Journal List > J Vet Sci > v.18(S1) > 1041528

Lalsiamthara and Lee: Development and trial of vaccines against Brucella

Abstract

The search for ideal brucellosis vaccines remains active today. Currently, no licensed human or canine anti-brucellosis vaccines are available. In bovines, the most successful vaccine (S19) is only used in calves, as adult vaccination results in orchitis in male, prolonged infection, and possible abortion complications in pregnant female cattle. Another widely deployed vaccine (RB51) has a low protective efficacy. An ideal vaccine should exhibit a safe profile as well as enhance protective efficacy. However, currently available vaccines exhibit one or more major drawbacks. Smooth live attenuated vaccines suffer shortcomings such as residual virulence and serodiagnostic interference. Inactivated vaccines, in general, confer relatively low levels of protection. Recent developments to improve brucellosis vaccines include generation of knockout mutants by targeting genes involved in metabolism, virulence, and the lipopolysaccharide synthesis pathway, as well as generation of DNA vaccines, mucosal vaccines, and live vectored vaccines, have all produced varying degrees of success. Herein, we briefly review the bacteriology, pathogenesis, immunological implications, candidate vaccines, vaccinations, and models related to Brucella.

Introduction

Recently, brucellosis has garnered increased importance due to its zoonotic potential and possible use in bio-warfare. Though the disease-causal agent relationship was established in the 19th century [14], it never reached the center of attention in the medical field. In 2009, the World Health Organization's, third International Conference on Neglected Zoonotic Diseases assessed and acknowledged initiative works into the burden of key diseases such as bovine tuberculosis, brucellosis, toxoplasmosis, and zoonotic schistosomiasis [6]. Brucellosis is a sub-acute or chronic disease, and in cattle, sheep, goats, other ruminants, and pigs the initial phase following infection is often not apparent [29]. Recently recognized Brucella types associated with disease in marine animals may also have the capacity to cause disease in humans [2972].
Control of zoonotic diseases in human populations has relied heavily on the control of animal disease. Over the last century, human brucellosis has been controlled by vaccination and culling within cattle, goat, and sheep herds [28]. Yet, despite past and current efforts to eradicate brucellosis many human cases are reported annually worldwide. There is a direct link between the disease and the economic status of a country [78]. It was the opinion of experts that resulted in the suggestion that vaccination efforts alone are insufficient to effectively control the disease. Still, the efficacy of vaccination-only strategies has not been seriously evaluated, and it depends in large part on the quality of the vaccine employed [28]. Since, brucellosis is closely tied to economic stature, improving livelihood would also alleviate disease burden and transmission in endemic areas.
Due to importance of controlling brucellosis in the animal population, Brucella abortus S19 and Brucella melitensis Rev. 1 vaccines have been widely used in certain developed countries, but both the vaccines induce abortions in pregnant animals and are virulent for humans; moreover, they elicit anti-Brucella antibodies that interfere with serodiagnosis. Further, Rev. 1 is streptomycin resistant, an important antibiotic used to treat the disease. However, the residual virulence issue can be solved by using further attenuated mutant strains, and the serodiagnosis problem can be solved by using lipopolysaccharide (LPS)-disrupted mutant strains, immunization via the conjunctival route, avoidance of adult vaccination, and by an individual serological follow up. Hence, a vaccine embodying all such prerequisite properties would be a boon to brucellosis control and a major breakthrough [7085].

Understanding Brucella for Strategic Development of Vaccine

Brucella organism

Interestingly, Brucella organisms are very closely related to plant microbes. It is remarkable how the proto-Brucella may jump between the two kingdoms, from plant to animal. Brucellae are classified under α-Proteobacteria phylogenetically, which are related to plant pathogens and symbionts such as Rhizobium, Agrobacterium, and an animal endosymbiont Wolbachia, to intracellular animal parasites such as Bartonella and Rickettsia, and to opportunistic and free-living bacteria like Ochrobactrum and Caulobacter [3269]. Accurate identification of Brucellae and differentiating them from closely related opportunistic Ochrobactrum is important for accurate diagnosis [83]. It is not well understood whether pre-existing anti-Ochrobactrum immunity may hinder successful vaccination against Brucella. Brucellae are gram-negative, non-sporulating, capnophilic, facultative intracellular bacteria [28].

Brucella genetic composition

With the increased availability of the Brucella whole genome sequence, a wide variety of genetic modification studies related to microbiological and vaccinology have been undertaken. The type strains and other representative strains of the six species of Brucella form a very tight cluster, with G + C values between 57.9% and 59.2%, DNA homologies close to 100%, Tm values between 79.2℃ and 80.5℃, and rRNA binding values between 0.166% and 0.198% [32]. Brucella species show a considerably high degree of homology, except for B. ovis which has been reported to show slight restriction endonuclease digestion pattern differences from other Brucellae [73]. The vaccine strain S19 genome is 3.2 Mbp and comprises two circular chromosomes: one 2122487 bp long and the other 1161449 bp long. The average G + C content of the two chromosomes of S19 strain is 57% [30]. It was shown that the S19 genome has remarkable similarity in size and structure to those of its virulent relatives, B. abortus strains 9–941 and 2308 [2249]. The S19 genome sequence shows over 99.5% similarity to the genomes of strains 9–941 and 2308 [30].

Brucella host interaction and pathogenesis

Understanding host-pathogen interactions and pathogenesis helps in developing rational vaccine designs. Like other pathogenic intracellular bacteria, Brucella infection requires the four following steps: adherence, invasion, establishment, and dissemination within the host [58]. The most striking characteristics of Brucella infection is that it operates in “stealth mode”, i.e., it can evade immune detection [961]. This mode of infection eventually leads to chronicity and prolonged infection [931]. Earlier ultrastructural work characterizing the morphology of B. abortus-infected cells revealed that Brucellae multiply in an intracellular multimembranous compartment that has similarities to the rough endoplasmic reticulum [5]. Weak interactions between host and Brucella LPS probably has a substantial role in intracellular survival [17]. The chronicity of brucellosis is multifactorial, utilizing both the ability of Brucella to evade immune detection and to adapt to intracellular survival inside both phagocytic and nonphagocytic cells [3180]. The inhibition of the phagosomal-lysosomal maturation pathway and the deviation of intracellular trafficking have important roles; in this scheme, the bacterium reaches its endoplasmic reticulum-derived replicative niche [20].

