbreaks of tilapia streptococcosis, including environmental conditions as well as the presence of certain viruses and fungi [14,15].
S. agalactiae, a Gram-positive bacterium, has a spherical shape with dimensions ranging from 0.2 to 1.0 microns in diameter [16]. When cultured, S. agalactiae may grow in pairs or chains. It is a facultative anaerobe and is catalase and oxidase negative; it possesses the capacity for lactic acid fermentation [17] and is classified into Lancefield group B streptococcus (based on the presence and type of its surface antigens) [18]. The serotypes of GBS strains are assessed based on a capsular polysaccharide antigen, and to date, GBS has been classified into ten kinds of distinct serotypes, including Ia, Ib, II-IX [19,20]. Among them, serotypes Ia, Ib, II, and III are the most prevalent in tilapia infections. Notably, in 2018, our group identified that among others also the IX serotype was an important agent in tilapia streptococcosis [11]. This serotype has the potential to become a major infectious strain for tilapia.
Tilapia streptococcosis is mainly observed in temperate and tropical tilapia-culturing areas, including China [13], Malaysia [21], Austria [22], Brazil [23], Columbia [24], and Thailand [25], among other areas. Typically, large-scale infections of tilapia will breakthrough in relatively warm seasons, especially summer. The mortality rate can reach 50–70% in less than a week [26].
Although there is variability among tilapia, some of the most common symptoms and pathological signs include erratic swimming and loss of orientation, unilateral or bilateral exophthalmia (also named “pop-eye”), anorexia, abdominal distention, darkening of the skin, and hemorrhaging skin around the anus or at the base of the fins, as well as abdominal dropsy, pale, but enlarged liver, inflammations around the heart and kidney, and meningitis [26,27]. There has been heated discussion about potential modes of transmission: the introduction of infected fish is the most common mode, yet there is also solid evidence supporting vertical transmission [26].

3. Virulence Factors

3.1. Capsular Polysaccharide

The capsular polysaccharide (CPS) is a pathogenic factor widely distributed among Streptococcus serotypes; this molecule comprises glucose, galactose, N-acetylglucosamine, and N-acetylneuraminate [28]. CPS has been a traditional epidemiological tool for investigating S. agalactiae infections in humans [29]; it is generally used for strain typing. CPS is known to contribute to disease severity [30], and molecular serotyping techniques have an elevated discriminatory power for epidemiological studies [31].
Experiments have shown that bacterial cells lacking the capacity to produce CPS lost their virulence in a neonatal rat model of lethal group B Streptococcus infection [32,33]. Similarly, using tilapia challenge assays, CPS-deficient GBS showed attenuated pathogenesis. And this mutant GBS was also cleared more easily from tilapia spleen tissue compared to the wild type GBS strain examined [34]. It is now understood that CPS can suppress the aggregation of the complement factor C3b to inhibit phagocytotic killing by host cells [35,36]. Sialylated CPS mimics cell surface carbohydrate epitopes and thereby decreases host immune recognition [37]. GBS is known to regulate CPS production in response to environmental signals to adjust the capacity for adherence and host invasion [38]. Further, Barato et al. (2016) used an infection model and showed that mutant GBS cells (unencapsulated) displayed increased adhesion to the tilapia intestinal epithelium [39].

3.2. CAMP Factor

The CAMP factor (also called co-hemolysin) is encoded by the cfb gene. This is an extracellular protein of 23.5 kDa [40] that functions to promote GBS pathogenesis [41,42,43]. In vivo assays have shown that partially purified CAMP factors can lead to death in rabbits [44]. Briefly, currently, understanding of the pathogenic impacts of CAMP includes its oligomerization to support forming discrete pores on host membranes and its binding to glycosylphosphatidylinositol (GPI)-anchored proteins, which can promote cell lysis [45]. Recently, two research groups identified the structure of CAMP, which provided more details about its perforating function [46,47]. Podbielski et al. (1994) demonstrated that a full-sized recombinant CAMP-factor exerts co-hemolytic activity [42]. Note that cfb is widely used as an identification marker for Streptococcus agalactiae due to its exclusive expression in GBS [40]; however, more recent studies have shown that there are homologs of cfb in Streptococcus pyogenes, Listeria monocytogenes, Mobiluncus curtisis, and Propionibacterium acnes [48,49].

3.3. HAase (Hyaluronidase)

Encoded by the cylB gene, GBS promotes its invasion of hosts by secreting HAase to specifically hydrolyze the host-cell-wall component hyaluronic acid into unsaturated disaccharide units as the end product. Its degradative enzyme function by cleaving the glycosidic bond between N-acetyl-β-D-glucosamine and D-glucuronic acid residues destroys the host’s normal connective tissues and nervous system, which leads to expose to the host tissue cells to bacterial toxins and further facilitate deep tissue penetration during infection [50,51]. It is also now clear that GBS uses HAase to counteract host immune responses [52]. Whereas a host can normally promptly respond by generating hyaluronan (HA) polymers, from which small fragments ultimately combine with Toll-like receptors (TLRs) to elicit inflammatory responses, the secreted HAase from GBS degrades proinflammatory HA fragments down into their component disaccharides, thus blocking the host’s TLR2/4 signaling responses [52].

3.4. Cel-EIIB

The GBS phosphotransferase system (PTS) system, which is known to regulate bacterial virulence, can phosphorylated sugar substrates, including lactose, fructose, cellobiose, mannose, and sorbose [53]. Cellobiose-PTS (cel-PTS) is ubiquitously expressed in different serotypes of GBS, and strains genetically deficient for cel-PTS have decreased colonization ability and virulence [54]. One study showed that there are different expression levels of the cel-PTS component cel-EIIB between low and high virulence GBS [55,56]. Xu et al. (2018) reported that a cel-EIIB knockout strain showed a decreased ability of cellobiose utilization, as well as significantly reduced biofilm formation ability compared with the wild-type GBS strain [57]. It was also notable that knockout of cel-EIIB caused a 20% decrease in the accumulative mortality of tilapia due to GBS infection; the authors speculated that cel-EIIB knockout significantly decreased invasion and colonization efficiency [57].

