Abstract
Brucellosis remains burdensome for livestock and humans worldwide. Better vaccines for protection are needed to reduce disease incidence. Immunity to brucellosis and barriers to protection are discussed. The benefits and limitations of conventional and experimental brucellosis vaccines are outlined, and novel vaccination strategies needed to ultimately protect against brucellosis are introduced.
Keywords: vaccine, mucosal immunity, bacteria
1. Brucellosis Overview
The most common zoonotic disease worldwide, brucellosis, is ranked third of eight most neglected zoonotic diseases [1]. This global disease remains problematic for countries on the Mediterranean rim, Middle East, Central Asia, South America, and the states in the USA that border Mexico [2-4]. Estimates claim that for some countries, brucellosis exceeds 500 cases/1×106 inhabitants [2, 5]. Brucellosis is often not recognized or is misdiagnosed resulting in underreporting by as much 26-fold [5, 6]. This unmitigated persistence of brucellosis results from chronic exposure to Brucella-infected livestock [7], often following consumption of unpasteurized milk or other dairy products [4, 8].
Brucellosis is caused by the Gram-negative cocciobacilli from the highly homogenous genus, Brucella [9], which contains 10 - 12 species [10,11], of which B. melitensis, B. abortus, and B. suis are the ones causing human disease [12-14]. In 1886, the British physician, David Bruce, first identified small Gram-negative cocciobacilli obtained from patients hospitalized with “Malta Fever” [15], and called these bacteria, Micrococcus melitensis [16]. A unique attribute of Brucella is their 94% DNA homology among species [6, 14, 17] allowing immunity to cross-protect across heterologous Brucella species. Most Brucella species have two circular chromosomes of varying size [18]. Each species is discriminated by its surface lipopolysaccharide imparting serological specificity [8]. Notably, Brucella LPS is weakly endotoxic with poor toll-like receptor 4 (TLR4) agonist activity due to the long fatty acid acylation of its lipid A. This feature adds to the stealth qualities of Brucella [8].
The primary route of infection for humans is by an oral exposure [4, 19], yet an airborne infection can occur resulting from exposure to Brucella-infected livestock [9]. Brucellosis can be lethal in livestock by inducing abortion [8, 19-21]; however, brucellosis is rarely fatal (<0.5% of cases) in humans, but instead results in a debilitating, systemic disease [3-5, 7], attributed to mostly to B. melitensis or B. abortus [22, 23]. Acute disease in humans often presents as a febrile disease with generic flu-like symptoms such as chills, malaise, headaches, hepatomegaly, and splenomegaly [4,5,7,9]. Despite oral exposures, intestinal disease rarely occurs [24, 25]. Chronic disease symptoms include a relapsing, undulant fever, chronic fatigue, malaise, and can produce positive Brucella blood cultures [7,9,24]. Brucellosis exacerbation can lead to more serious endocarditis and arthritis [26-29]. Brucella infections can be resolved with antibiotic treatment, but this requires a prolonged two-antibiotic regimen [9, 24]. Despite this rigorous treatment, it is not a guarantee that infection will be resolved as evidenced by sequelae occurring in ∼16% of the infected patients [24], of which, 50% have a persistent bacteremia [24]. What remains enigmatic following oral exposure is the absence of Brucella replication or pathology in the gut [19, 25], questioning the significance of oral infections. Recent data combined with past studies implicate the oro-pharyngeal tissues as the site where infections initiate in humans [30-34].
2. Correlates of Protection to Brucella Infections
2.1. Th1-type immunity required for protection to brucellosis
Brucella exposure by any route of infection will cause a bacteremia and progress to a systemic infection [5,9,24]. Brucella's primary target is macrophages [35], and once in these cells, brucellae become difficult to eliminate from the intracellular compartment. This pathogen has many stealth features enabling such infection including its low endotoxic LPS [8]; its ability to avoid fusion with lysosomes [35]; its ability to dampen dendritic cell maturation [36] and to interfere with TLR2 and TLR4 signaling via TcpB, an analog for mammalian Toll/interleukin 1 receptor (TIR) domain-containing adaptor protein (TIRAP) [37,38]; and to interfere with MHC class I [39] and class II antigen presentation [40,41]. Thus, macrophages need to be activated by IFN-γ [42,43] and TNF-α [23, 44] to effectively eliminate intracellular brucellae. Hence, IFN-γ is required for resolving Brucella infections of macrophages [45, 46] in an IL-12- [47] and TNF-α-dependent manner [48].
Brucella's astute capacity to avoid innate immune detection also impairs subsequent development of a suitable adaptive immune response further accentuating the importance of cellular immunity for protection against brucellosis. Immunity to Brucella requires the stimulation of IFN-γ for protection as evidenced by disease exacerbation and impotency of vaccines in IFN-γ-/- mice [49-51]. The source of IFN- γ can be CD4+ [51-59], CD8+ T cells [50, 58-60], or both [61]. Both T cell subset responses have been studied after Brucella infections in various immunization and challenge paradigms, but their role in Brucella infections remains controversial. The relevance of CD4+ or CD8+ T cell subsets depends on the vaccine and the route of administration. Some studies show that CD4+ T cells as the major source of IFN-γ, and critical in thwarting brucellosis [52-56]. Thought to be less important than CD4+ T cells, CD8+ T cells' role was assumed to be solely cytotoxicity [57]. The notion of CD8+ T cells being dispensable in protection was evident by the exquisite protection against virulent B. melitensis challenge retained in CD8-/- mice orally immunized with a purine auxotrophic B. melitensis mutant still retained [54]. The importance of CD4+ T cells for protection is evident after low-dose exposure of MHC class II-/- mice to virulent B. melitensis and subsequent treatment with antibiotics. At 9 wks post-immunization, MHC class II-/- mice were subjected to virulent challenge. At 50 days post-challenge, the mice had considerably more wild-type brucellae colonization of their spleens with concomitant reduction in IFN- γ -producing cells than did similarly treated wild-type mice [54]. In contrast, MHC class II-/- and TAP1-/- mice nasally infected with virulent B. melitensis, and then treated with antibiotics. Both showed protection equivalent to that of similarly immunized and challenged wild-type mice [61] implicating that as long as a source of IFN-g-producing cells is present, either CD4+ or CD8+ T cells can mediate this protection. At the minimum, these findings reveal the significance of CD4+ T cells in providing protection against wild-type Brucella and some mutant Brucella strains. In contrast, a protective role for CD8+ T cells has been reported as early as 1989, when adoptively transferred CD4+ or CD8+ T cells from S19 B. abortus-immunized mice conferred significant protection against virulent B. abortus challenge [58]. Immunization with a ΔnorD ΔznuA B. abortus mutant induced IFN-γ-producing CD4+ and CD8+ T cells, further implicating the type of mutant used to influence T cell responses [59]. This was accentuated by the observation that mice vaccinated with ΔznuA B. melitensis mutant were protected in a CD8+ T cell-dependent fashion since mice deficient in CD4, but not CD8, were resistant to virulent B. melitensis challenge [50]. Others have suggested that brucellae persistence is due to the lack of memory CD8+ T cells [60], unlike ΔznuA B. melitensis-vaccinated mice, which were able to generate memory CD8+ T cells and protected against virulent challenge [50].