Brucella virulency with special reference to LPS

Unlike most common bacterial pathogens, Brucella species do not produce exotoxins, appendages such as flagella, pili, or fimbriae, antiphagocytic capsules or thick cell walls, resistant forms, and do not exhibit antigenic variation [39]. The success of invasion is due to the inability of the host's immune system to effectively detect or clear the bacteria, in this way the bacteria gain entry to its replicative niche and thus proliferate within professional and nonprofessional phagocytic host cells [1880]. The successful evasion of host immunity is attributed to the LPS of the organism. Brucella possesses a peculiar nonclassical LPS that does not exhibit strong endotoxic activities similar to those of classical LPSs from enterobacteria such as Escherichia coli [56]. Further, LPS is recognized as a major virulence determinant as naturally occurring isolates lacking LPS showed reduced survival inside the host system.
Brucella LPS is comprised of lipid A, core oligosaccharide, and O-polysaccharide (OPS). Lipid A contains 2, 3-diamino-2, 3-dideoxy-D-glucose (diaminoglucose) as backbone, amide, and ester-linked long chain saturated and hydroxylated fatty acids. The core oligosaccharide is composed of mannose, glucose, 2-amino-2, 6-dideoxy-D-glucose (quinovosamine), 2-amino-2-deoxy-D-glucose (glucosamine), 3-deoxy-D-manno-2-octulosonic acid, and unidentified sugars. The OPS is an unbranched homopolymer (n = 96–100) of 1, 2-linked 4, 6-dideoxy-4-formamido-α-D-mannopyranosyl [1669]. In murine macrophages, it has been demonstrated that OPS is specifically involved in inhibition of early fusion between Brucella suis containing-phagosomes and lysosomes In contrast, phagosomes containing rough mutants, which fail to express the O antigen, rapidly fuse with lysosomes [81]. Due to its central role in virulency, much research has been undertaken into the disruption of LPS biosynthesis. The aim of that research is to generate a mutant vaccine strain that has a perfect balance between attenuation and protection, and also enables differentiating infected from vaccinated animals (DIVA) [54102].
Apart from the LPS involvement in virulence, the virB operon, encoding a type IV secretion system homologous to those encoded by Agrobacterium tumefaciens virB, and the Bordetella pertussis ptl operon are present in the Brucella genome [86]. The A. tumefaciens virB operon encodes a pilus-like structure necessary for secretion of transfer DNA, and the B. pertussis ptl operon encodes an apparatus that allows the secretion of pertussis toxin, suggesting that Brucella may secrete regulatory DNA or protein for intracellular survival. Superoxide dismutase (SOD) forms part of the antioxidant defense system that protects bacteria from the toxic effects of reactive oxygen intermediates by converting superoxide radicals into hydrogen peroxide and oxygen [40], suggesting that SOD has a crucial role in Brucella intracellular survival. As anticipated, anti-Brucella SOD-based vaccines confer substantial amounts of protection [235293].

Brucella interaction with host immunity

During the initial phase of infection, approximately 90% of bacteria are killed by phagocytic cells. Dendritic cells and macrophages have important roles in clearing the infection; however, failure to lyse the organism within phagosomes leads to formation of Brucella-containing vacuoles and, thus, replication in this niche [61]. Brucella can also subvert the autophagy process in order to evade efficient clearing [4890]. Neutrophils may have an important role in subsiding the infection via phagocytosis, as Brucella does not replicate within neutrophils, but it does resist neutrophil-mediated bacterial killing [9].
Specific antibodies have important roles in reducing the initial phase of a Brucella infection; however, they have limited roles following intracellular localization. Hence, strong humoral immunity unaccompanied by cell-mediated immunity (CMI) cannot provide total protection against the Brucella organism. B lymphocytes may act in favor of the organism by providing a replication niche [43] or B cells may produce interleukin (IL)-10 cytokine thereby antagonizing the production of IL-12 and interferon-γ, which are important for orchestrating the CMI response. It has been demonstrated that mice deficient in B cells are highly resistant to Brucella infection [44]. Several researchers have demonstrated the importance of Th1 immune response in controlling Brucella infection, as well as the involvement of IL-12 and INF-γ [1161]. Hence, it is prudent to design vaccines that can augment the Th1 response while reducing the Th2 response.

Approaches for Development of Anti-Brucella Vaccines

The general considerations associated with, and a summary of the different classes of, Brucella vaccines are listed in Table 1.

Classical and commercial vaccine

The most widely used vaccine for the prevention of brucellosis in cattle is the B. abortus S19 vaccine, which remains the reference vaccine to which other vaccines are compared. It is used as a live vaccine and is normally given to female calves aged between 3 and 6 months as a single subcutaneous dose of 5–8 × 1010 viable organisms or as a reduced dose of from 3 × 108 to 3 × 109 organisms that can be administered subcutaneously to adult cattle. Alternatively, it can be administered to cattle of any age as either one or two doses of 5 × 109 viable organisms, given via the conjunctival route [50]. B. abortus S19 has the normal properties of the biovar 1 strain of B. abortus, but it does not require CO2 for growth, does not grow in the presence of benzylpenicillin (3 µg/mL = 5 IU/mL), thionin blue (2 µg/mL), or i-erythritol (1 mg/mL) (all final concentrations), and presents with high L-glutamate use [50].
B. abortus strain 45/20 is a rough strain that is able to protect guinea pigs and cattle from Brucella infection; however, reversions to the wild smooth type has limited its use as a live vaccine [6297].
B. abortus strain RB51, a rough attenuated organism, was originally derived from a rifampicin-resistant mutant of B. abortus strain 2308 and has replaced B. abortus S19 strain as a vaccine candidate in some developed countries. Strain RB51 is very stable and has no, or highly reduced, abortifacient characteristics [84]. Protective efficacy and immunity induced by strain RB51 is similar to or better than that induced by strain 19 [2459]. However, although strain RB51 has an excellent record of stability, it is rifampicin resistant to important antibiotics used in the treatment of brucellosis; moreover, it is still infectious to humans and the exact nature of its mutations have not been described. Recently, it was reported that RB51 vaccinated cattle in the Greater Yellowstone Area in the USA were still susceptible to brucellosis [76103].

Genetically modified Brucella mutant strains

To counteract the disadvantageous properties of the smooth Brucella vaccines, research attention had been focused on the development of rough phenotype vaccines with greater attenuation. Rough Brucella mutants lack the LPS immunodominant N-formylperosamine OPS and are substantially attenuated [7085]. Some rough (R) vaccines or candidates are spontaneous mutants selected after repeated passage on antibiotic-containing media. Disruption of per, pgm, wboA, and wbkA (genes involved in the LPS biosynthesis pathway) results in rough mutants [67101108], showing that empirically, R vaccines can be improved. Other notable mutants that have metabolic genes disrupted are the purL, purD, and purE mutants that affect the purine biosynthesis pathway genes [1], the lipid A fatty acid transporting gene bacA mutant [3857], the ferrochelatase hemH mutant [3], the type IV secretion virB mutant [34], and the phosphoglycerate kinase encoding gene pgk mutant [99]. Many mutants have shown promising results by exhibiting a protection level similar to or higher than RB51. In addition, the rough strains are DIVA enabled. Despite several encouraging results, trials have not been undertaken to evaluate the protective capabilities of these mutants in the target host. Thus, conclusive data and substantial definitive host-vaccine efficacy findings have not been validated. Also due to variations in evaluation protocols, accurate comparisons between the mutants have not been possible [45110].