3.5. LuxS/AI-2 Quorum Sensing System

Quorum sensing (QS) refers to a coordinated mode of gene expression regulation that supports bacterial communication and group activity [58]. The LuxS gene encodes S-ribosyl homocysteinase, which catalyzes the biosynthesis of the QS signaling molecule known as autoinducer 2 (AI-2), a furanosyl borate diester. LuxS is conserved among GBS serotypes and is ubiquitously expressed [59,60]. This 483 bp gene is composed of a conserved active center (H57, H61, C127) and a Zn2+-binding site (H-T-I-E-H) [61]. Ma et al. have reported that a mutant strain deficient for LuxS was defective for quorum sensing and displayed a more than 30-fold reduction in acid resistance compared to the wild-type strain [61]. The cell adherence was also decreased in the mutant strain. A study in tilapia showed a significantly decreased extent of infection and demonstrated that reintroducing LuxS into the luxS rescued hypervirulence [61].

3.6. Other Virulence Factors

Besides the aforementioned virulence factors, there are also other virulence factors, which have not been experimentally confirmed in tilapia GBS infections, but, which have been widely studied in mammalian GBS infections, including, for example, fibrinogen receptor (FbsA, FbsB and FbsC) [62], superoxide dismutase (SOD), serine-threonine protein kinase (STPK), C5a peptidase (scp) [14], serine-rich repeat glycoproteins (srr1 and srr2) [63], β-hemolysin/cytolysin (cylE) [64], pili [65], proteins Cα (bca) [66], neul [67], and α-like protein (Alp) [68], among others.

4. Progress Made in GBS Vaccinology

4.1. Inactivated Whole-Cell Vaccine

Formalin-inactivated and heat-killed whole-cell vaccines have been widely used in studies of pathogenic infection. Pasnik et al. (2005) conducted experiments wherein the relative percentage of survival (RPS) upon post-vaccination challenge was 49–50% [69,70]. Evans et al. (2004) performed similar assays using an inactivated whole-cell vaccination approach and challenge methods and recorded an RPS of 80% [71]. Thus, inactivated whole-cell approaches are useful for inducing immune responses to support basic studies and can confer protection against GBS infection (Table 1).

4.2. SAGs (S. Agalactiae Ghosts)

So-called S. agalactiae ghosts (SAGs) are empty cell envelopes from dead cells that have been explored as vaccine candidates owing to the presence of too many innate immunostimulatory agonists and their potent ability to activate innate and adaptive immunity responses. SAGs have been shown to induce cytokine secretion, which, in turn, contributes to the recruitment of T and B cells to lymph nodes. The recruitment of lymphocytes can increase the recognition of foreign antigens to elicit strong immune responses [72]. Wang et al. (2018) reported that immunization of tilapia with SAG elicited significantly higher resistance against GBS compared to the PBS-immunized controls [73] (Table 1). Moreover, SAG-immunized tilapia has stronger innate immunity (including phagocytic activity, lysozyme and superoxide dismutase activities) and adaptive immunity, especially IgM antibody titers. Finally, they found that the SAG-immunized tilapia showed significantly higher cytokine production (IL-1β, TNF-α, and TGF-β) than control animals.

4.3. Sip (Surface Immunogenic Protein)

There are some issues like inter-batch variability that can make use of inactivated whole-cell approaches less attractive than using vaccination approaches, which rely on more narrowly focused antigens (especially subunit vaccines) [74,75].
Sip is encoded by a gene in the GBS chromosome; this cell surface protein has been demonstrated as highly conserved; it is present in all GBS serotypes [76,77]. Given this broad distribution, it is unsurprising that Sip has been explored as a candidate for developing a subunit vaccine against GBS infection [78,79]. A 2014 study explored the immunogenicity of a Sip DNA vaccine, reporting an RPS value of 57% [79]; this was considered to represent relative effective immunoprotection. He et al. (2014) evaluated the truncated surface immunogenic protein (tSip) subunit vaccine and reported an RPS of 90% [78] (Table 1).

4.4. Surface Antigen Metabolic Enzymes

There have been attempts to assess the immunogenicity of enzymes present at the GBS surface, including phosphoglycerate kinase (PGK) [80], ornithine carbamoyl-transferase (OCT) [80], and the GapA subunit of glyceraldehyde-3-phosphate dehydrogenase of GBS [75,80]. PGK is responsible for its virulence and immunoprotective functions [81]. Using the recombinant PGK protein, Wang et al. (2014) found that PGK (as a subunit vaccine) against GBS infection of tilapia gave an RPS value of 82.4% [80]. The same study investigated OCT, which catalyzes the production of citrulline and phosphate from ornithine and carbamoyl phosphate substrates. Using recombinant OCT protein as a vaccine subunit gave an RPS value of 58.8%. Another study examining GapA as a subunit vaccine showed an RPS value of 63.3% [75] (Table 1).