2.2. Th17 immunity in brucellosis
Evidence from the limited work on IL-17's role in brucellosis indicates that this cytokine has minimal impact upon protection against systemic brucellosis in immunocompetent animals [49, 50,62]. As described below under 3. Mucosal Vaccinations, the importance of IL-17 upon mucosal vaccination heightened when IL-17 compensated for the absence of IFN- γ [49,50]. Considered a primary source of IL-17, γδ T cells were found to be important for initial defense against B. abortus infections; however, resistance to infection was linked to γδ T cells' production of IFN-γ, not IL-17 [46], and IL-17 did not contribute to the development of osteoarthritis in mucosally or systemically Brucella challenged IFN-γ-/- mice [28]. Resistance to virulent nasal challenge with B. melitensis was also minimally affected in IL-17RA-/- and IL-23p19-/- mice [60].
Another source of Th17 cells resides with those T cells producing IL-22. Examination of IL-22 production revealed that mucosal immunization with ΔznuA B. melitensis mutant resulted in stimulation of IL-22 in immunocompetent and IFN-γ-/- mice [49,50]. Prominent IL-17 production was observed with antigen-restimulated CD8+ T cells [50]. The stimulation of IL-22 is associated with protection of the mucosal epithelium by enhancing the epithelial barrier and increasing defensins production [rev. in 63]. The stimulation of IL-22 may contribute to resistance to mucosal Brucella infections, and in particular, may account for the lack of oral infections as IL-22 production was elevated in orally vaccinated and nasally challenged mice [49]. Parenteral challenge of IL-22-/- mice with virulent B. melitensis 16M had minimal impact upon resistance to brucellosis [53]. Further studies are needed to determine IL-22's entire contribution to mucosal protection to brucellosis.
2.3. B cell immunity in brucellosis
Anti-Brucella polysaccharide antibodies are thought to be less important [53,64] than cell-mediated immunity in protection to brucellosis [8,10,13,65]. Although, passively transferred anti-Brucellas antiserum could reduce splenic B. abortus S19 colonization [58], as could passive transfer of an O-polysaccharide-specific monoclonal antibody in preventing infection with virulent B. abortus [66]. Further inquiry into B cells' role in immunity has found that B cell-deficient (μMT) mice were more resistant to B. abortus infection than were wild-type mice [64]. In fact, antibodies facilitated the uptake of GFP-expressing B. abortus into wild-type B cells but not in B cell-deficient (Jh-/-) mice [67]. Hence, B. abortus can infect B cells, resulting in the production of B cell-derived TGF-β1, having anti-inflammatory properties that may exacerbate chronic Brucella infections [67].
3. Mucosal Vaccinations
Thus far, we described the importance of eliciting cell-mediated immunity, be it CD4+ or CD8+ T cell-dependent that produces IFN-γ to protect against virulent Brucella challenge. Brucella infections produce a systemic disease regardless of the route of infection [3-5,7,10]. By convention brucellosis vaccinations are administered parenterally to livestock [65]. As a result, consideration of mucosal administration is often overlooked despite the fact that the majority of livestock and human infections occur mucosally. An interesting caveat is that even with oral exposures with Brucella not producing a gut infection [25, 68], brucellae can enter via the cellular prion protein on Peyer's patch microfold cells [69]. While unable to sustain an intestinal infection, oral gavage with live brucellosis vaccines has been successfully used for brucellosis vaccination of laboratory animals [49,70-73], livestock [74-76], red deer [77], and nonhuman primates [73]. In contrast, the naso-oropharyngeal lymph nodes (LNs) that drain the oral cavity and upper respiratory tract are susceptible to Brucella infections [78] as a result of animals sniffing or licking Brucella-infected aborted fetuses and/or infected placental tissues [79,80]. In humans, past clinical cases of brucellosis involved occurrence of pharyngitis [30-34], which can result in brucellae sequestration in the cervical LNs (CLNs) [34]. Such evidence suggests that the oral cavity and associated LNs and tonsils serve as the first site of mucosal exposure. This is consistent with the notion of oral exposures from consumption of contaminated dairy products, and these findings are recapitulated in experimental brucellosis in sheep [81] and in rhesus macaques [82].
3.1. Oral vaccination strategies for brucellosis
Only a handful of studies has examined the possibility of using mucosal vaccination strategies for protection against brucellosis to understand immune mechanisms of brucellosis resistance. When orally are vaccinated with the B. abortus RB51 cattle vaccine, mice show reduced splenic brucellae colonization by only ∼ 2 logs following virulent oral B. abortus challenge, and a little protection against parenteral challenge [72]. B. neotomae, a pathogen of wood rats and humans [83], was used as a heterologous species to orally vaccinate against B. abortus, and was irradiated to prevent its replication, while still maintaining its immunogenicity. Immunized mice were subsequently challenged parenterally with virulent B. abortus reducing brucellae colonization of spleen, livers, and lungs [70] to levels similar as others have shown for live attenuated Brucella mutants tested in mice. When orally administered, the purine auxotrophic mutant, ΔpurEK B. melitensis 16M (WR201) elicited potent protection in the lungs and reduced brucellae colonization of systemic tissues after nasal challenge with virulent B. melitensis 16M [71]. Such evidence demonstrates that if a suitable mutant is developed, it can be used to confer protection. In this same vein, outstanding protection against nasal challenge with virulent B. melitensis 16M was achieved in mice orally vaccinated with the ΔznuA B. melitensis mutant [49]. These orally vaccinated mice exhibited almost complete protection to brucellae colonization of systemic tissues. Interestingly, such protection was reduced, but not completely abrogated in IFN-γ-/- mice [49]. This remaining resistance in IFN-γ-/- mice orally vaccinated with ΔznuA B. melitensis was attributed to IL-17 since upon exogenous neutralization of IL-17, protection was abated [49]. IL-17 was less important in wild-type mice since in vivo IL-17 neutralization did not affect protection [49]. IL-17 had a much greater role in the IFN-γ-/- mice, given the absence of IFN-γ, and possibly, in compensating for this deficiency, elevations in IL-17-producing T cells were detected following in vitro antigen restimulation [49]. Such increases were greater than those detected for wild-type mice. One negative aspect for both mutants used in these studies was the high dose inoculum, ∼1011 CFUs, required for protection [49,71], especially when considering vaccination of humans; however, this dose is a similar order of magnitude used for oral vaccination of cattle [75,76].