Inactivated vaccines

Cell fractions and lysate

Cell components from the killed organism were tested during the early development of a Brucella vaccine. Preparations that were evaluated included outer membrane protein [68], soluble and insoluble extracts of cell envelopes [36107], whole killed cells [10], and periplasmic proteins and salt extractable proteins [94]. While infection-related issues were nil, several issues such as poor protection, local reactions, and serological problems hindered their popularity [64]. The local reactions were generally induced by adjuvants which were used to improve protective efficacy.

Subunit and DNA vaccine

Due to safety implications, several subunit vaccines have been evaluated. In addition, subunit vaccines have the advantage of being effective for pan-Brucella species because high homology protein can be selected as the candidate. Several subunit/DNA vaccine candidates have been evaluated with the central aim of evoking a Th1 response. These candidates include recombinant P39, bacterioferritin, L7/L12 [275], lumazine synthase [104], Bp26 and trigger factor together [109], InfC [21], L7/L12 [53], Omp16 and Omp19 [79], Omp25 [27], Omp28 [51], Omp31 [19], P39 [2], S-adenosyl-l-homocysteine hydrolase [111], DnaK and SurA [33], and SodC [71]. It has been demonstrated that subunit vaccines can be improved by including encapsulations such as escheriosomes and liposomes or via fortifying with IL-18 cytokines [6088]. The major disadvantage of these vaccines is their relatively low protection and the requirement for several booster doses or prime boosting. Further studies are required to assess the efficacy of these candidate antigens in livestock [110].

Synthetic peptide vaccine

In 1994, Tabatabai and Pugh [93] synthesized three peptides derived from the primary structure of B. abortus Cu-Zn SOD and used those peptides in vaccines against brucellosis. Their studies showed that only peptide 3 (GGAPGEKDGKIVPAG) possessed protective biological activity, which was demonstrated by its ability to modulate both splenomegaly and the extent of Brucella infection in spleen [91]. It was concluded that peptide 3 probably contains a specific sequence preferentially recognized by the cellular immune system. Hence, this study showed that a highly selective epitope capable of activating T cells can be selected and synthesized accordingly. However, in general, this class of vaccine displays relatively low protection, which would hinder its effective deployment.

Vector-delivered Brucella vaccines

Due to its overall similarity in infection, an intracellular pathogen such as an attenuated Salmonella strain can be employed as a vector to deliver Brucella antigen at immunologically critical sites. It has been reported that oral immunization of mice with Salmonella expressing a 31 kDa Brucella protein resulted in the production of local as well as serum antibodies against the protein but resulted in poor CMI responses [89]. Recently, our group developed Salmonella Typhimurium delivery-based Brucella vaccines. Using a mouse model, the suitability of the vectored Brucella vaccine in different routes of immunization was studied [52]. To improve the protective efficacy, we also investigated the usefulness of these vectored vaccines when provided as a “cocktail”. The cocktail of Salmonella delivering heterologous antigens of Brucella included SOD, BLS, PrpA, and Omp19 proteins. To further improve the vaccine's efficacy, purified LPS was included in the cocktail [55]. Single dose immunization results were at par with the reference RB51 vaccination. Food-grade genetically modified Lactococcus lactis strains that express B. abortus GroEL heat-shock protein have been developed [66]; however, protective efficacies against wild-type challenges were not evaluated. It was reported that several recombinant vaccinia viruses were able to express a variety of Brucella antigens (HtrA, GroEL, GroES, Cu-ZnSOD, and YajC), and that study revealed that the recombinant vaccinia viruses induced specific immune responses to these antigens in mice, but the levels of protection were not significant [98].

Vaccine Trials for Brucella

A general overview and summary of the different types of trial hosts are presented in Table 2.

Laboratory animals

Due to economical and ethical concerns associated with experimentation in hosts such as ruminants, humans, and other primates, various small laboratory animal species serve as important tools for investigating the pathophysiology and undertaking vaccine-related trials of brucellosis [46]. Many foremost experiments related to Brucella involve the use of chicken embryo as a model [15]. Rabbit has mostly served as an animal for producing antibodies against Brucella antigens as well as a model for Brucella toxicity and hypersensitivity, mainly due of the susceptibility of rabbit to bacterial endotoxins and toxins [435]. However, mouse (Mus musculus) has been the most convenient and commonly used brucellosis model [4687]. For World Organisation for Animal Health (OIE) vaccine batch potency testing and residual virulence evaluation, mice are routinely used. CD1 mice at 5 to 6 weeks of age are used for screening of candidate vaccines. Due to ease of handling, availability, and presence of large amounts of information and literature, mice are often the laboratory animal of choice [46]. The efficacy of a Brucella candidate vaccine is usually measured as a reduction in splenic colonization as compared to that in non-vaccinated but challenged controls. However, due to matching phases and pathophysiology, a guinea pig model serves as a better model than mouse for human-related anti-Brucella vaccine studies [87]. Guinea pigs exhibit similar pulmonary, hepatic, spleen, and genital lesions and have similar hypersensitivity reactions to those observed in humans; moreover, they exhibit comparative stages of infection caused by Brucella in natural hosts, including abortion [121341]. In addition, guinea pigs exhibit the highest susceptibility to Brucella infections among the tested laboratory animals [41]. Hence, guinea pig represents one of the best models for several immunological and vaccine studies [465074].

Livestock

Large animal experimentation and trials involving anti-Brucella vaccine are comparatively uncommon due to limited availability of resources. However, it is critical to characterize the equivalent immune responses in livestock hosts. Such information will provide a better understanding of the safety and efficacy of the vaccine. Brucellosis in cattle is usually caused by biovars of B. abortus. In some countries, particularly in southern Europe and western Asia, where cattle are kept in close association with sheep or goats, infection can also be caused by B. melitensis. In small ruminants, brucellosis is mainly due to B. melitensis [42]. Such brucellosis is mainly controlled by vaccination with the vaccine strain Rev. 1. This strain confers excellent immunity; however, it has two disadvantages: i) prolonged sensitization of animals resulting in interference with subsequent allergic tests; ii) formation of anti-Rev. 1 antibodies, which disappear at different rates among individual animals [37]. Occasionally, B. suis may produce a chronic infection in the mammary gland of cattle, but it has not been reported to cause abortion or spread to other animals [50]. Considering the disease's transmission cycle and zoonotic implications, livestock animals may be more suitably vaccinated with multivalent pan-Brucella species cross-protecting vaccines.