4.5. Fibrinogen-Binding Protein A, α-Enolase, and GroEL

FbsA and enolase are adhesion proteins on the surface of bacteria. Yi et al. (2014) assessed potential immune protection against GBS infection upon vaccination with FbsA and Enolase, reporting RPS values of 40.63% and 62.50%, respectively [82]. The above data stated that the FbsA and enolase are multifaceted functions, including activating the host’s innate immune responses and relevant antibody responses and immunoprotection function. Thus, both of them can be efficient subunit vaccine candidates (Table 1).
GroEL is a heat shock protein (Hsp) of the chaperonin family of molecular chaperones; it is ubiquitously expressed in many bacteria [83]. This highly conserved chaperone functions in the proper folding of proteins. Li et al. (2019) reported that immunization of tilapia with GroEL (delivered with FC or FIC adjuvants) conferred protection against GBS, with an RPS value of 68.61% [74]. At the same time, which can increase antibody titers by promoting lymphocyte proliferation [74,84] (Table 1).
Table 1. Inactivated whole-cell vaccines and subunit vaccines explored for controlling tilapia streptococcosis.
Table 1. Inactivated whole-cell vaccines and subunit vaccines explored for controlling tilapia streptococcosis.
VaccineRPSAdjuvantVaccinationChallengeYearReference
Inactivated49–80%WithoutIntraperitonealIntraperitoneal2004–2005[69,70,71]
Inactivated97%Without/feed-based adjuvantOralIntraperitoneal2004[71]
Sip57%--IntragastricalIntraperitoneal2014[79]
tSip90%FIAIntraperitonealIntraperitoneal2014[78]
PGK82.4%Montanide ISA 763 AVGIntraperitonealIntraperitoneal2014[80]
OCT58.8%Montanide ISA 763 AVGIntraperitonealIntraperitoneal2014[80]
GapA63.3%FCA + FIAIntraperitonealIntraperitoneal2016[75]
GapA45.6%Montanide ISA 763 AVGIntraperitonealIntraperitoneal2016[75]
FbsA40.63%AdjuvantIntraperitonealIntraperitoneal2014[82]
Enolase62.50%AdjuvantIntraperitonealIntraperitoneal2014[82]
LIC70%WithoutFeed-basedIntraperitoneally2014[85]
GroEL68.61%FCA + FIASubcutaneousIntraperitoneally2019[74]
CWSAP 46577.5%FIAIntraperitonealIntraperitoneal2016[86]
CWSAP103572.5%FIAIntraperitonealIntraperitoneal2016[86]
ISP48.61%FCA + FIAIntraperitonealIntraperitoneal2016[87]
SAG86.67%--IntraperitonealIntraperitoneal2018[73]
Notes: Inactivated—inactivated whole-cell vaccine, RPS—relative percentage of survival, FIA—Freund’s incomplete adjuvant, FCA—Freund’s complete adjuvant,—DNA vaccine without adjuvant, without—without adjuvant, LIC—cell wall surface anchor family protein, CWSAP—cell wall surface anchor family protein, ISP—immunogenic secreted protein.

4.6. Alternatives to Vaccines for Controlling Tilapia Streptococcosis

There has been research into other methods for managing, preventing, and treating tilapia streptococcosis, including intestinal microbiota modifier [88,89] and bacteriophages. Probiotics investigated to date include Bacillus subtilis, Bacillus licheniformis, and Lactobacillus rhamnosus. Liu et al. (2017) [90] reported that Bacillus subtilis could enhance tilapia growth, digestive enzyme activities, innate immune responses, and GBS resistance. Abarike et al. (2018) [91] reported that Bacillus licheniformis could also promote tilapia growth, immune responses, and GBS resistance. Xia et al. (2018) [92] showed supplementation with Lactobacillus rhamnosus increased tilapia growth, intestinal microbiota, immune responses, and GBS resistance. As for bacteriophages, Luo et al. (2018) [93] showed that tilapia treated with the bacteriophage HN48 had about 60% greater survival than control animals. Notably, there have been additional proposals about how to control GBS infections and tilapia streptococcosis, including, for example, the use of Chinese medicinal herbs [94] and alteration of water temperature [65,95], among others.

5. Conclusions and Prospects

This review has considered research progress into tilapia streptococcosis regarding etiology, pathology, epidemiology, and prospects for control, including immunology-based approaches. As genomics, transcriptomics, proteomics, metabonomics, and bioinformatics continue to mature, it is clear that ever-more-powerful tools can be applied to identify and better understand the biological basis and control prospects for GBS infections of many animals generally and of tilapia streptococcosis in particular [5,12,96]. At the same time, an increasing number of vaccine candidates and adjuvant (including Freund’s adjuvant, Montanide adjuvant, aluminum-based adjuvants and others) are being explored, and this is a promising area for control of disease outbreaks in aquaculture. There remain virulence factors of GBS isolated from tilapia, which are still not fully understood, so there is a knowledge gap for active research teams to fill. In the near future, it seems likely that antibiotic treatment will remain the main control approach; however, in the long run, as scientific research prompts breakthroughs in understanding and developing effective technologies, there may be some innovative treatments and control measures like vaccines plus probiotic and immune enhancers.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