3.2. Nasal vaccination strategies for brucellosis
A mucosal alternative to oral vaccination is immunizing via the nasal route. To test its effectiveness, nasal vaccination strategies were applied using RB51 or RB51 overexpressing Brucella's superoxide dismutase (RB51-SOD). Neither vaccine proved effective in protecting against nasal challenge with virulent B. abortus in contrast to mice nasally vaccinated with the smooth live cattle vaccine, strain 19 (S19) [84]. When combined with TLR agonists as adjuvants, nasal RB51 did improve efficacy [62]. Others found that nasal vaccination is effective against pulmonary challenge with virulent B. melitensis 16M [50]. A single, nasal dose of ΔznuA B. melitensis conferred potent protection against pulmonary B. melitensis challenge as evidenced by >50% of the mice having no detectable brucellae in their lungs or spleens [50]. This immune protection was IFN-g-dependent since IFN-γ-/- mice vaccinated with this mutant showed reduced efficacy, and what efficacy remained was abated subsequent in vivo IL-17 neutralization [50] similar to IFN-γ-/- mice orally immunized with ΔznuA B. melitensis [49]. This protective response was found to be CD8+ T cell-dependent in the lungs since ΔznuA B. melitensis nasally vaccinated CD8-/- mice showed no efficacy against pulmonary challenge with virulent B. melitensis in contrast to similarly vaccinated wild-type and CD4-/- mice [50]. Protection in the spleen was only partially compromised in ΔznuA B. melitensis-vaccinated CD8-/- mice, suggesting CD4+ T cells may compensate for the absence of CD8+ T cells. Likewise, CD8-/- mice nasally vaccinated with Rev. 1 lost protection in the lungs and spleen to pulmonary challenge [50]. Protection in the lungs was mediated by a pulmonary effector memory CD8+ T cells producing IFN-γ, TNF-α, and/or granzyme B [50]. Hence, both the vaccination strategy, e.g., route, and the vaccine used contribute to the type of T cell response generated for protection. This latter study [50] showed that the type of T cell needed for protection may also vary with immune compartment or tissue as systemic tissues were protected by both CD4+ and CD8+ T cells, whereas CD8+ T cells were required for the lungs. Additional work is needed to devise similar vaccination strategies for livestock.
4. Livestock and Experimental Vaccines
Brucellosis in livestock can result in fetal loss due to abortion, weak offspring, or reduced fertility [13]. This can impact livestock and milk production, with the latter often becoming contaminated with brucellae. The only commercial vaccines available for brucellosis are those for livestock [13,65]. No live or subunit vaccine has been approved for use in humans. Worldwide, three vaccines are currently used: the rough B. abortus mutant, RB51, used in cattle; the smooth vaccine, B. abortus S19, also used in cattle; and the smooth vaccine, B. melitensis Rev. 1, used in goats and sheep. These vaccines are normally given by the s.c. route. The oral B. suis strain 2 (S2) is administered via drinking water, and is used only in China [85]. 4.1 B. abortus S19 cattle vaccine.
Brucellosis in cattle was especially problematic in the US and many other European and South American countries during the 20th century. In the 1930s, as part of the “New Deal” sought to improve livestock industry and improve human health in the US, the State-Federal Cooperative Brucellosis Eradication Program was established to facilitate the elimination of brucellosis from livestock herds [86]. Brucellosis caused disease in livestock handlers inrural communities, and also affected urban areas because of the consumption of contaminated milk [31,86]. This program began with test and slaughter to screen for infected animals, and subsequently incorporated vaccination of seronegative animals to aid reducing the economic losses associated with contaminated milk and decline in beef cattle production [86,87].
In 1923, the cattle S19 vaccine arose initially as a spontaneous attenuated mutant [88], which was later found to be the result of the loss of the erythritol catabolic genes [89]. S19 vaccination of cattle proved highly efficacious against abortions caused by B. abortus [90], while having minimal impact upon normal calf births [91]. Importantly, S19 vaccine could reduce infections in animals already infected with B. abortus [87]. Although S19 was instrumental in the elimination of brucellosis in the US [87], the vaccine is only ∼70% efficacious in cattle [92]. S19 is normally given s.c. to heifer calves, but adults can be vaccinated using a reduced dose administered either s.c. or via the conjunctival route. Pregnant heifers orally vaccinated with S19 showed equivalent efficacy against Brucella-induced abortion as heifers vaccinated via the s.c. route [74, 75] demonstrating that S19 can be used as an oral vaccine. S19 has been used in humans in the former Soviet Union, where vaccination with S19 reduced B. melitensis infections in livestock workers [93], but no description of side-effects was included in this report. When similar vaccinations were performed in the US with S19 or with the live, attenuated small ruminant vaccine, B. melitensis Rev. 1, both vaccines caused reactogenicity in humans [94,95], abrogating further testing in humans.
4.2 B. abortus RB51 cattle vaccine
The current vaccine used in the US for protection to brucellosis is the rough B. abortus RB51, developing by repeated passage of wild-type B. abortus 2308 selected on rifampin- and penicillin-containing media [96]. RB51 exhibits a mutation that results in the interruption of the wboA glycosyltransferase, an enzyme important for O-Ag biosynthesis [97]. Bearing a rough phenotype readily enables serological diagnosis of B. abortus-infected animals, since cattle vaccinated with RB51 and who are not infected with wild-type B. abortus, do not produce antibodies to Brucella's O-Ag (LPS) [98]. This significant accomplishment permits serological herd surveillance. Furthermore, RB51 has a vaccine efficacy similar to that of S19 for preventing Brucella-induced abortion and fetal infection [87,99], not compromising the ability to vaccinate against brucellosis. However, variations in RB51's efficacy have been suggested to be age-related. Calves vaccinated at > 5-6 months of age are more resistant to Brucella-induced abortion during adult pregnancies than heifer calves vaccinated at 3 months of age [87,100]. In the US and in other countries, RB51 is used instead of S19 vaccine as part of a brucellosis eradication program, and is more effective where the prevalence of brucellosis is low [101]. Oral and s.c. RB51 vaccination also showed equivalent efficacy against Brucella-induced abortion and brucellae colonization in pregnant heifers [76].
4.3 B. melitensis Rev. 1 small ruminant vaccine
Brucellosis is also problematic for small ruminants, e.g., goats and sheep, mostly infected by B. melitensis [13,65,87]. B. melitensis Rev. 1 vaccine strain was originally derived from an avirulent streptomycin-resistant strain. This isolated revertant regained its sensitivity to streptomycin [102], and protects against Brucella-induced abortion in goats [103]. Rev. 1 is typically administered to sheep and goats either as a s.c. injection or applied via the conjunctiva [86], and is never given to pregnant animals so as to avoid vaccine-induced abortion [65, 87]. Rev. 1 is a smooth vaccine complicating serological surveillance in differentiating between vaccinated and naturally infected animals [87]. Incidentally, conjunctival administration can confer protection without producing a long-term antibody response [65].
4.4 B. suis strain 2 (S2) swine vaccine
Developed in China in the early 1950s, the B. suis strain 2 (S2) vaccine was attenuated as a result of serial passage of a virulent isolate from an aborted B. suis-infected fetal pig [85]. S2 vaccination is routinely done in China to curb B. suis and other Brucella infections in swine, and has significantly reduced brucellosis since its introduction in 1971 [85]. S2 is administered orally via the drinking water [85].