Companion animals

Brucella canis is important pathogen resulting in brucellosis in dogs, and, to date, no effective anti-B. canis vaccine is available. Very limited research has been initiated into this type of anti-Brucella vaccine [252682]. The site of injection and the type of adjuvant to be used needs to be determined before initiating vaccine trials [63]. Vaccines containing recombinant antigens may be less reactogenic and also less immunogenic, thus necessitating the inclusion of an adjuvant [26].

Other animals

Brucellosis has been reported in the one-humped camel (Camelus dromedarius), in the two-humped camel (Camelus bactrianus), and in South American camelids including llama (Lama glama), alpaca (Lama pacos), guanaco (Lama guinicoe), and vicuna (Vicugne vicugne). Those infections have been related to contact with large and small ruminants infected with B. abortus or B. melitensis [50106]. In addition, brucellosis has been observed in the domestic buffalo (Bubalus bubalus), American and European bison (Bison bison, Bison bonasus), yak (Bos grunniens), elk/wapiti (Cervus elaphus), the African buffalo (Syncerus caffer), and various African antelope species. The clinical manifestations of brucellosis in these animals are similar to those in cattle [50]. However, it is uncertain whether current vaccine candidates and immunization strategies would be suitable application in feral populations of those species. Rough strain RB51 has been tested for use in wild animals, such as bison in North America, and it conferred considerable protection in test subjects; moreover, the level of protection was improved with a booster vaccine regime [7677].

Human

To date, no vaccine licensed for human anti-Brucella is available. In addition, there is very little information and relevant data regarding human clinical vaccine trials. Although the threat due to bioterrorism has recently toned down [47], developing a safe and effective vaccine is of prime importance due to the chronic debilitating nature of brucellosis. The most extensive trial and study was held in the former Soviet Union in which over 3 million people were vaccinated with the S19 vaccine strain [105]. Most human brucellosis cases have been linked with infected sheep and goats [28]. There was a nearly 60% reduction in the number of human cases over the period 1952 to 1958. Despite conferring strong protection due to the prolonged persistency in vaccinated individuals [105], the deployment of S19 in humans has been limited.
Several candidate anti-Brucella vaccines have been proposed for the use against human brucellosis. Since B. melitensis is the most common species found in humans, development of a vaccine based on B. melitensis is prudent. Several candidate vaccines are reported to be capable of protecting against a virulent B. melitensis challenge in mice model [7895]. However, validation in a primate model may be necessary prior to undertaking large scale trials. Success stories involving the control of human brucellosis via its control in an animal population have been documented in different parts of the world [6578]. Nevertheless, with the increasing reports and re-emergence of brucellosis around the globe [9296100], research into developing safe, effective, cross-protecting, human exclusive vaccines must be continued.

Conclusions

The most effective strategy to control human brucellosis in the absence of an appropriate human vaccine is to control brucellosis in animal populations; thereby reducing zoonotic transmission and the number of carrier hosts. For human anti-Brucella vaccine, safety properties should be considered of prime importance, and, ideally, the vaccine should be cross protecting. Vaccine formulation comprising a cocktail of protective antigens may be used in conjunction with an appropriate adjuvant to augment the vaccine's protective capabilities. Newer immuno-modulators such as viability-associated pathogen-associated molecular patterns may also be included in such formulations in order to mimic live bacterial infections. A major challenge in the development of an ideal vaccine lies in evoking robust CMI in the host. In general, vaccine candidates that evoke a strong CMI response confer a better level of protection. Hence, targeting the CMI branch of host immunity via induction of IL-12 and INF-γ should prove to be useful.

Figures and Tables

Table 1

Summary of Brucella vaccine and their properties

jvs-18-281-i001
Table 2

Summary of hosts in vaccine development and trials

jvs-18-281-i002

Acknowledgments

This research was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Korea (grant No. HI16C2130).

Notes

Conflict of Interest The authors declare no conflicts of interest.