I would like to thank John Hugh Snyder (Director of Scientific Literature at NIBS) and Shun Wei for their careful and critical reading of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Suanyuk, N.; Kong, F.; Ko, D.; Gilbert, G.L.; Supamattaya, K. Occurrence of rare genotypes of Streptococcus agalactiae in cultured red tilapia Oreochromis sp. and Nile tilapia O. niloticus in Thailand—Relationship to human isolates? Aquaculture 2008, 284, 35–40. [Google Scholar] [CrossRef]
  2. Wang, E.L.; Wang, K.Y.; Chen, D.F.; Geng, Y.; Huang, L.Y.; Wang, J.; He, Y. Molecular cloning and bioinformatic analysis of the Streptococcus agalactiae neuA gene isolated from tilapia. Genet. Mol. Res. GMR 2015, 14, 6003–6017. [Google Scholar] [CrossRef]
  3. Wang, R.; Li, L.; Huang, Y.; Luo, F.; Liang, W.; Gan, X.; Huang, T.; Lei, A.; Chen, M.; Chen, L. Comparative genome analysis identifies two large deletions in the genome of highly-passaged attenuated Streptococcus agalactiae strain YM001 compared to the parental pathogenic strain HN016. BMC Genom. 2015, 16, 897. [Google Scholar] [CrossRef] [Green Version]
  4. Chen, M.; Li, L.P.; Wang, R.; Liang, W.W.; Huang, Y.; Li, J.; Lei, A.Y.; Huang, W.Y.; Gan, X. PCR detection and PFGE genotype analyses of streptococcal clinical isolates from tilapia in China. Vet. Microbiol. 2012, 159, 526–530. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, Z.; Yu, A.; Lan, J.; Zhang, Y.; Zhang, H.; Li, Y.; Hu, M.; Cheng, J.; Wei, S.; Lin, L. Draft Genome Sequence of an Attenuated Streptococcus agalactiae Strain Isolated from the Gut of a Nile Tilapia (Oreochromis niloticus). Genome Announc. 2017, 5, e01627-16. [Google Scholar] [CrossRef] [Green Version]
  6. Surve, M.V.; Anil, A.; Kamath, K.G.; Bhutda, S.; Sthanam, L.K.; Pradhan, A.; Srivastava, R.; Basu, B.; Dutta, S.; Sen, S. Membrane Vesicles of Group B Streptococcus Disrupt Feto-Maternal Barrier Leading to Preterm Birth. PLoS Pathog. 2016, 12, e1005816. [Google Scholar] [CrossRef] [PubMed]
  7. Armistead, B.; Oler, E.; Adams Waldorf, K.; Rajagopal, L. The Double Life of Group B Streptococcus: Asymptomatic Colonizer and Potent Pathogen. J. Mol. Biol. 2019, 431, 2914–2931. [Google Scholar] [CrossRef] [PubMed]
  8. Vornhagen, J.; Adams Waldorf, K.M.; Rajagopal, L. Perinatal Group B Streptococcal Infections: Virulence Factors, Immunity, and Prevention Strategies. Trends Microbiol. 2017, 25, 919–931. [Google Scholar] [CrossRef]
  9. Rajagopal, L. Understanding the regulation of Group B Streptococcal virulence factors. Futur. Microbiol. 2009, 4, 201–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Andrade, W.A.; Firon, A.; Schmidt, T.; Hornung, V.; Fitzgerald, K.A.; Kurt-Jones, E.A.; Trieu-Cuot, P.; Golenbock, D.T.; Kaminski, P.A. Group B Streptococcus Degrades Cyclic-di-AMP to Modulate STING-Dependent Type I Interferon Production. Cell Host Microbe 2016, 20, 49–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Zhang, Z.; Lan, J.; Li, Y.; Hu, M.; Yu, A.; Zhang, J.; Wei, S. The pathogenic and antimicrobial characteristics of an emerging Streptococcus agalactiae serotype IX in Tilapia. Microb. Pathog. 2018, 122, 39–45. [Google Scholar] [CrossRef]
  12. Zhang, Z.; Li, Y.; Hu, M.; Yu, A. Comparative transcriptome profiling reveals a mechanism of Streptococcus agalactiae resistance to florfenicol. Microb. Pathog. 2020, 142, 104098. [Google Scholar] [CrossRef]
  13. Wei, S.; Zhang, Z.; Li, Y.; Hu, M.; Yu, A.; Zhang, H.; Lan, J.; Zhang, Z.; Yan, Y.; Lin, L. Epidemic and antibiotic resistance of Streptococcus agalactiae isolated from tilapia (GIFT Oreochromis niloticus) in Guangdong Province. J. Fish. China 2016, 40, 503–511. [Google Scholar]
  14. Mishra, A.; Nam, G.H.; Gim, J.A.; Lee, H.E.; Jo, A.; Kim, H.S. Current Challenges of Streptococcus Infection and Effective Molecular, Cellular, and Environmental Control Methods in Aquaculture. Mol. Cells 2018, 41, 495–505. [Google Scholar]
  15. Laith, A.A.; Ambak, M.A.; Hassan, M.; Sheriff, S.M.; Nadirah, M.; Draman, A.S.; Wahab, W.; Ibrahim, W.N.; Aznan, A.S.; Jabar, A.; et al. Molecular identification and histopathological study of natural Streptococcus agalactiae infection in hybrid tilapia (Oreochromis niloticus). Vet. World 2017, 10, 101–111. [Google Scholar] [PubMed] [Green Version]
  16. Phares, C.R.; Lynfield, R.; Farley, M.M.; Mohle-Boetani, J.; Harrison, L.H.; Petit, S.; Craig, A.S.; Schaffner, W.; Zansky, S.M.; Gershman, K.; et al. Active Bacterial Core surveillance/Emerging Infections Program, N. Epidemiology of invasive group B streptococcal disease in the United States, 1999–2005. JAMA 2008, 299, 2056–2065. [Google Scholar] [CrossRef] [PubMed]
  17. Schuchat, A. Epidemiology of group B streptococcal disease in the United States: Shifting paradigms. Clin. Microbiol. Rev. 1998, 11, 497–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Lancefield, R.C. A Serological Differentiation of Specific Types of Bovine Hemolytic Streptococci (Group B). J. Exp. Med. 1934, 59, 441–458. [Google Scholar] [CrossRef] [Green Version]