4.5 Subunit vaccines for brucellosis
The discussion thus far has focused on live attenuated vaccines for brucellosis, in large part because of their success in controlling disease and recapitulating aspects of immunity required for protection [13,65,87]. Generally, subunit vaccines for brucellosis have not been able to achieve immunity to the level obtained with a live vaccine [65, 104]. Ordinarily, protein or DNA vaccines yield one- to two-log reduction in splenic colonization from a level of 5-6 log colonization by virulent Brucella. Subunit vaccines clearly have the advantage of avoiding side-effects associated with live vaccines, and readily facilitate serological surveillance in vaccinated livestock. Yet, another advantage is the ability to impact multiple Brucella species because their proteins are highly conserved. Unfortunately, no successful subunit vaccine for brucellosis has been developed despite the multiple efforts. Large scale evaluation of different protein candidates in livestock is cost-prohibitive, thus, initial screens have been performed in laboratory animals. In mice, the use of the immunodominant protein, S-adenosyl-L-homocysteine hydrolase, resulted in 2-log reduction in splenic colonization [105]. The combination of two proteins, Omp16 and Omp19 resulted in nearly 2-log protection [106]. Similar success was also achieved using DNA vaccine approaches with the combination of bp26 plus trigger factor [107], IalB [108], omp25 [108], and ribosomal protein L9 [109].
On the other hand, some success was achieved using a heterologous vaccine vector, influenza virus [110]. In this study, 50 pregnant cows (3-4 months of gestation) were subdivided into 5 groups: one group dosed s.c. with PBS in 20% Montanide Gel01 adjuvant (unvaccinated control); one group s.c. vaccinated with S19; one group s.c. vaccinated with RB51; and the last two groups primed-boosted either s.c. or vaccinated initially via the conjunctival route with H5N1-L7/L12 & omp16, and 28 days later boosted with a heterologous H1N1-L7/L12 & omp16. At 5-6 months gestation, pregnant heifers were challenged s.c. with virulent wild-type B. abortus 544. S19 and RB51 conferred 89% (8/9) and 70% (7/10) protection against abortion, respectively; and the s.c. and conjunctival flu virus vaccinated heifers showed 90% (9/10) and 80% (8/10) protection against abortion, respectively. Control heifers showed a 70% incidence of abortion. The vaccinates displayed a similar magnitude of protection against both infection and Brucella-induced abortion, and all of the control (from the adjuvant-dosed heifer) fetuses and calves were culture-positive for Brucella [110]. Hence, alternative vaccine vectors that mimic immunity required for protection to Brucella are needed to further subunit vaccine approaches.
5. Conclusion
Brucellosis is commonly viewed as a problem impacting livestock health and economic loss for livestock producers having an economic impact attributed to losses in milk and meat production, and because of its zoonosis, brucellosis also remains a threat to human health. Eradication programs seldom reimburse livestock producers for animal loss. Current vaccines for livestock are only about 70% efficacious. Hence, better vaccines are needed to abrogate brucellosis in livestock, the primary source of infection. Development of enhanced vaccines is dependent on a better understanding of Brucella's pathogenesis, particularly in animals that are naturally susceptible to Brucella infections [10]. If a live vaccine is to be developed, it will require a DIVA component to allow the ability to distinguish vaccinated from infected animals during surveillance. Any subunit vaccine must achieve an efficacy better than conventional live vaccines. To date, most protein and DNA vaccines have not achieved the efficacy of conventional brucellosis vaccines [104], likely attributable to the lack of identifying adjuvants suitable to produce the desired protective immune response. Improved efficacy may be accomplished via a novel vaccine delivery system used as suggested by recent studies that showed higher efficacy using an attenuated influenza-based vector [110]. Certainly, vaccine formulation and route of delivery can influence outcomes as suggested by our studies [49,50,59]. Consideration of alternative methods rather than reliance on parenteral methods for vaccination can lead to vaccination strategies that produce improved efficacy and long-term memory response. Such improvements in protection came about by considering brucellosis as a mucosal disease, rather one that solely produces a systemic disease. Empowering mucosal approaches could harness additional lymphocytes to protect against infection, particularly since most infections occur following a mucosal exposure. Vaccine formulations that license a strong response by both CD4+ and CD8+ T cells, rather than the reliance on a single T cell subset may hold the answer to immune protection.
Acknowledgments
This work was supported by U. S. Public Health Grants R01 AI-123244 and R01 AI-125546, and a USDA-NIFA 2013-01165 grant. ZG is in part supported by AI-123244-02S1.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Mableson HE, Okello A, Picozzi K, Welburn SC. Neglected zoonotic diseases-the long and winding road to advocacy. PLoS Negl Trop Dis. 2014;8:e2800. doi: 10.1371/journal.pntd.0002800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pappas G, Papadimitriou P, Akritidis N, Christou L, Tsianos EV. The new global map of human brucellosis. Lancet Infect Dis. 2006;6:91–9. doi: 10.1016/S1473-3099(06)70382-6. [DOI] [PubMed] [Google Scholar]
- 3.Corbel MJ. Brucellosis in humans and animals. WHO; Geneva: 2006. pp. 1–102. [Google Scholar]
- 4.Pappas G, Akritidis N, Bosilkovski M, Tsianos E. Brucellosis. N Engl J Med. 2005;352:2325–36. doi: 10.1056/NEJMra050570. [DOI] [PubMed] [Google Scholar]
- 5.Franco MP, Mulder M, Gilman RH, Smits HL. Human brucellosis. Lancet Infect Dis. 2007;7:775–86. doi: 10.1016/S1473-3099(07)70286-4. [DOI] [PubMed] [Google Scholar]
- 6.Van der Henst C, de Barsy M, Zorreguieta A, Letesson JJ, De Bolle X. The Brucella pathogens are polarized bacteria. Microbes Infect. 2013;15:998–1004. doi: 10.1016/j.micinf.2013.10.008. [DOI] [PubMed] [Google Scholar]