References

1. Alcantara RB, Read RD, Valderas MW, Brown TD, Roop RM 2nd. Intact purine biosynthesis pathways are required for wild-type virulence of Brucella abortus 2308 in the BALB/c mouse model. Infect Immun. 2004; 72:4911–4917.
crossref
2. Al-Mariri A, Tibor A, Mertens P, De Bolle X, Michel P, Godefroid J, Walravens K, Letesson JJ. Protection of BALB/c mice against Brucella abortus 544 challenge by vaccination with bacterioferritin or P39 recombinant proteins with CpG oligodeoxynucleotides as adjuvant. Infect Immun. 2001; 69:4816–4822.
crossref
3. Almirón M, Martínez M, Sanjuan N, Ugalde RA. Ferrochelatase is present in Brucella abortus and is critical for its intracellular survival and virulence. Infect Immun. 2001; 69:6225–6230.
crossref
4. Alton GG, Jones LM, Angus RD, Verger JM. Techniques for the brucellosis laboratory. Paris: Institut National de la Recherche Agronomique;1988.
5. Anderson TD, Cheville NF. Ultrastructural morphometric analysis of Brucella abortus-infected trophoblasts in experimental placentitis. Bacterial replication occurs in rough endoplasmic reticulum. Am J Pathol. 1986; 124:226–237.
6. APPMG (All-Party Parliamentary Group on Malaria and Neglected Tropical Diseases). The Neglected Tropical Diseases: A Challenge We Could Rise to -- Will We? Geneva: World Health Organization;2009.
7. Arenas-Gamboa AM, Ficht TA, Kahl-McDonagh MM, Rice-Ficht AC. Immunization with a single dose of a microencapsulated Brucella melitensis mutant enhances protection against wild-type challenge. Infect Immun. 2008; 76:2448–2455.
crossref
8. Arenas-Gamboa AM, Rice-Ficht AC, Kahl-McDonagh MM, Ficht TA. Protective efficacy and safety of Brucella melitensis 16MΔmucR against intraperitoneal and aerosol challenge in BALB/c mice. Infect Immun. 2011; 79:3653–3658.
crossref
9. Barquero-Calvo E, Chaves-Olarte E, Weiss DS, Guzmán-Verri C, Chacón-Díaz C, Rucavado A, Moriyón I, Moreno E. Brucella abortus uses a stealthy strategy to avoid activation of the innate immune system during the onset of infection. PLoS One. 2007; 2:e631.
10. Blasco JM, Gamazo C, Winter AJ, Jiménez de Bagüés MP, Marín C, Barberán M, Moriyón I, Alonso-Urmeneta B, Díaz R. Evaluation of whole cell and subcellular vaccines against Brucella ovis in rams. Vet Immunol Immunopathol. 1993; 37:257–270.
crossref
11. Brandão AP, Oliveira FS, Carvalho NB, Vieira LQ, Azevedo V, Macedo GC, Oliveira SC. Host susceptibility to Brucella abortus infection is more pronounced in IFN-γ knockout than IL-12/β2-microglobulin double-deficient mice. Clin Dev Immunol. 2012; 2012:589494.
12. Braude AI. Studies in the pathology and pathogenesis of experimental brucellosis. II. The formation of the hepatic granuloma and its evolution. J Infect Dis. 1951; 89:87–94.
crossref
13. Braude AI, Spink WW. Studies in the pathology and pathogenesis of experimental brucellosis. III. Investigations pertaining to the function of the spleen. J Infect Dis. 1951; 89:272–276.
crossref
14. Bruce D. Note on the discovery of a microorganism in Malta fever. Practitioner. 1887; 39:161–170.
15. Buddingh GJ, Womack FC Jr. Observations on the infection of chick embryos with Bacterium tularense, Brucella, and Pasteurella pestis. J Exp Med. 1941; 74:213–222.
crossref
16. Bundle DR, Cherwonogrodzky JW, Gidney MA, Meikle PJ, Perry MB, Peters T. Definition of Brucella A and M epitopes by monoclonal typing reagents and synthetic oligosaccharides. Infect Immun. 1989; 57:2829–2836.
crossref
17. Canning PC, Roth JA, Deyoe BL. Release of 5'-guanosine monophosphate and adenine by Brucella abortus and their role in the intracellular survival of the bacteria. J Infect Dis. 1986; 154:464–470.
crossref
18. Cardoso PG, Macedo GC, Azevedo V, Oliveira SC. Brucella spp noncanonical LPS: structure, biosynthesis, and interaction with host immune system. Microb Cell Fact. 2006; 5:13.
19. Cassataro J, Velikovsky CA, de la Barrera S, Estein SM, Bruno L, Bowden R, Pasquevich KA, Fossati CA, Giambartolomei GH. A DNA vaccine coding for the Brucella outer membrane protein 31 confers protection against B. melitensis and B. ovis infection by eliciting a specific cytotoxic response. Infect Immun. 2005; 73:6537–6546.
crossref
20. Celli J, de Chastellier C, Franchini DM, Pizarro-Cerda J, Moreno E, Gorvel JP. Brucella evades macrophage killing via VirB-dependent sustained interactions with the endoplasmic reticulum. J Exp Med. 2003; 198:545–556.
crossref
21. Cespedes S, Andrews E, Folch H, Oñate A. Identification and partial characterisation of a new protective antigen of Brucella abortus. J Med Microbiol. 2000; 49:165–170.
crossref
22. Chain PS, Comerci DJ, Tolmasky ME, Larimer FW, Malfatti SA, Vergez LM, Aguero F, Land ML, Ugalde RA, Garcia E. Whole-genome analyses of speciation events in pathogenic Brucellae. Infect Immun. 2005; 73:8353–8361.
crossref
23. Chaudhuri P, Singha H, Goswami TK, Jana C, Shukla G. DNA prime and protein boost immunization with combined SOD-L7/L12 antigen confers protection to mice against Brucella abortus 544 challenge. Adv Anim Vet Sci. 2013; 1:143–147.
24. Cheville NF, Olsen SC, Jensen AE, Stevens MG, Palmer MV, Florance AM. Effects of age at vaccination on efficacy of Brucella abortus strain RB51 to protect cattle against brucellosis. Am J Vet Res. 1996; 57:1153–1156.
25. Clausse M, Díaz AG, Ghersi G, Zylberman V, Cassataro J, Giambartolomei GH, Goldbaum FA, Estein SM. The vaccine candidate BLSOmp31 protects mice against Brucella canis infection. Vaccine. 2013; 31:6129–6135.
crossref
26. Clausse M, Díaz AG, Ibañez AE, Cassataro J, Giambartolomei GH, Estein SM. Evaluation of the efficacy of outer membrane protein 31 vaccine formulations for protection against Brucella canis in BALB/c mice. Clin Vaccine Immunol. 2014; 21:1689–1694.
crossref
27. Commander NJ, Spencer SA, Wren BW, MacMillan AP. The identification of two protective DNA vaccines from a panel of five plasmid constructs encoding Brucella melitensis 16M genes. Vaccine. 2007; 25:43–54.