  19. Slotved, H.C.; Kong, F.; Lambertsen, L.; Sauer, S.; Gilbert, G.L. Serotype IX, a Proposed New Streptococcus agalactiae Serotype. J. Clin. Microbiol. 2007, 45, 2929–2936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Gherardi, G.; Imperi, M.; Baldassarri, L.; Pataracchia, M.; Alfarone, G.; Recchia, S.; Orefici, G.; Dicuonzo, G.; Creti, R. Molecular epidemiology and distribution of serotypes, surface proteins, and antibiotic resistance among group B streptococci in Italy. J. Clin. Microbiol. 2007, 45, 2909–2916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Amal, M.N.; Zamri-Saad, M.; Iftikhar, A.R.; Siti-Zahrah, A.; Aziel, S.; Fahmi, S. An outbreak of Streptococcus agalactiae infection in cage-cultured golden pompano, Trachinotus blochii (Lacepede), in Malaysia. J. Fish Dis. 2012, 35, 849–852. [Google Scholar] [CrossRef] [PubMed]
  22. Bowater, R.O.; Forbes-Faulkner, J.; Anderson, I.G.; Condon, K.; Robinson, B.; Kong, F.; Gilbert, G.L.; Reynolds, A.; Hyland, S.; McPherson, G.; et al. Natural outbreak of Streptococcus agalactiae (GBS) infection in wild giant Queensland grouper, Epinephelus lanceolatus (Bloch), and other wild fish in northern Queensland, Australia. J. Fish Dis. 2012, 35, 173–186. [Google Scholar] [CrossRef] [PubMed]
  23. Pereira Ude, P.; Dos Santos, A.R.; Hassan, S.S.; Aburjaile, F.F.; Soares Sde, C.; Ramos, R.T.; Carneiro, A.R.; Guimaraes, L.C.; Silva de Almeida, S.; Diniz, C.A.; et al. Complete genome sequence of Streptococcus agalactiae strain SA20-06, a fish pathogen associated to meningoencephalitis outbreaks. Stand. Genom. Sci. 2013, 8, 188–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hernandez, E.; Figueroa, J.; Iregui, C. Streptococcosis on a red tilapia, Oreochromis sp. farm: A case study. J. Fish Dis. 2009, 32, 247–252. [Google Scholar] [CrossRef] [PubMed]
  25. Suwannasang, A.; Suanyuk, N.; Tantikitti, C. Phenotypic and genotypic characterization of Streptococcus agalactiae isolates from cultured tilapia in Thailand. Fish Shellfish Immunol. 2013, 34, 243–249. [Google Scholar] [CrossRef]
  26. Pradeep, P.J.; Suebsing, R.; Sirthammajak, S.; Kampeera, J.; Jitrakorn, S.; Saksmerprome, V.; Turner, W.; Palang, I.; Vanichviriyakit, R.; Senapin, S. Evidence of vertical transmission and tissue tropism of Streptococcosis from naturally infected red tilapia ( Oreochromis spp.). Aquac. Rep. 2016, 3, 58–66. [Google Scholar] [CrossRef] [Green Version]
  27. Amal, M.N.A.; Zamrisaad, M. Streptococcosis in tilapia (Oreochromis niloticus): A review. Pertanika J. Trop. Agric. Sci. 2011, 2, 195–206. [Google Scholar]
  28. Wessels, M.R.; Benedi, W.J.; Jennings, H.J.; Michon, F.; DiFabio, J.L.; Kasper, D.L. Isolation and characterization of type IV group B Streptococcus capsular polysaccharide. Infect. Immun. 1989, 57, 1089–1094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Dogan, B.; Schukken, Y.H.; Santisteban, C.; Boor, K.J. Distribution of serotypes and antimicrobial resistance genes among Streptococcus agalactiae isolates from bovine and human hosts. J. Clin. Microbiol. 2006, 43, 5899–5906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Zhao, Z.; Kong, F.X.; Gidding, H.; Morgan, J.; Gilbert, G. Distribution of genotypes and antibiotic resistance genes among invasive Streptococcus agalactiae (group B streptococcus) isolates from Australasian patients belonging to different age groups. Clin. Microbiol. Infect. 2008, 14, 260–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Kong, F.; Ma, L.; Gilbert, G.L. Simultaneous detection and serotype identification of Streptococcus agalactiae using multiplex PCR and reverse line blot hybridization. J. Med. Microbiol. 2005, 54 Pt 12, 1133–1138. [Google Scholar] [CrossRef]
  32. Rubens, C.E.; Wessels, M.R.; Heggen, L.M.; Kasper, D.L. Transposon mutagenesis of type III group B Streptococcus: Correlation of capsule expression with virulence. Proc. Natl. Acad. Sci. USA 1987, 84, 7208–7212. [Google Scholar] [CrossRef] [Green Version]
  33. Wessels, M.R.; Rubens, C.E.; Benedi, V.J.; Kasper, D.L. Definition of a bacterial virulence factor: Sialylation of the group B streptococcal capsule. Proc. Natl. Acad. Sci. USA 1989, 86, 8983–8987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhang, D.; Ke, X.; Liu, Z.; Cao, J.; Su, Y.; Lu, M.; Gao, F.; Wang, M.; Yi, M.; Qin, F. Capsular polysaccharide of Streptococcus agalactiae is an essential virulence factor for infection in Nile tilapia (Oreochromis niloticus Linn.). J. Fish Dis. 2019, 42, 293–302. [Google Scholar] [CrossRef] [PubMed]
  35. Marques, M.B.; Kasper, D.L.; Pangburn, M.K.; Wessels, M.R. Prevention of C3 deposition by capsular polysaccharide is a virulence mechanism of type III group B streptococci. Infect. Immun. 1992, 60, 3986–3993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Campbell, J.R.; Baker, C.J.; Edwards, M.S. Deposition and degradation of C3 on type III group B streptococci. Infect. Immun. 1991, 59, 1978–1983. [Google Scholar] [CrossRef] [Green Version]
  37. Lemire, P.; Houde, M.; Lecours, M.P.; Fittipaldi, N.; Segura, M. Role of capsular polysaccharide in Group B Streptococccus interactions with dendritic cells. Microbes Infect. 2012, 14, 1064–1076. [Google Scholar] [CrossRef] [PubMed]
  38. Nizet, V.; Kim, K.S.; Stins, M.; Jonas, M.; Chi, E.Y.; Nguyen, D.; Rubens, C.E. Invasion of brain microvascular endothelial cells by group B streptococci. Infect. Immun. 1997, 65, 5074–5081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Barato, P.; Martins, E.R.; Vasquez, G.M.; Ramirez, M.; Melo-Cristino, J.; Martinez, N.; Iregui, C. Capsule impairs efficient adherence of Streptococcus agalactiae to intestinal epithelium in tilapias Oreochromis sp. Microb. Pathog. 2016, 100, 30–36. [Google Scholar] [CrossRef] [PubMed]