- 7.Young EJ. Human brucellosis. Rev Infect Dis. 1983;5:821–42. doi: 10.1093/clinids/5.5.821. [DOI] [PubMed] [Google Scholar]
- 8.Byndloss MX, Tsolis RM. Brucella spp. virulence vactors and immunity. Annu Rev Anim Biosci. 2016;4:111–27. doi: 10.1146/annurev-animal-021815-111326. [DOI] [PubMed] [Google Scholar]
- 9.Corbel M. Brucellosis: an overview. Emerg Infect Dis. 1997;3:213–21. doi: 10.3201/eid0302.970219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.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;185:1505–17. doi: 10.1016/j.ajpath.2015.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.He Y. Analyses of Brucella pathogenesis, host immunity, and vaccine targets usingsystems biology and bioinformatics. Front Cell Infect Microbiol. 2012;2:2. doi: 10.3389/fcimb.2012.00002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Guzmán-Verri C, González-Barrientos R, Hernández-Mora G, Morales JA, Baquero-Calvo E, Chaves-Olarte E, Moreno E. Brucella ceti and brucellosis in cetaceans. Front Cell Infect Microbiol. 2012;2:3. doi: 10.3389/fcimb.2012.00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Olsen SC, Palmer MV. Advancement of knowledge of Brucella over the past 50 years. Vet Pathol. 2014;51:1076–89. doi: 10.1177/0300985814540545. [DOI] [PubMed] [Google Scholar]
- 14.Whatmore AM. Current understanding of the genetic diversity of Brucella, an expanding genus of zoonotic pathogens. Infect Genet Evol. 2009;9:1168–84. doi: 10.1016/j.meegid.2009.07.001. [DOI] [PubMed] [Google Scholar]
- 15.Bruce D. Observations on Malta Fever. Br Med J. 1889;1:1101–5. doi: 10.1136/bmj.1.1481.1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bruce D. Note on the discovery of a micro·organism in Malta Fever. Practitioner. 1887;39:161–70. [Google Scholar]
- 17.Wattam AR, Williams KP, Snyder EE, Almeida NF, Jr, Shukla M, Dickerman AW, et al. Analysis of ten Brucella genomes reveals evidence for horizontal gene transfer despite a preferred intracellular lifestyle. J Bacteriol. 2009;191:3569–79. doi: 10.1128/JB.01767-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.De Bolle X, Crosson S, Matroule JY, Letesson JJ. Brucella abortus cell cycle and infection are coordinated. Trends Microbiol. 2015;23:812–21. doi: 10.1016/j.tim.2015.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Godfroid J, Scholz HC, Barbier T, Nicolas C, Wattiau P, Fretin D, et al. Brucellosis at the animal/ecosystem/human interface at the beginning of the 21st century. Prev Vet Med. 2011;102:118–31. doi: 10.1016/j.prevetmed.2011.04.007. [DOI] [PubMed] [Google Scholar]
- 20.Bang B. Infectious abortion in cattle. J Comp Pathol Ther. 1906;19:191–202. [Google Scholar]
- 21.Poester FP, Samartino LE, Santos RL. Pathogenesis and pathobiology of brucellosis in livestock. Rev Sci Tech. 2013;32:105–15. doi: 10.20506/rst.32.1.2193. [DOI] [PubMed] [Google Scholar]
- 22.Godfroid J, DeBolle X, Roop RM, O'Callaghan D, Tsolis RM, Baldwin C, et al. The quest for a true One Health perspective of brucellosis. Rev Sci Tech Off Int Epiz. 2014;33:521–38. doi: 10.20506/rst.33.2.2290. [DOI] [PubMed] [Google Scholar]
- 23.Tsolis RM, Young GM, Solnick JV, Bäumler AJ. From bench to bedside: stealth of enteroinvasive pathogens. Nat Rev Microbiol. 2008;6:883–92. doi: 10.1038/nrmicro2012. [DOI] [PubMed] [Google Scholar]
- 24.Ariza J, Corredoira J, Pallares R, Viladrich PF, Rufi G, Pujol M, Gudiol F. Characteristics of and risk factors for relapse of brucellosis in humans. Clin Infect Dis. 1995;20:1241–9. doi: 10.1093/clinids/20.5.1241. [DOI] [PubMed] [Google Scholar]
- 25.Ablin J, Mevorach D, Eliakim R. Brucellosis and the gastrointestinal tract. The odd couple. J Clin Gastroenterol. 1997;24:25–9. doi: 10.1097/00004836-199701000-00005. [DOI] [PubMed] [Google Scholar]
- 26.Reguera JM, Alarcón A, Miralles F, Pachón J, Juárez C, Colmenero JD. Brucella endocarditis: clinical, diagnostic, and therapeutic approach. Eur J Clin Microbiol Infect Dis. 2003;22:647–50. doi: 10.1007/s10096-003-1026-z. [DOI] [PubMed] [Google Scholar]
- 27.Rajapakse CN. Bacterial infections: osteoarticular brucellosis. Baillieres Clin Rheumatol. 1995;9:161–77. doi: 10.1016/s0950-3579(05)80153-0. [DOI] [PubMed] [Google Scholar]
- 28.Skyberg JA, Thornburg T, Kochetkova I, Layton W, Callis G, Rollins MF, et al. IFN- γ-deficient mice develop IL-1-dependent cutaneous and musculoskeletal inflammation during experimental brucellosis. J Leukoc Biol. 2012;92:375–87. doi: 10.1189/jlb.1211626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lacey CA, Mitchell WJ, Brown CR, Skyberg, JA Temporal role for MyD88 in a model of Brucella-induced arthritis and musculoskeletal inflammation. Infect Immun. 2017;85:e00961–16. doi: 10.1128/IAI.00961-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ron-Román J, Ron-Garrido L, Abatih E, Celi-Erazo M, Vizcaíno-Ordóñez L, Calva-Pacheco J, et al. Human brucellosis in northwest Ecuador: typifying Brucella spp., seroprevalence, and associated risk factors. Vector Borne Zoonotic Dis. 2014;14:124–33. doi: 10.1089/vbz.2012.1191. [DOI] [PubMed] [Google Scholar]
- 31.Carpenter CM, Boak RA. The isolation of Brucella abortus from tonsils. JAMA. 1932;99:296–98. [Google Scholar]
- 32.Yinnon AM, Morali GA, Goren A, Rudensky B, Isacsohn M, Michel J, Hershko C. Effect of age and duration of disease on the clinical manifestations of brucellosis. A study of 73 consecutive patients in Israel. Isr J Med Sci. 1993;29:11–6. [PubMed] [Google Scholar]
- 33.Zachou K, Papamichalis PA, Dalekos GN. Severe pharyngitis in stockbreeders: an unusual presentation of brucellosis. Occup Med (Lond) 2008;58:305–7. doi: 10.1093/occmed/kqn020. [DOI] [PubMed] [Google Scholar]