crossref
28. Corbel MJ. Brucellosis: an overview. Emerg Infect Dis. 1997; 3:213–221.
crossref
29. Corbel MJ. Brucellosis in humans and animals. Geneva: World Health Organization;2006.
30. Crasta OR, Folkerts O, Fei Z, Mane SP, Evans C, Martino-Catt S, Bricker B, Yu G, Du L, Sobral BW. Genome sequence of Brucella abortus vaccine strain S19 compared to virulent strains yields candidate virulence genes. PLoS One. 2008; 3:e2193.
31. de Figueiredo P, Ficht TA, Rice-Ficht A, Rossetti CA, Adams LG. Pathogenesis and immunobiology of brucellosis: review of Brucella-host interactions. Am J Pathol. 2015; 185:1505–1517.
32. De Ley J, Mannheim W, Segers P, Lievens A, Denijn M, Vanhoucke M, Gillis M. Ribosomal ribonucleic acid cistron similarities and taxonomic neighborhood of Brucella and CDC group Vd. Int J Syst Bacteriol. 1987; 37:35–42.
crossref
33. Delpino MV, Estein SM, Fossati CA, Baldi PC, Cassataro J. Vaccination with Brucella recombinant DnaK and SurA proteins induces protection against Brucella abortus infection in BALB/c mice. Vaccine. 2007; 25:6721–6729.
crossref
34. den Hartigh AB, Sun YH, Sondervan D, Heuvelmans N, Reinders MO, Ficht TA, Tsolis RM. Differential requirements for VirB1 and VirB2 during Brucella abortus infection. Infect Immun. 2004; 72:5143–5149.
crossref
35. Diaz R, Oyeledun MA. Studies of some biological activities of “brucella” endotoxin in normal and infected animals and the role of the hypersensitivity factor. Ann Sclavo. 1977; 19:117–130.
36. Dzata GK, Confer AW, Wyckoff JH 3rd. The effects of adjuvants on immune responses in cattle injected with a Brucella abortus soluble antigen. Vet Microbiol. 1991; 29:27–48.
crossref
37. Fensterbank R. Brucellosis in cattle, sheep and goats: diagnosis, control and vaccination. Rev Sci Tech Off Int Epiz. 1986; 5:605–618.
38. Ferguson GP, Datta A, Baumgartner J, Roop RM 2nd, Carlson RW, Walker GC. Similarity to peroxisomal-membrane protein family reveals that Sinorhizobium and Brucella BacA affect lipid-A fatty acids. Proc Natl Acad Sci U S A. 2004; 101:5012–5017.
crossref
39. Finlay BB, Falkow S. Common themes in microbial pathogenicity. Microbiol Rev. 1989; 53:210–230.
crossref
40. Fridovich I. Superoxide dismutases. An adaptation to a paramagnetic gas. J Biol Chem. 1989; 264:7761–7764.
41. García-Carrillo C. Laboratory animal models for brucellosis studies. In : Nielsen K, Duncan , editors. Animal Brucellosis. Boca Raton: CRC Press;1990. p. 423–442.
42. Garin-Bastuji B, Blasco JM, Grayon M, Verger JM. Brucella melitensis infection in sheep: present and future. Vet Res. 1998; 29:255–274.
43. Goenka R, Guirnalda PD, Black SJ, Baldwin CL. B lymphocytes provide an infection niche for intracellular bacterium Brucella abortus. J Infect Dis. 2012; 206:91–98.
crossref
44. Goenka R, Parent MA, Elzer PH, Baldwin CL. B cell-deficient mice display markedly enhanced resistance to the intracellular bacterium Brucella abortus. J Infect Dis. 2011; 203:1136–1146.
crossref
45. González D, Grilló MJ, De Miguel MJ, Ali T, Arce-Gorvel V, Delrue RM, Conde-Alvarez R, Muñoz P, López-Goñi I, Iriarte M, Marín CM, Weintraub A, Widmalm G, Zygmunt M, Letesson JJ, Gorvel JP, Blasco JM, Moriyón I. Brucellosis vaccines: assessment of Brucella melitensis lipopolysaccharide rough mutants defective in core and O-polysaccharide synthesis and export. PLoS One. 2008; 3:e2760.
46. Grilló MJ, Blasco JM, Gorvel JP, Moriyón I, Moreno E. What have we learned from brucellosis in the mouse model? Vet Res. 2012; 43:29.
crossref
47. Guihot A, Bossi P, Bricaire F. [Bioterrorism with brucellosis]. Presse Med. 2004; 33:119–122. French.
48. Guo F, Zhang H, Chen C, Hu S, Wang Y, Qiao J, Ren Y, Zhang K, Wang Y, Du G. Autophagy favors Brucella melitensis survival in infected macrophages. Cell Mol Biol Lett. 2012; 17:249–257.
49. Halling SM, Peterson-Burch BD, Bricker BJ, Zuerner RL, Qing Z, Li LL, Kapur V, Alt DP, Olsen SC. Completion of the genome sequence of Brucella abortus and comparison to the highly similar genomes of Brucella melitensis and Brucella suis. J Bacteriol. 2005; 187:2715–2726.
crossref
50. International Office of Epizootics. Bovine brucellosis. International Office of Epizootics. Manual of Diagnostic Tests and Vaccines for Terrestrial Animals: Mammals, Birds and Bees. 7th ed. Paris: World Organisation for Animal Health (OIE);2009. p. 1–35.
51. Kaushik P, Singh DK, Kumar SV, Tiwari AK, Shukla G, Dayal S, Chaudhuri P. Protection of mice against Brucella abortus 544 challenge by vaccination with recombinant OMP28 adjuvanted with CpG oligonucleotides. Vet Res Commun. 2010; 34:119–132.
crossref
52. Kim WK, Moon JY, Kim S, Hur J. Comparison between immunization routes of live attenuated Salmonella Typhimurium strains expressing BCSP31, Omp3b, and SOD of Brucella abortus in murine model. Front Microbiol. 2016; 7:550.
53. Kurar E, Splitter GA. Nucleic acid vaccination of Brucella abortus ribosomal L7/L12 gene elicits immune response. Vaccine. 1997; 15:1851–1857.
crossref
54. Lalsiamthara J, Gogia N, Goswami TK, Singh RK, Chaudhuri P. Intermediate rough Brucella abortus S19Δper mutant is DIVA enable, safe to pregnant guinea pigs and confers protection to mice. Vaccine. 2015; 33:2577–2583.
crossref
55. Lalsiamthara J, Lee JH. Brucella lipopolysaccharide reinforced Salmonella delivering Brucella immunogens protects mice against virulent challenge. Vet Microbiol. 2017; 205:84–91.
crossref
56. Lapaque N, Moriyon I, Moreno E, Gorvel JP. Brucella lipopolysaccharide acts as a virulence factor. Curr Opin Microbiol. 2005; 8:60–66.
57. LeVier K, Phillips RW, Grippe VK, Roop RM 2nd, Walker GC. Similar requirements of a plant symbiont and a mammalian pathogen for prolonged intracellular survival. Science. 2000; 287:2492–2493.
crossref
58. Liautard JP, Gross A, Dornand J, Köhler S. Interactions between professional phagocytes and Brucella spp. Microbiologia. 1996; 12:197–206.