  40. Bernheimer, A.W.; Linder, R.; Avigad, L.S. Nature and mechanism of action of the CAMP protein of group B streptococci. Infect. Immun. 1979, 23, 838–844. [Google Scholar] [CrossRef] [Green Version]
  41. Lang, S.; Palmer, M. Characterization of Streptococcus agalactiae CAMP factor as a pore-forming toxin. J. Biol. Chem. 2003, 278, 38167–38173. [Google Scholar] [CrossRef] [Green Version]
  42. Podbielski, A.; Blankenstein, O.; Lutticken, R. Molecular characterization of the cfb gene encoding group B streptococcal CAMP-factor. Med Microbiol. Immunol. 1994, 183, 239–256. [Google Scholar] [CrossRef]
  43. Jurgens, D.; Sterzik, B.; Fehrenbach, F.J. Unspecific binding of group B streptococcal cocytolysin (CAMP factor) to immunoglobulins and its possible role in pathogenicity. J. Exp. Med. 1987, 165, 720–732. [Google Scholar] [CrossRef]
  44. Skalka, B.; Smola, J. Lethal effect of CAMP-factor and UBERIS-factor—A new finding about diffusible exosubstances of streptococcus agalactiae and Streptococcus uberis. Zent. fur Bakteriol. 1 Abt. Orig. A Med. Mikrobiol. Infekt. und Parasitol. 1981, 249, 190–194. [Google Scholar] [CrossRef]
  45. Hensler, M.E.; Quach, D.; Hsieh, C.J.; Doran, K.S.; Nizet, V. CAMP factor is not essential for systemic virulence of Group B Streptococcus. Microb. Pathog. 2008, 44, 84–88. [Google Scholar] [CrossRef] [Green Version]
  46. Jin, T.; Brefo-Mensah, E.; Fan, W.; Zeng, W.; Li, Y.; Zhang, Y.; Palmer, M. Crystal structure of the Streptococcus agalactiae CAMP factor provides insights into its membrane-permeabilizing activity. J. Biol. Chem. 2018, 293, 11867–11877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Li, Y.; Zeng, W.; Li, Y.; Fan, W.; Ma, H.; Fan, X.; Jiang, J.; Brefo-Mensah, E.; Zhang, Y.; Yang, M.; et al. Structure determination of the CAMP factor of Streptococcus agalactiae with the aid of an MBP tag and insights into membrane-surface attachment. Acta Crystallogr. D Struct. Biol. 2019, 75 Pt 8, 772–781. [Google Scholar] [CrossRef] [Green Version]
  48. Gase, K.; Ferretti, J.J.; Primeaux, C.; McShan, W.M. Identification, cloning, and expression of the CAMP factor gene (cfa) of group A streptococci. Infect. Immun. 1999, 67, 4725–4731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Sorensen, M.; Mak, T.N.; Hurwitz, R.; Ogilvie, L.A.; Mollenkopf, H.J.; Meyer, T.F.; Bruggemann, H. Mutagenesis of Propionibacterium acnes and analysis of two CAMP factor knock-out mutants. J. Microbiol. Methods 2010, 83, 211–216. [Google Scholar] [CrossRef] [PubMed]
  50. Li, S.; Kelly, S.J.; Lamani, E.; Ferraroni, M.; Jedrzejas, M.J. Structural basis of hyaluronan degradation by Streptococcus pneumoniae hyaluronate lyase. EMBO J. 2000, 19, 1228–1240. [Google Scholar] [CrossRef] [Green Version]
  51. Mello, L.V.; De Groot, B.L.; Li, S.; Jedrzejas, M.J. Structure and flexibility of Streptococcus agalactiae hyaluronate lyase complex with its substrate. Insights into the mechanism of processive degradation of hyaluronan. J. Biol. Chem. 2002, 277, 36678–36688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kolar, S.L.; Kyme, P.; Tseng, C.W.; Soliman, A.; Kaplan, A.; Liang, J.; Nizet, V.; Jiang, D.; Murali, R.; Arditi, M.; et al. Group B Streptococcus Evades Host Immunity by Degrading Hyaluronan. Cell Host Microbe 2015, 18, 694–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Sutcliffe, I.C.; Black, G.W.; Harrington, D.J. Bioinformatic insights into the biosynthesis of the Group B carbohydrate in Streptococcus agalactiae. Microbiology 2008, 154 Pt 5, 1354–1363. [Google Scholar] [CrossRef] [Green Version]
  54. McAllister, L.J.; Ogunniyi, A.D.; Stroeher, U.H.; Paton, J.C. Contribution of a genomic accessory region encoding a putative cellobiose phosphotransferase system to virulence of Streptococcus pneumoniae. PLoS ONE 2012, 7, e32385. [Google Scholar]
  55. Li, W.; Su, Y.L.; Mai, Y.Z.; Li, Y.W.; Mo, Z.Q.; Li, A.X. Comparative proteome analysis of two Streptococcus agalactiae strains from cultured tilapia with different virulence. Vet. Microbiol. 2014, 170, 135–143. [Google Scholar] [CrossRef]
  56. Su, Y.L.; Feng, J.; Liu, C.; Li, W.; Xie, Y.D.; Li, A.X. Dynamic bacterial colonization and microscopic lesions in multiple organs of tilapia infected with low and high pathogenic Streptococcus agalactiae strains. Aquaculture 2017, 471, 190–203. [Google Scholar] [CrossRef]
  57. Xu, J.; Xie, Y.D.; Liu, L.; Guo, S.; Su, Y.L.; Li, A.X. Virulence regulation of cel-EIIB protein mediated PTS system in Streptococcus agalactiae in Nile tilapia. J. Fish Dis. 2019, 42, 11–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Miller, M.B.; Bassler, B.L. Quorum sensing in bacteria. Annu. Rev. Microbiol. 2001, 55, 165–199. [Google Scholar] [CrossRef] [Green Version]
  59. Wang, Y.; Zhang, W.; Wu, Z.; Zhu, X.; Lu, C. Functional analysis of luxS in Streptococcus suis reveals a key role in biofilm formation and virulence. Vet. Microbiol. 2011, 152, 151–160. [Google Scholar] [CrossRef]
  60. Zhao, L.; Xue, T.; Shang, F.; Sun, H.; Sun, B. Staphylococcus aureus AI-2 quorum sensing associates with the KdpDE two-component system to regulate capsular polysaccharide synthesis and virulence. Infect. Immun. 2010, 78, 3506–3515. [Google Scholar] [CrossRef] [Green Version]