- 34.von Bargen K, Gagnaire A, Arce-Gorvel V, de Bovis B, Baudimont F, Chasson L, et al. Cervical lymph nodes as a selective niche for Brucella during oral infections. PLoS One. 2015;10:e121790. doi: 10.1371/journal.pone.0121790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Celli J. The changing nature of the Brucella-containing vacuole. Cell Microbiol. 2015;17:951–8. doi: 10.1111/cmi.12452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Salcedo SP, Marchesini MI, Lelouard H, Fugier E, Jolly G, Balor S, et al. Brucella control of dendritic cell maturation is dependent on the TIR-containing protein Btp1. PLoS Pathog. 2008;4:e21. doi: 10.1371/journal.ppat.0040021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Alaidarous M, Ve T, Casey LW, Valkov E, Ericsson DJ, Ullah MO, et al. Mechanism of bacterial interference with TLR4 signaling by Brucella Toll/interleukin-1 receptor domain-containing protein TcpB. J Biol Chem. 2014;289:654–68. doi: 10.1074/jbc.M113.523274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Snyder GA, Deredge D, Waldhuber A, Fresquez T, Wilkins DZ, Smith PT, et al. Crystal structures of the Toll/Interleukin-1 receptor (TIR) domains from the Brucella protein TcpB and host adaptor TIRAP reveal mechanisms of molecular mimicry. J Biol Chem. 2014;289:669–79. doi: 10.1074/jbc.M113.523407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Barrionuevo P, Delpino MV, Pozner RG, Velásquez LN, Cassataro J, Giambartolomei GH. Brucella abortus induces intracellular retention of MHC-I molecules in human macrophages down-modulating cytotoxic CD8+ T cell responses. Cell Microbiol. 2013;15:487–502. doi: 10.1111/cmi.12058. [DOI] [PubMed] [Google Scholar]
- 40.Velásquez LN, Milillo MA, Delpino MV, Trotta A, Fernández P, Pozner RG, et al. Brucella abortus down-regulates MHC class II by the IL-6-dependent inhibition of CIITA through the downmodulation of IFN regulatory factor-1 (IRF-1) J Leukoc Biol. 2017;101:759–73. doi: 10.1189/jlb.4A0416-196R. [DOI] [PubMed] [Google Scholar]
- 41.Barrionuevo P, Cassataro J, Delpino MV, Zwerdling A, Pasquevich KA, García Samartino C, et al. Brucella abortus inhibits major histocompatibility complex class II expression and antigen processing through interleukin-6 secretion via Toll-like receptor 2. Infect Immun. 2008;76:250–62. doi: 10.1128/IAI.00949-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jiang X, Baldwin CL. Effects of cytokines on intracellular growth of Brucella abortus. Infect Immun. 1993;61:124–34. doi: 10.1128/iai.61.1.124-134.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Rodriguez-Zapata M, Salmeron I, Manzano L, Salmeron OJ, Prieto A, Alvarez-Mon M. Defective interferon-gamma production by T-lymphocytes from patients with acute brucellosis. Eur J Clin Invest. 1996;26:136–40. doi: 10.1046/j.1365-2362.1996.108250.x. [DOI] [PubMed] [Google Scholar]
- 44.Macedo GC, Magnani DM, Carvalho NB, Bruna-Romero O, Gazzinelli RT, Oliveira SC. Central role of MyD88-dependent dendritic cell maturation and proinflammatory cytokine production to control Brucella abortus infection. J Immunol. 2008;180:1080–7. doi: 10.4049/jimmunol.180.2.1080. [DOI] [PubMed] [Google Scholar]
- 45.Eze MO, Yuan L, Crawford RM, Paranavitana CM, Hadfield TL, Bhattacharjee AK, et al. Effects of opsonization and gamma interferon on growth of Brucella melitensis 16M in mouse peritoneal macrophages in vitro. Infect Immun. 2000;68:257–63. doi: 10.1128/iai.68.1.257-263.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Skyberg JA, Thornburg T, Rollins M, Huarte E, Jutila MA, Pascual DW. Murine and bovine γδ T cells enhance innate immunity against Brucella abortus infections. PloS-ONE. 2011;6:e21978. doi: 10.1371/journal.pone.0021978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhan Y, Cheers C. Endogenous interleukin-12 is involved in resistance to Brucella abortus infection. Infect Immun. 1995;63:1387–90. doi: 10.1128/iai.63.4.1387-1390.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhan Y, Cheers C. Control of IL-12 and IFN-γ production in response to live or dead bacteria by TNF and other factors. J Immunol. 1998;161:1447–53. [PubMed] [Google Scholar]
- 49.Clapp B, Skyberg JA, Yang X, Thornburg T, Walters N, Pascual DW. Protective live oral brucellosis vaccines stimulate Th1 and Th17 cell responses. Infect Immun. 2011;79:4165–74. doi: 10.1128/IAI.05080-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Clapp B, Yang X, Thornburg T, Walters N, Pascual DW. Nasal vaccination stimulates CD8+ T cells for potent protection against mucosal Brucella melitensis challenge. Immunol Cell Biol. 2016;94:496–508. doi: 10.1038/icb.2016.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Murphy EA, Sathiyaseelan J, Parent MA, Zou B, Baldwin CL. Interferon-γ is crucial for surviving a Brucella abortus infection in both resistant C57BL/6 and susceptible BALB/c mice. Immunology. 2001;103:511–8. doi: 10.1046/j.1365-2567.2001.01258.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vitry MA, Hanot Mambres D, De Trez C, Akira S, Ryffel B, Letesson JJ, Muraille E. Humoral immunity and CD4+ Th1 cells are both necessary for a fully protective immune response upon secondary infection with Brucella melitensis. J Immunol. 2014;192:3740–52. doi: 10.4049/jimmunol.1302561. [DOI] [PubMed] [Google Scholar]
- 53.Vitry MA, De Trez C, Goriely S, Dumoutier L, Akira S, Ryffel B, et al. Crucial role of IFN-γ-producing CD4+ Th1 cells but dispensable function of CD8+ T cell, B cell, Th2, and Th17 responses in the control of Brucella melitensis infection in mice. Infect Immun. 2012;80:4271–80. doi: 10.1128/IAI.00761-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yingst SL, Izadjoo M, Hoover DL. CD8 knockout mice are protected from challenge by vaccination with WR201, a live attenuated mutant of Brucella melitensis. Clin Dev Immunol. 2013;2013:686919. doi: 10.1155/2013/686919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhan Y, Kelso A, Cheers C. Differential activation of Brucella-reactive CD4+ T cells by Brucella infection or immunization with antigenic extracts. Infect Immun. 1995;63:969–75. doi: 10.1128/iai.63.3.969-975.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Svetić A, Jian YC, Lu P, Finkelman FD, Gause WC. Brucella abortus induces a novel cytokine gene expression pattern characterized by elevated IL-10 and IFN-gamma in CD4+ T cells. Int Immunol. 1993;5:877–83. doi: 10.1093/intimm/5.8.877. [DOI] [PubMed] [Google Scholar]
- 57.He Y, Vemulapalli R, Zeytun A, Schurig GG. Induction of specific cytotoxic lymphocytes in mice vaccinated with Brucella abortus RB51. Infect Immun. 2001;69:5502–8. doi: 10.1128/IAI.69.9.5502-5508.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Araya LN, Elzer PH, Rowe GE, Enright FM, Winter AJ. Temporal development of protective cell-mediated and humoral immunity in BALB/c mice infected with Brucella abortus. J Immunol. 1989;143:3330–7. [PubMed] [Google Scholar]
- 59.Yang X, Clapp B, Thornburg T, Hoffman C, Pascual DW. Vaccination with a ΔnorD ΔznuA Brucella abortus mutant confers potent protection against virulent challenge. Vaccine. 2016;34:5290–7. doi: 10.1016/j.vaccine.2016.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Durward-Diioia M, Harms J, Khan M, Hall C, Smith JA, Splitter GA. CD8+ T cell exhaustion, suppressed gamma interferon production, and delayed memory response induced by chronic Brucella melitensis infection. Infect Immun. 2015;83:4759–71. doi: 10.1128/IAI.01184-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hanot Mambres D, Machelart A, Potemberg G, De Trez C, Ryffel B, Letesson JJ, Muraille E. Identification of immune effectors essential to the control of primary and secondary intranasal infection with Brucella melitensis in mice. J Immunol. 2016;196:3780–93. doi: 10.4049/jimmunol.1502265. [DOI] [PubMed] [Google Scholar]
- 62.Surendran N, Sriranganathan N, Boyle SM, Hiltbold EM, Tenpenny N, Walker M, et al. Protection to respiratory challenge of Brucella abortus strain 2308 in the lung. Vaccine. 2013;31:4103–10. doi: 10.1016/j.vaccine.2013.06.078. [DOI] [PubMed] [Google Scholar]
- 63.Valeri M, Raffatellu M. Cytokines IL-17 and IL-22 in the host response to infection. Pathog Dis. 2016;74:ftw111. doi: 10.1093/femspd/ftw111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.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;203:1136–46. doi: 10.1093/infdis/jiq171. [DOI] [PubMed] [Google Scholar]
- 65.Goodwin ZI, Pascual DW. Brucellosis vaccines for livestock. Vet Immunol Immunopathol. 2016;181:51–8. doi: 10.1016/j.vetimm.2016.03.011. [DOI] [PubMed] [Google Scholar]
- 66.Winter AJ, Duncan JR, Santisteban CG, Douglas JT, Adams LG. Capacity of passively administered antibody to prevent establishment of Brucella abortus infection in mice. Infect Immun. 1989;57:3438–44. doi: 10.1128/iai.57.11.3438-3444.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Goenka R, Guirnalda PD, Black SJ, Baldwin CL. B Lymphocytes provide an infection niche for intracellular bacterium Brucella abortus. J Infect Dis. 2012;206:91–8. doi: 10.1093/infdis/jis310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Delpino MV, Marchesini MI, Estein SM, Comerci DJ, Cassataro J, Fossati CA, Baldi PC. A bile salt hydrolase of Brucella abortus contributes to the establishment of a successful infection through the oral route in mice. Infect Immun. 2007;75:299–305. doi: 10.1128/IAI.00952-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Nakato G, Hase K, Suzuki M, Kimura M, Ato M, Hanazato M, et al. Cutting Edge: Brucella abortus exploits a cellular prion protein on intestinal M cells as an invasive receptor. J Immunol. 2012;189:1540–4. doi: 10.4049/jimmunol.1103332. [DOI] [PubMed] [Google Scholar]
- 70.Dabral N, Moreno-Lafont M, Sriranganathan N, Vemulapalli R. Oral immunization of mice with gamma-irradiated Brucella neotomae induces protection against intraperitoneal and intranasal challenge with virulent B. abortus 2308. PLoS One. 2014;9:e107180. doi: 10.1371/journal.pone.0107180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Izadjoo MJ, Bhattacharjee AK, Paranavitana CM, Hadfield TL, Hoover DL. Oral vaccination with Brucella melitensis WR201 protects mice against intranasal challenge with virulent Brucella melitensis 16M. Infect Immun. 2004;72:4031–9. doi: 10.1128/IAI.72.7.4031-4039.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Pasquali P, Rosanna A, Pistoia C, Petrucci P, Ciuchini F. Brucella abortus RB51 induces protection in mice orally infected with the virulent strain B. abortus 2308. Infect Immun. 2003;71:2326–30. doi: 10.1128/IAI.71.5.2326-2330.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chen TH, Elberg SS. Immunization against Brucella infections: immune response of mice, guinea pigs, and Cynomolgus philipinensis to live and killed Brucella melitensis strain Rev. I administered by various methods. J Infect Dis. 1970;122:489–500. doi: 10.1093/infdis/122.6.489. [DOI] [PubMed] [Google Scholar]
- 74.Nicoletti P, Milward FW. Protection by oral administration of Brucella abortus strain 19 against an oral challenge exposure with a pathogenic strain of Brucella. Am J Vet Res. 1983;44:1641–3. [PubMed] [Google Scholar]
- 75.Nicoletti P. Vaccination of cattle with Brucella abortus strain 19 administered by differing routes and doses. Vaccine. 1984;2:133–5. doi: 10.1016/0264-410x(84)90004-5. [DOI] [PubMed] [Google Scholar]
- 76.Elzer PH, Enright FM, Colby L, Hagius SD, Walker JV, Fatemi MB, et al. Protection against infection and abortion induced by virulent challenge exposure after oral vaccination of cattle with Brucella abortus strain RB51. Am J Vet Res. 1998;59:1575–8. [PubMed] [Google Scholar]
- 77.Arenas-Gamboa AM, Ficht TA, Davis DS, Elzer PH, Kahl-McDonagh M, Wong-Gonzalez A, Rice-Ficht AC. Oral vaccination with microencapsuled strain 19 vaccine confers enhanced protection against Brucella abortus strain 2308 challenge in red deer (Cervus elaphus elaphus) J Wildl Dis. 2009;45:1021–9. doi: 10.7589/0090-3558-45.4.1021. [DOI] [PubMed] [Google Scholar]
- 78.Meador VP, Warner DP, Deyoe BL. Distribution of Brucella abortus organisms in calves after conjunctival exposure. Am J Vet Res. 1988;49:2015–7. [PubMed] [Google Scholar]
- 79.Samartino LE, Enright FM. Pathogenesis of abortion of bovine brucellosis. Comp Immunol Microbiol Infect Dis. 1993;16:95–101. doi: 10.1016/0147-9571(93)90001-l. [DOI] [PubMed] [Google Scholar]
- 80.Schumaker B. Risks of Brucella abortus spillover in the Greater Yellowstone area. Rev Sci Tech. 2013;32:71–7. doi: 10.20506/rst.32.1.2185. [DOI] [PubMed] [Google Scholar]
- 81.Suraud V, Olivier M, Bodier CC, Guilloteau LA. Differential expression of homing receptors and vascular addressins in tonsils and draining lymph nodes: effect of Brucella infection in sheep. Vet Immunol Immunopathol. 2007;115:239–50. doi: 10.1016/j.vetimm.2006.11.008. [DOI] [PubMed] [Google Scholar]
- 82.Mense MG, Borschel RH, Wilhelmsen CL, Pitt ML, Hoover DL. Pathologic changes associated with brucellosis experimentally induced by aerosol exposure in rhesus macaques (Macaca mulatta) Am J Vet Res. 2004;65:644–52. doi: 10.2460/ajvr.2004.65.644. [DOI] [PubMed] [Google Scholar]
- 83.Suárez-Esquivel M, Ruiz-Villalobos N, Jiménez-Rojas C, Barquero-Calvo E, Chacón-Díaz C, Víquez-Ruiz E, et al. Brucella neotomae infection in humans, Costa Rica. Emerg Infect Dis. 2017;23:997–1000. doi: 10.3201/eid2306.162018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Surendran N, Sriranganathan N, Lawler H, Boyle SM, Hiltbold EM, Heid B, et al. Efficacy of vaccination strategies against intranasal challenge with Brucella abortus in BALB/c mice. Vaccine. 2011;29:2749–55. doi: 10.1016/j.vaccine.2011.01.090. [DOI] [PubMed] [Google Scholar]
- 85.Xin X. Orally administrable brucellosis vaccine: Brucella suis strain 2 vaccine. Vaccine. 1986;4:212–6. doi: 10.1016/0264-410x(86)90131-3. [DOI] [PubMed] [Google Scholar]
- 86.Busch LA, Parker RL. Brucellosis in the United States. J Infect Dis. 1972;125:289–94. doi: 10.1093/infdis/125.3.289. [DOI] [PubMed] [Google Scholar]
- 87.Olsen SC, Stoffregen WS. Essential role of vaccines in brucellosis control and eradication programs for livestock. Expert Rev Vaccines. 2005;4:915–28. doi: 10.1586/14760584.4.6.915. [DOI] [PubMed] [Google Scholar]
- 88.Buck JM. Studies of vaccination during calfhood to prevent bovine infectious abortion. J Agr Res. 1930;41:667–89. [Google Scholar]
- 89.Sangari FJ, García-Lobo JM, Agüero J. The Brucella abortus vaccine strain B19 carries a deletion in the erythritol catabolic genes. FEMS Microbiol Lett. 1994;121:337–42. doi: 10.1111/j.1574-6968.1994.tb07123.x. [DOI] [PubMed] [Google Scholar]
- 90.Confer AW, Hall SM, Faulkner CB, Espe BH, Deyoe BL, Morton RJ, Smith RA. Effects of challenge dose on the clinical and immune responses of cattle vaccinated with reduced doses of Brucella abortus strain 19. Vet Microbiol. 1985;10:561–75. doi: 10.1016/0378-1135(85)90065-3. [DOI] [PubMed] [Google Scholar]
- 91.Wright AE. Report of the co-operative bovine brucellosis work in the United States. Proc US Livestock San Assoc. 1942;47:149–54. [Google Scholar]
- 92.Lubroth J, Rweyemamu MM, Viljoen G, Diallo A, Dungu B, Amanfu W. Veterinary vaccines and their use in developing countries. Rev Sci Tech. 2007;26:179–201. [PubMed] [Google Scholar]
- 93.Vershilova PA. The use of live vaccine for vaccination of human beings against brucellosis in the USSR. Bull World Health Organ. 1961;24:85–9. [PMC free article] [PubMed] [Google Scholar]
- 94.Spink WW, Hall JW, 3rd, Finstad J, Mallet E. Immunization with viable Brucella organisms. Results of a safety test in humans. Bull World Health Organ. 1962;26:409–19. [PMC free article] [PubMed] [Google Scholar]
- 95.Pappagianis D, Elberg SS, Crouch D. Immunization against Brucella infections. Effects of graded doses of viable attenuated Brucella melitensis in humans. Am J Epidemiol. 1966;84:21–31. doi: 10.1093/oxfordjournals.aje.a120624. [DOI] [PubMed] [Google Scholar]
- 96.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;28:171–88. doi: 10.1016/0378-1135(91)90091-s. [DOI] [PubMed] [Google Scholar]
- 97.Vemulapalli R, McQuiston JR, Schurig GG, Sriranganathan N, Halling SM, Boyle SM. Identification of an IS711 element interrupting the wboA gene of Brucella abortus vaccine strain RB51 and a PCR assay to distinguish strain RB51 from other Brucella species and strains. Clin Diagn Lab Immunol. 1999;6:760–4. doi: 10.1128/cdli.6.5.760-764.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Stevens MG, Hennager SG, Olsen SC, Cheville NF. Serologic responses in diagnostic tests for brucellosis in cattle vaccinated with Brucella abortus 19 or RB51. J Clin Microbiol. 1994;32:1065–6. doi: 10.1128/jcm.32.4.1065-1066.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Olsen SC. Immune responses and efficacy after administration of a commercial Brucella abortus strain RB51 vaccine to cattle. Vet Ther. 2000;1:183–91. [PubMed] [Google Scholar]
- 100.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;57:1153–56. [PubMed] [Google Scholar]
- 101.Moriyón I, Grilló MJ, Monreal D, González D, Marín C, López-Goñi I, et al. Rough vaccines in animal brucellosis: structural and genetic basis and present status. Vet Res. 2004;35:1–38. doi: 10.1051/vetres:2003037. [DOI] [PubMed] [Google Scholar]
- 102.Herzberg M, Elberg SS. Immunization against Brucella infection. III. Response of mice and guinea pigs to injection of viable and nonviable suspensions of a streptomycin-dependent mutant of Brucella melitensis. J Bacteriol. 1955;69:432–5. doi: 10.1128/jb.69.4.432-435.1955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Elberg SS, Faunce K., Jr Immunization against Brucella infection. VI. Immunity conferred on goats by a nondependent mutant from a streptomycin-dependent mutant strain of Brucella melitensis. J Bacteriol. 1957;73:211–7. doi: 10.1128/jb.73.2.211-217.1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Yang X, Skyberg JA, Cao L, Thornburg T, Clapp B, Pascual DW. Progress in Brucella vaccine development. Front Biol. 2013;8:60–77. doi: 10.1007/s11515-012-1196-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.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;61:159–67. doi: 10.1111/j.1574-695X.2010.00758.x. [DOI] [PubMed] [Google Scholar]
- 106.Pasquevich KA, Estein SM, García Samartino C, Zwerdling A, Coria LM, Barrionuevo P, et al. 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;77:436–45. doi: 10.1128/IAI.01151-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yang X, Hudson M, Walters N, Bargatze RF, Pascual DW. Selection of protective epitopes for Brucella melitensis using DNA vaccination. Infect Immun. 2005;73:7297–303. doi: 10.1128/IAI.73.11.7297-7303.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.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. doi: 10.1016/j.vaccine.2006.07.046. [DOI] [PubMed] [Google Scholar]
- 109.Jain S, Afley P, Dohre SK, Saxena N, Kumar S. Evaluation of immunogenicity and protective efficacy of a plasmid DNA vaccine encoding ribosomal protein L9 of Brucella abortus in BALB/c mice. Vaccine. 2014;32:4537–42. doi: 10.1016/j.vaccine.2014.06.012. [DOI] [PubMed] [Google Scholar]
- 110.Tabynov K, Kydyrbayev Z, Ryskeldinova S, Yespembetov B, Zinina N, Assanzhanova N, et al. Novel influenza virus vectors expressing Brucella L7/L12 or Omp16 proteins in cattle induced a strong T-cell immune response, as well as high protectiveness against B. abortus infection. Vaccine. 2014;32:2034–41. doi: 10.1016/j.vaccine.2014.02.058. [DOI] [PubMed] [Google Scholar]