59. Lord VR, Schurig GG, Cherwonogrodzky JW, Marcano MJ, Melendez GE. Field study of vaccination of cattle with Brucella abortus strains RB51 and 19 under high and low disease prevalence. Am J Vet Res. 1998; 59:1016–1020.
60. Mallick AI, Singha H, Chaudhuri P, Nadeem A, Khan SA, Dar KA, Owais M. Liposomised recombinant ribosomal L7/L12 protein protects BALB/c mice against Brucella abortus 544 infection. Vaccine. 2007; 25:3692–3704.
crossref
61. Martirosyan A, Moreno E, Gorvel JP. An evolutionary strategy for a stealthy intracellular Brucella pathogen. Immunol Rev. 2011; 240:211–234.
crossref
62. McEwen AD. Experiments on contagious abortion. The immunity of cattle inoculated with vaccines of grades virulence. Vet Rec. 1940; 52:815–819.
63. Meeusen EN, Walker J, Peters A, Pastoret PP, Jungersen G. Current status of veterinary vaccines. Clin Microbiol Rev. 2007; 20:489–510.
crossref
64. Meyer ME, Gibbons RW. Results of trial use of H-38 vaccine for immunizing beef heifers against experimental exposure to Brucella abortus, strain 2308. Proc Annu Meet U S Anim Health Assoc. 1978; (82):106–119.
65. Minas A, Minas M, Stournara A, Tselepidis S. The “effects” of Rev-1 vaccination of sheep and goats on human brucellosis in Greece. Prev Vet Med. 2004; 64:41–47.
crossref
66. Miyoshi A, Bermúdez-Humarán LG, Ribeiro LA, Le Loir Y, Oliveira SC, Langella P, Azevedo V. Heterologous expression of Brucella abortus GroEL heat-shock protein in Lactococcus lactis. Microb Cell Fact. 2006; 5:14.
67. Monreal D, Grilló MJ, González D, Marín CM, De Miguel MJ, López-Goñi I, Blasco JM, Cloeckaert A, Moriyón I. Characterization of Brucella abortus O-polysaccharide and core lipopolysaccharide mutants and demonstration that a complete core is required for rough vaccines to be efficient against Brucella abortus and Brucella ovis in the mouse model. Infect Immun. 2003; 71:3261–3271.
crossref
68. Montaraz JA, Winter AJ. Comparison of living and nonliving vaccines for Brucella abortus in BALB/c mice. Infect Immun. 1986; 53:245–251.
crossref
69. Moreno E, Stackebrandt E, Dorsch M, Wolters J, Busch M, Mayer H. Brucella abortus 16S rRNA and lipid A reveal a phylogenetic relationship with members of the alpha-2 subdivision of the class Proteobacteria. J Bacteriol. 1990; 172:3569–3576.
crossref
70. Moriyón I, Grilló MJ, Monreal D, González D, Marín C, López-Goñi I, Mainar-Jaime RC, Moreno E, Blasco JM. Rough vaccines in animal brucellosis: structural and genetic basis and present status. Vet Res. 2004; 35:1–38.
crossref
71. Muñoz-Montesino C, Andrews E, Rivers R, González-Smith A, Moraga-Cid G, Folch H, Céspedes S, Oñate AA. Intraspleen delivery of a DNA vaccine coding for superoxide dismutase (SOD) of Brucella abortus induces SOD-specific CD4+ and CD8+ T cells. Infect Immun. 2004; 72:2081–2087.
crossref
72. Nymo IH, Tryland M, Godfroid J. A review of Brucella infection in marine mammals, with special emphasis on Brucella pinnipedialis in the hooded seal (Cystophora cristata). Vet Res. 2011; 42:93.
crossref
73. O'Hara MJ, Collins DM, de Lisle GW. Restriction endonuclease analysis of Brucella ovis and other Brucella species. Vet Microbiol. 1985; 10:425–429.
74. Oliveira SC, Harms JS, Banai M, Splitter GA. Recombinant Brucella abortus proteins that induce proliferation and gamma-interferon secretion by CD4+ T cells from Brucella-vaccinated mice and delayed-type hypersensitivity in sensitized guinea pigs. Cell Immunol. 1996; 172:262–268.
crossref
75. Oliveira SC, Splitter GA. Immunization of mice with recombinant L7/L12 ribosomal protein confers protection against Brucella abortus infection. Vaccine. 1996; 14:959–962.
crossref
76. Olsen SC, Boyle SM, Schurig GG, Sriranganathan NN. Immune responses and protection against experimental challenge after vaccination of bison with Brucella abortus strain RB51 or RB51 overexpressing superoxide dismutase and glycosyltransferase genes. Clin Vaccine Immunol. 2009; 16:535–540.
crossref
77. Olsen SC, Johnson C. Immune responses and safety after dart or booster vaccination of bison with Brucella abortus strain RB51. Clin Vaccine Immunol. 2012; 19:642–648.
crossref
78. Pappas G, Papadimitriou P, Akritidis N, Christou L, Tsianos EV. The new global map of human brucellosis. Lancet Infect Dis. 2006; 6:91–99.
crossref
79. Pasquevich KA, Estein SM, García Samartino C, Zwerdling A, Coria LM, Barrionuevo P, Fossati CA, Giambartolomei GH, Cassataro J. Immunization with recombinant Brucella species outer membrane protein Omp16 or Omp19 in adjuvant induces specific CD4+ and CD8+ T cells as well as systemic and oral protection against Brucella abortus infection. Infect Immun. 2009; 77:436–445.
crossref
80. Pizarro-Cerdá J, Méresse S, Parton RG, van der Goot G, Sola-Landa A, Lopez-Goñi I, Moreno E, Gorvel JP. Brucella abortus transits through the autophagic pathway and replicates in the endoplasmic reticulum of nonprofessional phagocytes. Infect Immun. 1998; 66:5711–5724.
crossref
81. Porte F, Naroeni A, Ouahrani-Bettache S, Liautard JP. Role of the Brucella suis lipopolysaccharide O antigen in phagosomal genesis and in inhibition of phagosome-lysosome fusion in murine macrophages. Infect Immun. 2003; 71:1481–1490.
crossref
82. Qian J, Bu Z, Lang X, Yan G, Yang Y, Wang X, Wang X. A safe and molecular-tagged Brucella canis ghosts confers protection against virulent challenge in mice. Vet Microbiol. 2017; 204:121–128.
crossref
83. Scholz HC, Pfeffer M, Witte A, Neubauer H, Al Dahouk S, Wernery U, Tomaso H. Specific detection and differentiation of Ochrobactrum anthropi, Ochrobactrum intermedium and Brucella spp. by a multi-primer PCR that targets the recA gene. J Med Microbiol. 2008; 57:64–71.
crossref
84. Schurig GG, Roop RM 2nd, Bagchi T, Boyle S, Buhrman D, Sriranganathan N. Biological properties of RB51; a stable rough strain of Brucella abortus. Vet Microbiol. 1991; 28:171–188.
crossref
85. Schurig GG, Sriranganathan N, Corbel MJ. Brucellosis vaccines: past, present and future. Vet Microbiol. 2002; 90:479–496.