  61. Ma, Y.; Hao, L.; Ke, H.; Liang, Z.; Ma, J.; Liu, Z.; Li, Y. LuxS/AI-2 in Streptococcus agalactiae reveals a key role in acid tolerance and virulence. Res. Vet. Sci. 2017, 115, 501–507. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, Y.; Li, L.; Huang, T.; Wu, W.; Liang, W.; Chen, M. The Interaction between Phagocytes and Streptococcus agalactiae (GBS) Mediated by the Activated Complement System is the Key to GBS Inducing Acute Bacterial Meningitis of Tilapia. Animals 2019, 9, 818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Su, Y.; Liu, C.; Deng, Y.; Cheng, C.; Ma, H.; Guo, Z.; Feng, J. Molecular typing of Streptococcus agalactiae isolates of serotype Ia from tilapia in southern China. FEMS Microbiol. Lett. 2019, 366, 13. [Google Scholar] [CrossRef] [PubMed]
  64. Kayansamruaj, P.; Pirarat, N.; Katagiri, T.; Hirono, I.; Rodkhum, C. Molecular characterization and virulence gene profiling of pathogenic Streptococcus agalactiae populations from tilapia (Oreochromis sp.) farms in Thailand. J. Vet. Diagn. Invest. 2014, 26, 488–495. [Google Scholar] [CrossRef] [Green Version]
  65. Kayansamruaj, P.; Pirarat, N.; Hirono, I.; Rodkhum, C. Increasing of temperature induces pathogenicity of Streptococcus agalactiae and the up-regulation of inflammatory related genes in infected Nile tilapia (Oreochromis niloticus). Vet. Microbiol. 2014, 172, 265–271. [Google Scholar] [CrossRef]
  66. Wu, W.; Li, L.; Liu, Y.; Huang, T.; Liang, W.; Chen, M. Multiomics analyses reveal that NOD-like signaling pathway plays an important role against Streptococcus agalactiae in the spleen of tilapia. Fish Shellfish Immunol. 2019, 95, 336–348. [Google Scholar] [CrossRef]
  67. Yamaguchi, M.; Hirose, Y.; Nakata, M.; Uchiyama, S.; Yamaguchi, Y.; Goto, K.; Sumitomo, T.; Lewis, A.L.; Kawabata, S.; Nizet, V. Evolutionary inactivation of a sialidase in group B Streptococcus. Sci. Rep. 2016, 6, 28852. [Google Scholar] [CrossRef] [Green Version]
  68. Jalalifar, S.; Havaei, S.A.; Motallebirad, T.; Moghim, S.; Fazeli, H.; Esfahani, B.N. Determination of surface proteins profile, capsular genotyping, and antibiotic susceptibility patterns of Group B Streptococcus isolated from urinary tract infection of Iranian patients. BMC Res. Notes 2019, 12, 437. [Google Scholar] [CrossRef]
  69. Pasnik, D.J.; Evans, J.J.; Klesius, P.H. Duration of protective antibodies and correlation with survival in Nile tilapia Oreochromis niloticus following Streptococcus agalactiae vaccination. Dis. Aquat. Org. 2005, 66, 129–134. [Google Scholar] [CrossRef]
  70. Pasnik, D.J.; Evans, J.J.; Panangala, V.S.; Klesius, P.H.; Shelby, R.A.; Shoemaker, C.A. Antigenicity of Streptococcus agalactiae extracellular products and vaccine efficacy. J. Fish Dis. 2005, 28, 205–212. [Google Scholar] [CrossRef]
  71. Evans, J.J.; Klesius, P.H.; Shoemaker, C.A. Efficacy of Streptococcus agalactiae (group B) vaccine in tilapia (Oreochromis niloticus) by intraperitoneal and bath immersion administration. Vaccine 2004, 22, 3769–3773. [Google Scholar] [CrossRef] [PubMed]
  72. Hajam, I.A.; Dar, P.A.; Won, G.; Lee, J.H. Bacterial ghosts as adjuvants: Mechanisms and potential. Vet. Res. 2017, 48, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wang, Q.; Wang, X.; Wang, X.; Feng, R.; Luo, Q.; Huang, J. Generation of a novel Streptococcus agalactiae ghost vaccine and examination of its immunogenicity against virulent challenge in tilapia. Fish Shellfish Immunol. 2018, 81, 49–56. [Google Scholar] [CrossRef] [PubMed]
  74. Li, W.; Li, Y.; Hu, Y.Z.; Mo, X.B.; Xu, G.H.; Xie, L.W.; Li, A.X. GroEL, a novel vaccine candidate of piscine Streptococcus agalactiae identified by immunoproteome. Fish Shellfish Immunol. 2019, 84, 377–383. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, Z.; Yu, A.; Lan, J.; Zhang, H.; Hu, M.; Cheng, J.; Zhao, L.; Lin, L.; Wei, S. GapA, a potential vaccine candidate antigen against Streptococcus agalactiae in Nile tilapia (Oreochromis niloticus). Fish Shellfish Immunol. 2017, 63, 255–260. [Google Scholar] [CrossRef] [PubMed]
  76. Rioux, S.; Martin, D.; Ackermann, H.W.; Dumont, J.; Hamel, J.; Brodeur, B.R. Localization of surface immunogenic protein on group B streptococcus. Infect. Immun. 2001, 69, 5162–5165. [Google Scholar] [CrossRef] [Green Version]
  77. Xue, G.; Yu, L.; Li, S.; Shen, X. Intranasal immunization with GBS surface protein Sip and ScpB induces specific mucosal and systemic immune responses in mice. FEMS Immunol. Med. Microbiol. 2010, 58, 202–210. [Google Scholar] [CrossRef]
  78. He, Y.; Wang, K.Y.; Xiao, D.; Chen, D.F.; Huang, L.; Liu, T.; Wang, J.; Geng, Y.; Wang, E.L.; Yang, Q. A recombinant truncated surface immunogenic protein (tSip) plus adjuvant FIA confers active protection against Group B streptococcus infection in tilapia. Vaccine 2014, 32, 7025–7032. [Google Scholar] [CrossRef] [PubMed]
  79. Huang, L.Y.; Wang, K.Y.; Xiao, D.; Chen, D.F.; Geng, Y.; Wang, J.; He, Y.; Wang, E.L.; Huang, J.L.; Xiao, G.Y. Safety and immunogenicity of an oral DNA vaccine encoding Sip of Streptococcus agalactiae from Nile tilapia Oreochromis niloticus delivered by live attenuated Salmonella typhimurium. Fish Shellfish Immunol. 2014, 38, 34–41. [Google Scholar] [CrossRef] [PubMed]