crossref
86. Sieira R, Comerci DJ, Sánchez DO, Ugalde RA. A homologue of an operon required for DNA transfer in Agrobacterium is required in Brucella abortus for virulence and intracellular multiplication. J Bacteriol. 2000; 182:4849–4855.
crossref
87. Silva TM, Costa EA, Paixão TA, Tsolis RM, Santos RL. Laboratory animal models for brucellosis research. J Biomed Biotechnol. 2011; 2011:518323.
crossref
88. Singha H, Mallick AI, Jana C, Fatima N, Owais M, Chaudhuri P. Co-immunization with interlukin-18 enhances the protective efficacy of liposomes encapsulated recombinant Cu-Zn superoxide dismutase protein against Brucella abortus. Vaccine. 2011; 29:4720–4727.
crossref
89. Stabel TJ, Mayfield JE, Morfitt DC, Wannemuehler MJ. Oral immunization of mice and swine with an attenuated Salmonella choleraesuis [delta cya-12 delta(crp-cdt)19] mutant containing a recombinant plasmid. Infect Immun. 1993; 61:610–618.
crossref
90. Starr T, Child R, Wehrly TD, Hansen B, Hwang S, López-Otin C, Virgin HW, Celli J. Selective subversion of autophagy complexes facilitates completion of the Brucella intracellular cycle. Cell Host Microbe. 2012; 11:33–45.
crossref
91. Stevens MG, Tabatabai LB, Olsen SC, Cheville NF. Immune responses to superoxide dismutase and synthetic peptides of superoxide dismutase in cattle vaccinated with Brucella abortus strain 19 or RB51. Vet Microbiol. 1994; 41:383–389.
crossref
92. Suárez-Esquivel M, Ruiz-Villalobos N, Jiménez-Rojas C, Barquero-Calvo E, Chacón-Díaz C, Víquez-Ruiz E, Rojas-Campos N, Baker KS, Oviedo-Sánchez G, Amuy E, Chaves-Olarte E, Thomson NR, Moreno E, Guzmán-Verri C. Brucella neotomae infection in humans, Costa Rica. Emerg Infect Dis. 2017; 23:997–1000.
93. Tabatabai LB, Pugh GW Jr. Modulation of immune responses in Balb/c mice vaccinated with Brucella abortus Cu-Zn superoxide dismutase synthetic peptide vaccine. Vaccine. 1994; 12:919–924.
crossref
94. Tabatabai LB, Pugh GW Jr, Stevens MG, Phillips M, McDonald TJ. Monophosphoryl lipid A-induced immune enhancement of Brucella abortus salt-extractable protein and lipopolysaccharide vaccines in BALB/c mice. Am J Vet Res. 1992; 53:1900–1907.
95. Tabynov K, Yespembetov B, Matikhan N, Ryskeldinova S, Zinina N, Kydyrbayev Z, Assanzhanova N, Tabynov K, Renukaradhya GJ, Mukhitdinova G, Sansyzbay A. First evaluation of an influenza viral vector based Brucella abortus vaccine in sheep and goats: assessment of safety, immunogenicity and protective efficacy against Brucella melitensis infection. Vet Microbiol. 2016; 197:15–20.
crossref
96. Tasiame W, Emikpe BO, Folitse RD, Fofie CO, Burimuah V, Johnson S, Awuni JA, Afari E, Yebuah N, Wurapa F. The prevalence of brucellosis in cattle and their handlers in North Tongu District of Volta Region, Ghana. Afr J Infect Dis. 2016; 10:111–117.
crossref
97. Taylor AW, McDiarmid A. The stability of the avirulent characters of Brucella abortus strain 19 and strain 45/20 in lactating and pregnant cows. Vet Rec. 1949; 61:317–318.
98. Toth TE, Cobb JA, Boyle SM, Roop RM, Schurig GG. Selective humoral immune response of Balb/C mice to Brucella abortus proteins expressed by vaccinia virus recombinants. Vet Microbiol. 1995; 45:171–183.
crossref
99. Trant CG, Lacerda TL, Carvalho NB, Azevedo V, Rosinha GM, Salcedo SP, Gorvel JP, Oliveira SC. The Brucella abortus phosphoglycerate kinase mutant is highly attenuated and induces protection superior to that of vaccine strain 19 in immunocompromised and immunocompetent mice. Infect Immun. 2010; 78:2283–2291.
crossref
100. Tukana A, Gummow B. Dairy farm demographics and management factors that played a role in the re-emergence of brucellosis on dairy cattle farms in Fiji. Trop Anim Health Prod. 2017; 49:1171–1178.
crossref
101. Ugalde JE, Comerci DJ, Leguizamón MS, Ugalde RA. Evaluation of Brucella abortus phosphoglucomutase (pgm) mutant as a new live rough-phenotype vaccine. Infect Immun. 2003; 71:6264–6269.
crossref
102. Ugalde JE, Czibener C, Feldman MF, Ugalde RA. Identification and characterization of the Brucella abortus phosphoglucomutase gene: role of lipopolysaccharide in virulence and intracellular multiplication. Infect Immun. 2000; 68:5716–5723.
crossref
103. Van Campen H, Rhyan J. The role of wildlife in diseases of cattle. Vet Clin North Am Food Anim Pract. 2010; 26:147–161.
crossref
104. Velikovsky CA, Cassataro J, Giambartolomei GH, Goldbaum FA, Estein S, Bowden RA, Bruno L, Fossati CA, Spitz M. A DNA vaccine encoding lumazine synthase from Brucella abortus induces protective immunity in BALB/c mice. Infect Immun. 2002; 70:2507–2511.
crossref
105. Vershilova PA. The use of live vaccine for vaccination of human beings against brucellosis in the USSR. Bull World Health Organ. 1961; 24:85–89.
106. Wernery U. Camelid brucellosis: a review. Rev Sci Tech. 2014; 33:839–857.
crossref
107. Winter AJ, Rowe GE, Duncan JR, Eis MJ, Widom J, Ganem B, Morein B. Effectiveness of natural and synthetic complexes of porin and O polysaccharide as vaccines against Brucella abortus in mice. Infect Immun. 1988; 56:2808–2817.
crossref
108. Winter AJ, Schurig GG, Boyle SM, Sriranganathan N, Bevins JS, Enright FM, Elzer PH, Kopec JD. Protection of BALB/c mice against homologous and heterologous species of Brucella by rough strain vaccines derived from Brucella melitensis and Brucella suis biovar 4. Am J Vet Res. 1996; 57:677–683.
109. Yang X, Hudson M, Walters N, Bargatze RF, Pascual DW. Selection of protective epitopes for Brucella melitensis by DNA vaccination. Infect Immun. 2005; 73:7297–7303.
crossref
110. Yang X, Skyberg JA, Cao L, Clapp B, Thornburg T, Pascual DW. Progress in Brucella vaccine development. Front Biol (Beijing). 2013; 8:60–77.
111. Yang Y, Yin J, Guo D, Lang X, Wang X. Immunization of mice with recombinant S-adenosyl-L-homocysteine hydrolase protein confers protection against Brucella melitensis infection. FEMS Immunol Med Microbiol. 2011; 61:159–167.
crossref
TOOLS
Similar articles