  80. Wang, Y.T.; Huang, H.Y.; Tsai, M.A.; Wang, P.C.; Jiang, B.H.; Chen, S.C. Phosphoglycerate kinase enhanced immunity of the whole cell of Streptococcus agalactiae in tilapia, Oreochromis niloticus. Fish Shellfish Immunol. 2014, 41, 250–259. [Google Scholar] [CrossRef]
  81. Lee, K.W.; Shalaby, K.A.; Thakur, A.; Medhat, A.M.; Karim, A.M.; LoVerde, P.T. Cloning of the gene for phosphoglycerate kinase from Schistosoma mansoni and characterization of its gene product. Mol. Biochem. Parasitol. 1995, 71, 221–231. [Google Scholar] [CrossRef]
  82. Yi, T.; Li, Y.W.; Liu, L.; Xiao, X.X.; Li, A.X. Protection of Nile tilapia (Oreochromis niloticus L.) against Streptococcus agalactiae following immunization with recombinant FbsA and α-enolase. Aquaculture 2014, 428–429, 35–40. [Google Scholar] [CrossRef]
  83. Zeilstra-Ryalls, J.; Fayet, O.; Georgopoulos, C. The universally conserved GroE (Hsp60) chaperonins. Annu. Rev. Microbiol. 1991, 45, 301–325. [Google Scholar] [CrossRef] [PubMed]
  84. Yi, L.; Wang, Y.; Ma, Z.; Lin, H.X.; Xu, B.; Grenier, D.; Fan, H.J.; Lu, C.P. Identification and characterization of a Streptococcus equi ssp. zooepidemicus immunogenic GroEL protein involved in biofilm formation. Vet. Res. 2016, 47, 50. [Google Scholar] [CrossRef] [Green Version]
  85. Nur-Nazifah, M.; Sabri, M.Y.; Siti-Zahrah, A. Development and efficacy of feed-based recombinant vaccine encoding the cell wall surface anchor family protein of Streptococcus agalactiae against streptococcosis in Oreochromis sp. Fish Shellfish Immunol. 2014, 37, 193–200. [Google Scholar] [CrossRef] [PubMed]
  86. Liu, H.; Zhang, S.; Shen, Z.; Ren, G.; Liu, L.; Ma, Y.; Zhang, Y.; Wang, W. Development of a vaccine against Streptococcus agalactiae in fish based on truncated cell wall surface anchor proteins. Vet. Rec. 2016, 179, 359. [Google Scholar] [CrossRef] [PubMed]
  87. Li, W.; Wang, H.Q.; He, R.Z.; Li, Y.W.; Su, Y.L.; Li, A.X. Major surfome and secretome profile of Streptococcus agalactiae from Nile tilapia (Oreochromis niloticus): Insight into vaccine development. Fish Shellfish Immunol. 2016, 55, 737–746. [Google Scholar] [CrossRef] [PubMed]
  88. Opiyo, M.A.; Jumbe, J.; Ngugi, C.C.; Charo-Karisa, H. Dietary administration of probiotics modulates non-specific immunity and gut microbiota of Nile tilapia (Oreochromis niloticus) cultured in low input ponds. Int. J. Vet. Sci. Med. 2019, 7, 1–9. [Google Scholar] [CrossRef] [Green Version]
  89. Xia, Y.; Wang, M.; Gao, F.; Lu, M.; Chen, G. Effects of dietary probiotic supplementation on the growth, gut health and disease resistance of juvenile Nile tilapia (Oreochromis niloticus). Anim. Nutr. 2020, 6, 69–79. [Google Scholar] [CrossRef] [PubMed]
  90. Liu, H.; Wang, S.; Cai, Y.; Guo, X.; Cao, Z.; Zhang, Y.; Liu, S.; Yuan, W.; Zhu, W.; Zheng, Y.; et al. Dietary administration of Bacillus subtilis HAINUP40 enhances growth, digestive enzyme activities, innate immune responses and disease resistance of tilapia, Oreochromis niloticus. Fish Shellfish Immunol. 2017, 60, 326–333. [Google Scholar] [CrossRef] [PubMed]
  91. Abarike, E.D.; Cai, J.; Lu, Y.; Yu, H.; Chen, L.; Jian, J.; Tang, J.; Jun, L.; Kuebutornye, F.K.A. Effects of a commercial probiotic BS containing Bacillus subtilis and Bacillus licheniformis on growth, immune response and disease resistance in Nile tilapia, Oreochromis niloticus. Fish Shellfish Immunol. 2018, 82, 229–238. [Google Scholar] [CrossRef] [PubMed]
  92. Xia, Y.; Lu, M.; Chen, G.; Cao, J.; Gao, F.; Wang, M.; Liu, Z.; Zhang, D.; Zhu, H.; Yi, M. Effects of dietary Lactobacillus rhamnosus JCM1136 and Lactococcus lactis subsp. lactis JCM5805 on the growth, intestinal microbiota, morphology, immune response and disease resistance of juvenile Nile tilapia, Oreochromis niloticus. Fish Shellfish Immunol. 2018, 76, 368–379. [Google Scholar] [CrossRef] [PubMed]
  93. Luo, X.; Liao, G.; Liu, C.; Jiang, X.; Lin, M.; Zhao, C.; Tao, J.; Huang, Z. Characterization of bacteriophage HN48 and its protective effects in Nile tilapia Oreochromis niloticus against Streptococcus agalactiae infections. J. Fish Dis. 2018, 41, 1477–1484. [Google Scholar] [CrossRef]
  94. Abarike, E.D.; Jian, J.; Tang, J.; Cai, J.; Yu, H.; Chen, L. Traditional Chinese Medicine Enhances Growth, Immune Response, and Resistance to Streptococcus agalactiae in Nile Tilapia. J. Aquat. Anim. Health 2019, 31, 46–55. [Google Scholar] [CrossRef]
  95. Tavares, G.C.; Carvalho, A.F.; Pereira, F.L.; Rezende, C.P.; Azevedo, V.A.C.; Leal, C.A.G.; Figueiredo, H.C.P. Transcriptome and Proteome of Fish-Pathogenic Streptococcus agalactiae Are Modulated by Temperature. Front. Microbiol. 2018, 9, 2639. [Google Scholar] [CrossRef] [PubMed]
  96. Zhang, Z.; Li, Y.; Hu, M.; Yu, A. Genome reanalysis to decipher resistome, virulome, and attenuated characters of attenuated Streptococcus agalactiae strain HZAUSC001. Microb. Pathog. 2020, 147, 104416. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, Z. Research Advances on Tilapia Streptococcosis. Pathogens 2021, 10, 558. https://doi.org/10.3390/pathogens10050558

AMA Style

Zhang Z. Research Advances on Tilapia Streptococcosis. Pathogens. 2021; 10(5):558. https://doi.org/10.3390/pathogens10050558

Chicago/Turabian Style

Zhang, Ze. 2021. "Research Advances on Tilapia Streptococcosis" Pathogens 10, no. 5: 558. https://doi.org/10.3390/pathogens10050558

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop