Open access peer-reviewed chapter

Genome-Based Vaccinology Applied to Bovine Anaplasmosis

Written By

Itzel Amaro-Estrada and Sergio D. Rodríguez-Camarillo

Submitted: 25 September 2017 Reviewed: 22 November 2017 Published: 21 March 2018

DOI: 10.5772/intechopen.72637

From the Monograph

Farm Animals Diseases, Recent Omic Trends and New Strategies of Treatment

Edited by Rosa Estela Quiroz-Castañeda

Chapter metrics overview

1,451 Chapter Downloads

View Full Metrics

Abstract

Bovine anaplasmosis is an infectious non-contagious disease transmitted mainly by ticks or fomites contaminated with Anaplasma marginale. Once cattle have developed the disease it can be treated with antibiotics or chemotherapy, although with partial success. Still, there is no effective and global prophylactic method available, mainly because of variability and diversity showed by different A. marginale strains distributed worldwide. In this regard, several proteins have been proposed as immunogens, MSPs, OMPs, Type IV Secretion System Proteins and some other hypothetical proteins, which have been chosen either by experimental evidence or more recently by genome-based analysis. So far, the results suggest that a single molecule will not be enough to trigger a protective immune response in the host, so it is necessary to identify other proteins or epitopes with adequate immunological properties, a process in which omics tools have potential. In order to develop a vaccine against bovine anaplasmosis, it has been proposed by the use of combinations of molecules, exposure formats and application protocols to provide an effective control of the disease.

Keywords

  • Anaplasma marginale
  • genomics
  • vaccinology
  • bovine anaplasmosis
  • OMP
  • MSP

1. Introduction

Tick-borne diseases are considered a major obstacle and the cause of great economic impact for livestock production [1]. Control measures currently available for tick-borne diseases include the use of acaricides for reduction or tick populations, specific chemotherapy, chemoprophylaxis, controlled exposure and vaccination. These measures limit losses caused by ticks and the diseases they transmit [2]. Globally, the most important rickettsial disease in cattle is bovine anaplasmosis caused by Anaplasma marginale [3].

Vaccination is the method of choice for preventing infectious diseases. In the case of bovine anaplasmosis, while there are live vaccines, these pose many risks, including: (i) spread of other blood-borne pathogens, such as Babesia spp. and virus-like bovine leukemia virus to mention a few [4]; (ii) standardization of vaccine dose; (iii) maintenance of carrier animals; (iv) quality control and production; (v) maintenance and transportation of vaccines to the end user, including the need of a cold chain [5, 6].

Inactivated vaccines based on the use of the extracted bacteria while effective, are restricted due to: (i) potential contamination with erythrocyte membrane antigens; (ii) wide antigenic variation between Anaplasma strains [7]; (iii) possible short-term immunity; and (iv) amelioration of clinical signs while not preventing infection, so the animals remain carriers for the rest of their lives [8].

Vaccine design is compounded by the large antigenic and genetic diversity found in strains from a region to another, within the same herd and even within the same animal [9, 10]. Current investigations focus on the search for new alternatives for designing vaccines and diagnostic assays [11, 12, 13]. In this review, besides discussing some fundamental aspects of anaplasmosis, we focus on the molecular characteristics that make A. marginale capable to persist in nature including: (i) the mechanisms of evasion of the host’s immune response; (ii) diversity; (iii) hypervariability of some of its components; or (iv) replication.

Advertisement

2. The causal agent

Anaplasma marginale is a tick-borne pathogen and the causative agent of bovine anaplasmosis [14]. A. marginale is classified in the Rickettsial order, reorganized into two families such as Anaplasmataceae and Rickettsiaceae [15]. Anaplasma organisms are obligate intracellular Gram-negative rickettsia, found exclusively within vacuoles derived from the erythrocyte membrane, and are membrane-bound within the cytoplasm of the host cell. A. marginale persist in nature in mammalian and ticks hosts, which serve as reservoirs of infection [16]. In the bovine, A. marginale infect erythrocytes and endothelial cells [17]. The infection process in endothelial cell has not been described and it is considered as of no relevance within the persistence mechanisms for the rickettsia [18]. Ticks transmit rickettsia from the salivary glands during feeding (Figure 1), and within the erythrocyte, the rickettsia replicates by binary fission to form 8–12 initial bodies and exit from the erythrocyte does not involve destruction of the host’s cell [19]. Once out of the host cell, the initial bodies invade new erythrocytes in endless cycles. Ticks acquires the rickettsia while feeding on carrier hosts. In the tick, the rickettsia infects midgut cells, where there is a first cycle of replication and from here dense forms move to other tissues. After several rounds of replication, dense forms travel to the salivary glands where the rickettsia is transmitted to a new mammalian host [16, 20].

Figure 1.

Proposed life cycle of A. marginale in bovine. Modified from [20].

Many species of ticks have been implicated in the transmission of A. marginale, although Dermacentor andersoni is the most studied of all [16]. Recent studies have focused on the role of ticks Rhipicephalus (Boophilus) microplus as vectors of bovine anaplasmosis, an issue discussed below [21, 22].

The first A. marginale sequenced genome published [23] present a very complete description of the known features to that date. Up to now, there are at least 2 full genome sequences published (St. Maries and Florida), other 2 almost complete sequences, and 10 more partially annotated sequences (NCBI/Genome). Comparative studies with the available genome sequences have been carried out with very interesting results as far as the study of hypervariable genes/antigens [24], while other genome/transcriptome analysis have focused on the transmission phenotype genes involved [25].

In this chapter, we will review the information available, since the publication of the St. Maries genome to the specific genes that have been studied as vaccine candidates. Vaccination against bovine Anaplasmosis traditionally relied on attenuated [5, 26] and inactivated organisms [6, 27]. Both of these types of vaccines will, in most cases, induce a degree of immunity, which do not prevent infection. There are a number of examples of experimental vaccines, but in this review only those cases where there has been modification of the organism or recombinant antigens are included as vaccine candidates will be discussed.

2.1. Major surface proteins as vaccine candidates

Over the last 30 years, six neutralization-sensitive membrane-exposed proteins were originally reported in 1984 [28, 29] and later named Major Surface Proteins [30]. This group of proteins has been the subject of a great number of studies aimed at developing a vaccine.

Msp1 is a heterodimer composed by Msp1a (100 kDa) and Msp1b (105 kDa) joined in a non-covalent manner and are exposed on the surface of A. marginale [31]. Details of the genetics, structure, and composition of these two peptides have been described elsewhere [24]. Msp1a is coded by a single gene and its product is composed of a variable number of tandem-repeat units of 28–32 aa in length at the amino terminus. The carboxyl end is conserved and extends mostly as an intracellular domain [32]. Msp1b is coded by a multigene family that expresses several variants during the acute and chronic phases of the infection [33]. Msp1 is an adhesin toward erythrocytes and tick-gut cells whereas Msp1b only toward bovine erythrocytes [34, 35]. It is now known that the adhesion function in Msp1a is located in the variable region which is composed of several short amino acid sequences (repeats). Analysis of Msp1a repeats from different isolates has shown no association between tick-transmission capabilities and the type or number of repeats present within this variable region [36]. Repeat sequences though have been used for defining genotypes associated to other markers such as the pseudogenes present in Msp2 [37] or distinguishing genotypes in cattle superinfected with two or more different Anaplasma strains [10, 38, 39].

Bioinformatic analysis carried out with Msp1a amino acid sequence have shown that Msp1a variable region is rich in highly immunogenic B cell epitopes, yet these sequences are considered distracters to the immune system of the host, despite the fact specific monoclonal antibodies neutralize infection to both the erythrocytes and tick cells [40]. Msp1a also contains Th1 cell epitopes in the carboxyl conserved region, which may be involved in immunoprotection [41]. Initial immunization experiments with recombinant Msp1a showed that autologous immunity was afforded; yet heterologous immunity was poor [42]. In a semi-controlled experiment where Msp1a was used as marker for matching the vaccine strain and the local strain in an inactivated vaccine trial in the field, which resulted in partial immunity except when the challenge was carried with the autologous strain but not when challenge was carried with heterologous Msp1a-matched strains [7].

More recently, two epitopes, STSSQL (Am1), located within the consensus sequence of the repeat and SEASTSSQLGA (Am2), which is located in C-terminal end of the 28-aa repetitive motif of the MSP1a protein, were identified using phage display technology for identification of immunodominant epitopes recognized by a neutralizing monoclonal antibody against MSP1a; these peptides were recognized by many but not all healthy infected animals tested by ELISA assay [43]. These synthetic peptides were conjugated to bovine serum albumin and used for immunization of mice, which the authors claim were infected with A. marginale was achieved. Details of the challenge strain used in this experiment are absent [44] and there have been no follow-up articles using the same peptides in cattle.

The inclusion of Msp1a or its “conserved epitopes” should be carried with caution as the diversity of this marker is wider in regions where the tick vector is R. microplus than in its absence [10, 45]. Vaccination based on Msp1a epitopes is further confounded by the fact that the Msp1a variable region may contain one or several repeats which may include the same or different repeats and that there are more than 300 repeats reported [45, 46] and this number is bound to increase as the number of epidemiology studies is published. Furthermore, the number of Msp1a-distinct strains (up to nine) within the same herd or even within the same animal [10, 47, 48] complicates even more the design of an effective vaccine.

While the adhesion function of Msp1a protein is located on the variable region, number or type of different Msp1a repeat sequences has not been associated with tick-transmission phenotype [49, 50]. Repeat sequences though have been used for defining genotypes associated to other markers such as the pseudogenes present in Msp2 [37] or distinguishing genotypes in cattle superinfected with two or more different Anaplasma strains [10, 38, 39].

As for Msp1b, initial experiments indicated that it was a poor immunogen [51, 52]. Further evidence indicates that immunization of naive calves with a recombinant fraction containing the Msp1a-T cell epitopes linked to recombinant Msp1b1, induced a much greater antibody titer to Msp1b than what was previously observed [52, 53].

DNA vaccines based on either one of Msp1a or Msp1b have given disappointing results. In an early effort using a construction pVCL/MSP1a for the immunization of mice and cattle [54], a predominant IgG1 antibody pattern was observed in the two immunized calves. No challenge was performed, however, as it has been proven, an IgG2 response is necessary in order to achieve protection [11, 55]. In a similar study with an Msp1b DNA construct, immunization not only did not induce immunity but immunized animals developed more severe clinical disease than controls [56]. These latter authors did not test for the type of immunoglobulin induced by vaccination.

Further studies with DNA vaccines constructs which included bovine herpes virus 1 tegument protein, BVP22 domain and an invariant-chain major histocompatibility complex class II-targeting motif capable of enhancing dendritic cell antigen uptake and presentation were fused to a sequence encoding a B and T cell antigen from the A. marginale Msp1a [57, 58]. This approach included the intradermal inoculation with a mixture of 2 mg of DNA encoding the molecular adjuvants bovine FLT3L and GM-CSF to recruit DCs to the intradermal immunization site, the results of this experiment were very encouraging as they stimulated the desired type of immune response with rapid recall of antibody production over a reasonable period after a single immunization. This study, however, suffers of several flaws (i) the inoculation in different points of the dendritic cells stimulant and the vaccine itself, (ii) while the responses as measured, were in all senses the appropriate ones for resistance, there was no challenge of vaccinated animals and second, the age of the vaccines would not allow for distinction between a solid immune response induced by vaccination or natural resistance commonly observed in animals under 1 year of age [3].

Msp2 is a highly immunodominant 36 kDa protein coded by a multigene family consisting of a functional gene that codes for the amino and carboxyl ends and a variable number of pseudogenes (5 in the St. Maries strain) [59] which recombine with the main gene, through gene conversion, in a single expression site such that the protein is expressed as a new variant in each cycle of rickettsemia, every 6–8 weeks [60]. msp2 pseudogenes code for hypervariable, hydrophilic sequences containing highly immunogenic B cell epitopes which induce a new immune response consequent to a new Msp2 variant [61]. The amino and carboxyl ends of the protein are hydrophobic conserved segments inserted into the membrane of the rickettsia [62] (and contain Th1 cell type epitopes that are preserved along different geographic strains) [29, 55].

Just like Msp1 and Msp1b, Msp2 was discovered through the neutralization of infection by specific monoclonal antibodies and it was considered a vaccine candidate [28], this has not been the case. While the hypervariable region of the protein contains a number of highly immunogenic type B cell epitopes, it has been recognized that antibody directed to these epitopes are distractors of the immune response during the periodical appearance of Msp2 variants [60, 63].

Msp3 is a very immunogenic 86 k Da protein located on the surface of the rickettsia [28]. msp3 is also composed by a central hypervariable region coded by several pseudogenes which recombine with the conserved amino and carboxyl ends [64, 65]. Based on previous studies [64] and analysis of the published genome sequence, it is speculated that MSP2 and MSP3 originated from a common ancestor [66], and have diverged since that event. Sequence identity between msp3 and msp2 pseudogenes is reduced, an average of 38%, identity within msp3 pseudogenes 68% and, within msp2 pseudogenes 78% [66, 67]. Immunologically, Msp2 and Msp3 share epitopes recognized in vitro by CD4+ cells clones from vaccinated cattle [55]. Recombination of pseudogenes in a mosaic pattern also adds to the presentation of polymorphic antigens that, when resolved through 2D electrophoresis, are observed as a series of antigens with the same molecular weight and different but very close isoelectric points [68]. Appearance of Msp2 and Msp3 variants in the persistently infected bovine gives rise to a more complex situation with negative implications for immune protection. Though Msp3 induces production of large amounts of antibodies [69], protection afforded is very limited [70]. Msp3 is known to cross-react with other rickettsiae such as A. ovis, Ehrlichia risticii, E. wengii, E. equi and E. ruminantium, which make it unsuitable for specific A. marginale sero-diagnosis [71].

Msp4 is a 31 kDa protein, encoded by single highly conserved gene, msp4 [72]. Msp4 is also present in A. marginale subsp. centrale with 83% identity in the nucleotide sequences and 91.7% in the amino acid level [73]. To date, there is no solid evidence that Msp4 may be involved in protection as, initial studies showed lack of recognition by sera of animals immunized with an initial body membrane fraction [42], however when animals are immunized with a recombinant Msp4 adjuvated with Iscometrix as adjuvant, there seems to induce an antibody response. msp4 has been used as a base for phylogenetic studies which have shown that there are variations in 168 bp and, of these, 39 bp show utility in parsimony analysis such that isolates from several countries in the Americas can be grouped according to their geographic location [50]. Msp4 is highly conserved over several Mexican isolates [9].

Msp5 is a highly conserved 19 kDa MW protein in A. marginale, A. marginale subsp. centrale and A. phagocytophilum [23, 73, 74, 75]. Immunization with Msp5 induces the production of large quantities of non-protective antibodies [69] so it is no suitable for vaccination. Animals naturally infected with the rickettsia produce specific antibodies that can be found in recent and old infections so the protein has been used successfully in a diagnostic competitive-ELISA test [76]. Despite the cross-reaction of antibody between A. marginale and A. marginale subsp. centrale at the competitive-ELISA [74, 77], the test has been adopted as the standard for serologic diagnostic of bovine anaplasmosis.

2.2. Type IV secretion system proteins

Secretion systems in bacteria are complex structures by which they communicate with its environment. There are several secretion systems some which span both the inner membrane (IM) and the outer membrane (OM), and those that span the OM [78]. Among several secretion systems described in nature, type 4 secretion systems (T4SSs) have the unique ability to mediate translocation of DNA (in addition to proteins) into bacterial or eukaryotic target cells. T4SSs are found in both Gram-negative and Gram-positive bacteria and also in some archaea [79]. Their most common role is to mediate the conjugation of plasmid DNA; thus, these systems contribute to the spread of plasmid-borne antibiotic resistance genes. As the ability to conjugate is a common bacterial trait, T4SSs are the most ubiquitous secretion systems in nature. In addition, T4SSs are involved in bacterial pathogenesis in a few organisms, and they mediate the secretion of transforming proteins in Helicobacter pylori, toxins in Bordetella pertussis and other effector proteins required to support an intracellular lifestyle in bacteria such as Legionella pneumophila [79].

Along with the publication of the first complete genome of A. marginale, and its annotation, some real or putative homologous genes of T4SS were described in St. Maries genome [23]. Although many studies have been done about MSPs, so far, we still require other approaches to find better vaccine candidates. An approach was shotgun sequencing of the proteins of a membrane-enriched fraction of A. marginale, which induced an antibody response in naive calves [80]. In this study, 25 immunoblot positive spots were sequenced and identified through their annotation in the genome. Among the proteins identified VirB9, VirB10 and conjugal transfer protein (CTP), were shown to stimulate an antibody response. Further studies using the same membrane-enriched fractions for the immunization of young cattle showed that their antibodies (IgG2) and Th cells reacted with the recombinant versions of CTP, VirB9 and VirB10 proteins [12]. In a more sophisticated study using far-Western blotting to identify protein linkage between possible antigenic proteins, it was shown that VirB proteins, VirB9-1, VirB9-2 and Virb10 when physically linked, could stimulate a more specific and stronger immune response than when used individually [81]. While presence of B cell epitopes is important in any protein to be used as vaccine candidate, Th cell epitopes are also important as their presence might determine the actual potential use of any antigen in a vaccine. An interesting study, takes synthetic overlapping peptides from VirB9-1, VirB9-2 and VirB10 to test for the presence of such epitopes [82]. T cells from six different MHC Class-II phenotypes outer membrane fraction immunized animals were tested and as expected, it was observed that not all animals reacted with peptides from all three TFSS proteins. While all six animals reacted to the membrane fraction which contained all three VirB9-1, VirB9-2 and VirB10, some animals did not react against rVirB9–1, others reacted poorly against rVirB9-2 or rVirB10 or against only one or two of the overlapping synthetic peptides [82]. The differences in response of T cells from these animals are explained in the context of the Class-II MHC molecules involved in presentation of the epitopes. Interestingly, these authors restricted themselves to Holstein cattle as the subjects to their studies yet, at least in Mexico and other Latin American countries, Holstein cattle are used for milk production and most of them are reared under conditions which preclude the contact with ticks.

These studies clearly show that an immune response that fills the criteria for protection as described is induced [11, 55], these authors though, fall short of proving that the induced immunity is protective as there was no actual confrontation with the virulent live agent. In a different study recombinant VirB9, Virb10 and Elongation Factor Tu (EF-Tu) were tested against the sera of immunoprotected animals naturally infected with two A. marginale isolates [83]. These works showed that while all experimentally infected cattle with the autologous isolate had relevant antibodies (IgG2) against VirB9, VirB10 and EF-Tu, only 87% of the animals naturally infected with a heterologous isolate reacted with the recombinant protein by ELISA.

2.3. Outer membrane proteins

The outer membrane of bacteria delimits its structure and is the interface with the host cell. Outer membrane proteins (OMPs) are key components of Gram-negative bacteria and because are involved in adhesion and infection processes they are targeted on vaccine development.

Some effective attempts have been achieved using whole membrane fractions as immunogens against bacterial diseases and due to their relevance OMPs from several pathogens have been extensively studied and proposed as vaccine component. It is known that in addition to composition, OMPs show diversity in function too, but they share structural patterns. Usually, regions with antigenic properties are located on the extracellular loops and show variable composition, meanwhile residues in the transmembrane β-barrel show the highest conservation [61, 84, 85, 86]. However, the use of individual components has only been partially successful [86, 87, 88, 89, 90, 91, 92], although, it appears that the most relevant OMP antigens have not yet been identified.

High-throughput sequencing technologies are currently available and allow the identification of several genes with potential important functions in the metabolism of the pathogen or in the interaction with its surroundings. For example, from the complete genome of Anaplasma marginale [23], the existence of additional outer membrane protein has been elucidated. Additionally, to Msp2 and Msp3, new members of pfam01617 family have been identified, and designated as Omp 1-14. omp2, omp3 and omp6 genes are not transcribed in A. marginale-infected erythrocytes, tick midgut and salivary glands, and the IDE8 tick cell line, while OMPs 1, 4, 7, 8, 9 and 11 were confirmed to be differentially expressed as proteins in those cell types [93]. Unlike Msp2 and Msp3, these OMPs exhibit high conservation at sequence level as seen in the follow-up of the infection and in comparative analyses with the St. Maries and Florida strain genomes, which increases the possibility of choosing molecules capable of inducing a protective immune response against bovine Anaplasmosis. Omp7 to Omp9 appear as tandem repeats with almost 75% amino acid identity, Omp10 is related to Omp7 to Omp9 with ~ 30% residues identity and Omp6 is a truncated and it is not expressed version of Omp10. Omp7 to Omp9 are part of protective outer membrane fraction and are highly expressed than Omp10 [23, 25, 93, 94].

In spite of the fact that none of these molecules has induced protection when is applied as an individual protein, the importance of OMPs in protective immune response induction has been revealed above all as complexes or associated with a membrane environment.

For example, protection against Leptospira was reached using OmpL1 and LipL41 expressed simultaneously in the context of the E. coli membrane but, immunization with either membrane-associated protein or as part of a mixture of non-membrane-associated proteins was not protective [53, 95].

These results confirm the importance of OMPs in the infection process and the generation of a protective immune response against pathogens and also reveal the interactions between OMPs and other proteins as well as with their environment. However, production, solubilization and purification of membrane-associated recombinant proteins is not easily achieved [96].

2.4. Hypothetical proteins

Genomic analyses of A. marginale have allowed identification of novel annotated proteins whose function has not yet been determined, however, in silico analysis and predictions may provide unrevealed information about immunogenic potential.

Some hypothetical proteins have been identified by structure prediction of β-barrel outer membrane and orthology and bioinformatic analysis, such as Am1108, Am127, Am216, Am202, Am936, Am854, Am368, Am854, Am1041, Am109 and Am1096. Some of these proteins have been evaluated as recombinant molecules and recognized by IgG from immunized animals with outer membranes protein, in this case, Am1108 and Am216 elicited specific T cell response proliferation [13, 97]. On the other hand, cattle immunized with recombinant Am854 or Am936 developed higher bacteremia as compared to adjuvant-only controls and outer membrane vaccinates after challenge [13].

The absence of a protective immune response after application of recombinant proteins presumably exposed to A. marginale, and therefore, with antigenic characteristics still seems to be insufficient to develop prophylactic methods against bovine anaplasmosis.

Although genomic analyses have revealed valuable information about the composition of A. marginale, it will be necessary to complement this knowledge with experimental evidence based in other methods, such as proteomic and transcriptomic tools.

Advertisement

Acknowledgments

Authors thank financial support to projects SEP-CONACYT 161618 granted to SDR and INIFAP SIGI 11343533011 granted to IAE.

References

  1. 1. Jonsson NN, Bock RE, Jorgensen WK. Productivity and health effects of anaplasmosis and babesiosis on Bos Indicus cattle and their crosses, and the effects of differing intensity of tick control in Australia. Veterinary Parasitology. 2008;155(1–2):1-9
  2. 2. Kocan KM, de la Fuente J, Blouin EF. Advances toward understanding the molecular biology of the Anaplasma-tick interface. Frontiers in Bioscience. 2008;13:7032-7045
  3. 3. Aubry P, Geale DW. A review of bovine anaplasmosis. Transboundary and Emerging Diseases. 2011;58(1):1-30
  4. 4. Rogers R, Dimmock CK, de Vos AJ, Rodwell BJ. Bovine leucosis virus contamination of a vaccine produced in vivo against bovine babesiosis and anaplasmosis. Australian Veterinary Journal. 1988;65(9):285-287. Available from: http://dx.doi.org/10.1111/j.1751-0813.1988.tb16144.x
  5. 5. Abdala AA, Pipano E, Aguirre DH, Gaido AB, Zurbriggen MA, Mangold AJ, et al. Frozen and fresh Anaplasma centrale vaccines in the protection of cattle against Anaplasma marginale infection. Revue d'Élevage et de Médecine Vétérinaire des Pays Tropicaux. 1990;43(2):155-158
  6. 6. Rodríguez SD, Garcîa Ortiz MA, Hernández Salgado G, Santos Cerda NA, Aboytes Torre R, Cantó Alarcón GJ. Anaplasma marginale inactivated vaccine: Dose titration against a homologous challenge. Comparative Immunology, Microbiology and Infectious Diseases. 2000;23(4):239-252
  7. 7. Ocampo Espinoza V, Vázquez JES, Aguilar MD, Ortiz MÁG, Alarcón GJC, Rodríguez SD. Anaplasma marginale: Lack of cross-protection between strains that share MSP1a variable region and MSP4. Veterinary Microbiology. 2006;114(1–2):34-40. Available from: http://www.sciencedirect.com/science/article/pii/S037811350500444X
  8. 8. Reinbold JB, Coetzee JF, Hollis LC, Nickell JS, Riegel CM, Christopher JA, et al. Comparison of iatrogenic transmission of Anaplasma marginale in Holstein steers via needle and needle-free injection techniques. American Journal of Veterinary Research. 2010;71(10):1178-1188
  9. 9. Jiménez-Ocampo R, Vega y Murguía CA, Oviedo N, Rojas Ramírez EE, García-Ortiz MA, Preciado de la Torre JF, et al. Diversidad genética de la región variable de los genes msp1a y msp4 en cepas de Anaplasma marginale de México genetic diversity of the msp1a gene variable region and msp4 gene of Anaplasma marginale strains from Mexico. Revista Mexicana de Ciencias Pecuarias. 2012;3(3):373-387
  10. 10. Castañeda-Ortiz EJ, Ueti MW, Camacho-Nuez M, Mosqueda JJ, Mousel MR, Johnson WC, et al. Association of Anaplasma marginale strain superinfection with infection prevalence within tropical regions. PLoS One. 2015;10(3):e0120748. Available from: http://dx.plos.org/10.1371/journal.pone.0120748
  11. 11. Barigye R, García-Ortiz MA, Rojas Ramírez EE, Rodríguez SD. Identification of IgG2-specific antigens in Mexican Anaplasma marginale strains. Annals of the New York Academy of Sciences. 2004;1026:84-94
  12. 12. Lopez JE, Palmer GH, Brayton KA, Dark MJ, Leach SE, Brown WC. Immunogenicity of Anaplasma marginale type IV secretion system proteins in a protective outer membrane vaccine. Infection and Immunity. 2007;75(5):2333-2342. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1865776/
  13. 13. Ducken DR, Brown WC, Alperin DC, Brayton KA, Reif KE, Turse JE, et al. Subdominant outer membrane antigens in Anaplasma marginale: Conservation, antigenicity, and protective capacity using recombinant protein. PLoS One. 2015;10(6):e0129309. Available from: http://dx.plos.org/10.1371/journal.pone.0129309
  14. 14. Ristic M. Anaplasmosis. In: Diseases of Cattle in the Tropics. Dordrecht: Springer Netherlands; 1981. pp. 327-344
  15. 15. Dumler JS, Barbet AF, Bekker CP, Dasch GA, Palmer GH, Ray SC, et al. Reorganization of genera in the families Rickettsiaceae and Anaplasmataceae in the order Rickettsiales: Unification of some species of Ehrlichia with Anaplasma, Cowdria with Ehrlichia and Ehrlichia with Neorickettsia, descriptions of six new species combinations and designation of Ehrlichia equi and “HGE agent” as subjective synonyms of Ehrlichia phagocytophila. International Journal of Systematic and Evolutionary Microbiology. 2001;51(Pt 6):2145-2165
  16. 16. Kocan KM, de la Fuente J, Blouin EF, Coetzee JF, S a E. The natural history of Anaplasma marginale. Veterinary Parasitology. 2010;167(2–4):95-107
  17. 17. Carreño AD, Alleman AR, Barbet AF, Palmer GH, Noh SM, Johnson CM. In vivo endothelial cell infection by Anaplasma marginale. Veterinary Pathology. 2007;44(1):116-118
  18. 18. Wamsley HL, Alleman AR, Johnson CM, Barbet AF, Abbott JR. Investigation of endothelial cells as an in vivo nidus of Anaplasma marginale infection in cattle. Veterinary Microbiology. 2011;153(3–4):264-273
  19. 19. Kessler RH, Ristic M. In vitro cultivation of Anaplasma marginale: Invasion of and development in noninfected erythrocytes. American Journal of Veterinary Research. 1979;40(12):1774-1776
  20. 20. Rodríguez SD, García Ortiz MÁ, Jiménez Ocampo R, Vega y Murguía CA. Molecular epidemiology of bovine anaplasmosis with a particular focus in Mexico. Infection, Genetics and Evolution. 2009;9(6):1092-1101
  21. 21. Futse JE, Ueti MW, Knowles DP, Palmer GH. Transmission of Anaplasma marginale by Boophilus Microplus: Retention of vector competence in the absence of vector-pathogen interaction. Journal of Clinical Microbiology. 2003;41(8):3829-3834
  22. 22. Ruiz PMG, Passos LMF, Ribeiro MFB. Lack of infectivity of a Brazilian Anaplasma marginale isolate for Boophilus microplus ticks. Veterinary Parasitology. 2005;128(3–4):325-331
  23. 23. Brayton KA, Kappmeyer LS, Herndon DR, Dark MJ, Tibbals DL, Palmer GH, et al. Complete genome sequencing of Anaplasma marginale reveals that the surface is skewed to two superfamilies of outer membrane proteins. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(3):844-849
  24. 24. Dark MJ, Al-Khedery B, Barbet AF. Multistrain genome analysis identifies candidate vaccine antigens of Anaplasma marginale. Vaccine. 2011;29(31):4923-4932
  25. 25. Pierlé SA, Dark MJ, Dahmen D, Palmer GH, Brayton KA. Comparative genomics and transcriptomics of trait-gene association. BMC Genomics. 2012;13:669. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3542260&tool=pmcentrez&rendertype=abstract
  26. 26. Ristic M, Carson CA. Methods of immunoprophylaxis against bovine anaplasmosis with emphasis on use of the attenuated Anaplasma marginale vaccine. Advances in Experimental Medicine and Biology. 1977;93:151-188
  27. 27. Wilson JS, Trace JC. A new inactivated vaccine for bovine anaplasmosis. Bulletin - Office International Des Epizooties. 1966;66(1):897-902
  28. 28. Palmer GH, McGuire TC. Immune serum against Anaplasma marginale initial bodies neutralizes infectivity for cattle. Journal of Immunology. 1984;133(2):1010-1015
  29. 29. McGuire TC, Palmer GH, Goff WL, Johnson MI, Davis WC. Common and isolate-restricted antigens of Anaplasma marginale detected with monoclonal antibodies. Infection and Immunity. 1984;45(3):697-700
  30. 30. Palmer GH, Kocan KM, Barron SJ, Hair JA, Barbet AF, Davis WC, et al. Presence of common antigens, including major surface protein epitopes, between the cattle (intraerythrocytic) and tick stages of Anaplasma marginale. Infection and Immunity. 1985;50(3):881-886
  31. 31. Barbet AF, Palmer GH, Myler PJ, McGuire TC. Characterization of an immunoprotective protein complex of Anaplasma marginale by cloning and expression of the gene coding for polypeptide Am105L. Infection and Immunity. 1987;55(10):2428-2435
  32. 32. Allred DR, McGuire TC, Palmer GH, Leib SR, Harkins TM, McElwain TF, et al. Molecular basis for surface antigen size polymorphisms and conservation of a neutralization-sensitive epitope in Anaplasma marginale. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(8):3220-3224. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC53867/
  33. 33. Viseshakul N, Kamper S, Bowie MV, Barbet AF. Sequence and expression analysis of a surface antigen gene family of the rickettsia Anaplasma marginale. Gene. 2000;253(1):45-53
  34. 34. McGarey DJ, Allred DR. Characterization of hemagglutinating components on the Anaplasma marginale initial body surface and identification of possible adhesins. Infection and Immunity. 1994;62(10):4587-4593
  35. 35. McGarey DJ, Barbet AF, Palmer GH, McGuire TC, Allred DR. Putative adhesins of Anaplasma marginale: Major surface polypeptides 1a and 1b. Infection and Immunity. 1994;62(10):4594-4601
  36. 36. Cabezas-Cruz A, Passos LMF, Lis K, Kenneil R, Valdes JJ, Ferrolho J, et al. Functional and immunological relevance of Anaplasma marginale major surface protein 1a sequence and structural analysis. PLoS One. 2013;8(6)
  37. 37. Palmer GH, Futse JE, Leverich CK, Knowles DP, Rurangirwa FR, Brayton KA. Selection for simple major surface protein 2 variants during Anaplasma marginale transmission to immunologically naïve animals. Infection and Immunity. 2007;75(3):1502-1506
  38. 38. Futse JE, Brayton KA, Dark MJ, Knowles DP, Palmer GH. Superinfection as a driver of genomic diversification in antigenically variant pathogens. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(6):2123-2127
  39. 39. Leverich CK, Palmer GH, Knowles DP, Brayton KA. Tick-borne transmission of two genetically distinct Anaplasma marginale strains following superinfection of the mammalian reservoir host. Infection and Immunity. 2008;76(9):4066-4070
  40. 40. Palmer GH, Waghela SD, Barbet AF, Davis WC, McGuire TC. Characterization of a neutralization-sensitive epitope on the am 105 surface protein of Anaplasma marginale. International Journal for Parasitology. 1987;17(7):1279-1285
  41. 41. Brown WC, McGuire TC, Mwangi W, Kegerreis KA, Macmillan H, Lewin HA, et al. Major histocompatibility complex class II DR-restricted memory CD4(+) T lymphocytes recognize conserved immunodominant epitopes of Anaplasma marginale major surface protein 1a. Infection and Immunity. 2002;70(10):5521-5532
  42. 42. Tebele N, Palmer GH. Crossprotective immunity between the Florida and a Zimbabwe stock of Anaplasma marginale. Tropical Animal Health and Production. 1991;23(4):197-202
  43. 43. Santos PS, Nascimento R, Rodrigues LP, Santos FAA, Faria PCB, Martins JRS, et al. Functional epitope core motif of the Anaplasma marginale major surface protein 1a and its incorporation onto bioelectrodes for antibody detection. PLoS One. 2012;7(3):e33045
  44. 44. Santos PS, Sena AAS, Nascimento R, Araújo TG, Mendes MM, Martins JRS, et al. Epitope-based vaccines with the Anaplasma marginale MSP1a functional motif induce a balanced humoral and cellular immune response in mice. PLoS One. 2013;8(4):1-9
  45. 45. Quiroz-Castañeda RE, Amaro-Estrada I, Rodríguez-Camarillo SD. Anaplasma marginale: Diversity, virulence, and vaccine landscape through a genomics approach. BioMed Research International. 2016;2016:9032085. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC5005611/
  46. 46. Cabezas-Cruz A, de la Fuente J. Anaplasma marginale major surface protein 1a: A marker of strain diversity with implications for control of bovine anaplasmosis. Ticks and Tick-borne Diseases. 2015;6(3):205-210
  47. 47. Palmer GH, Knowles DP, Rodriguez J-L, Gnad DP, Hollis LC, Marston T, et al. Stochastic transmission of multiple genotypically distinct Anaplasma marginale strains in a herd with high prevalence of anaplasma infection. Journal of Clinical Microbiology. 2004;42(11):5381-5384. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC525272/
  48. 48. Ybañez AP, Ybañez RHD, Claveria FG, Cruz-Flores MJ, Xuenan X, Yokoyama N, et al. High genetic diversity of Anaplasma marginale detected from Philippine cattle. The Journal of Veterinary Medical Science. 2014;76(7):1009-1014. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24717413
  49. 49. de la Fuente J, Garcia-Garcia JC, Blouin EF, Kocan KM. Characterization of the functional domain of major surface protein 1a involved in adhesion of the rickettsia Anaplasma marginale to host cells. Veterinary Microbiology. 2003;91(2–3):265-283
  50. 50. de la Fuente J, Ruybal P, Mtshali MS, Naranjo V, Shuqing L, Mangold AJ, et al. Analysis of world strains of Anaplasma marginale using major surface protein 1a repeat sequences. Veterinary Microbiology. 2007;119(2–4):382-390 [cited 2016 Jan 4]. Available from: http://www.sciencedirect.com/science/article/pii/S0378113506003828
  51. 51. Palmer GH, Barbet AF, Kuttler KL, McGuire TC. Detection of an Anaplasma marginale common surface protein present in all stages of infection. Journal of Clinical Microbiology. 1986;23(6):1078-1083
  52. 52. Brown WC, Palmer GH, Lewin HA, McGuire TC. CD4(+) T lymphocytes from calves immunized with Anaplasma marginale major surface protein 1 (MSP1), a heteromeric complex of MSP1a and MSP1b, preferentially recognize the MSP1a carboxyl terminus that is conserved among strains. Infection and Immunity. 2001;69(11):6853-6862
  53. 53. Macmillan H, Norimine J, Brayton KA, Palmer GH, Brown WC. Physical linkage of naturally complexed bacterial outer membrane proteins enhances immunogenicity. Infection and Immunity. 2008;76(3):1223-1229
  54. 54. Arulkanthan A, Brown WC, McGuire TC, Knowles DP. Biased immunoglobulin G1 isotype responses induced in cattle with DNA expressing msp1a of Anaplasma marginale. Infection and Immunity. 1999;67(7):3481-3487
  55. 55. Brown WC, Shkap V, Zhu D, McGuire TC, Tuo W, McElwain TF, et al. CD4(+) T-lymphocyte and immunoglobulin G2 responses in calves immunized with Anaplasma marginale outer membranes and protected against homologous challenge. Infection and Immunity. 1998;66(11):5406-5413
  56. 56. de Andrade GM, Machado RZ, Vidotto MC, Vidotto O. Immunization of bovines using a DNA vaccine (pcDNA3.1/MSP1b) prepared from the Jaboticabal strain of Anaplasma marginale. Annals of the New York Academy of Sciences 2004;1026:257–266
  57. 57. Mwangi W, Brown WC, Splitter GA, Zhuang Y, Kegerreis K, Palmer GH. Enhancement of antigen acquisition by dendritic cells and MHC class II-restricted epitope presentation to CD4+ T cells using VP22 DNA vaccine vectors that promote intercellular spreading following initial transfection. Journal of Leukocyte Biology. 2005;78(2):401-411
  58. 58. Mwangi W, Brown WC, Splitter GA, Davies CJ, Howard CJ, Hope JC, et al. DNA vaccine construct incorporating intercellular trafficking and intracellular targeting motifs effectively primes and induces memory B- and T-cell responses in outbred animals. Clinical and Vaccine Immunology. 2007;14(3):304-311
  59. 59. Palmer GH, Eid G, Barbet AF, McGuire TC, McElwain TF. The immunoprotective Anaplasma marginale major surface protein 2 is encoded by a polymorphic multigene family. Infection and Immunity. 1994;62(9):3808-3816
  60. 60. French DM, Brown WC, Palmer GH. Emergence of Anaplasma marginale antigenic variants during persistent rickettsemia. Infection and Immunity. 1999;67(11):5834-5840
  61. 61. Barbet AF, Lundgren A, Yi J, Rurangirwa FR, Palmer GH. Antigenic variation of Anaplasma marginale by expression of MSP2 mosaics. Infection and Immunity. 2000;68(11):6133-6138
  62. 62. Noh SM, Brayton KA, Knowles DP, Agnes JT, Dark MJ, Brown WC, et al. Differential expression and sequence conservation of the Anaplasma marginale msp2 gene superfamily outer membrane proteins. Infection and Immunity. 2006;74(6):3471-3479
  63. 63. Abbott JR, Palmer GH, Howard CJ, Hope JC, Brown WC. Anaplasma marginale major surface protein 2 CD4+−T-cell epitopes are evenly distributed in conserved and Hypervariable regions (HVR), whereas linear B-cell epitopes are predominantly located in the HVR. Infection and Immunity. 2004;72(12):7360-7366
  64. 64. Alleman AR, Palmer GH, McGuire TC, McElwain TF, Perryman LE, Barbet AF. Anaplasma marginale major surface protein 3 is encoded by a polymorphic, multigene family. Infection and Immunity. 1997;65(1):156-163
  65. 65. Meeus PFM, Brayton KA, Palmer GH, Barbet AF. Conservation of a gene conversion mechanism in two distantly related paralogues of Anaplasma marginale. Molecular Microbiology. 2003;47(3):633-643 [cited 2016 Feb 3]. Available from: http://doi.wiley.com/10.1046/j.1365-2958.2003.03331.x
  66. 66. Meeus PF, Barbet AF. Ingenious gene generation. Trends in Microbiology. 2001;9(8):353-356
  67. 67. Brayton KA, Knowles DP, McGuire TC, Palmer GH. Efficient use of a small genome to generate antigenic diversity in tick-borne ehrlichial pathogens. Proceedings of the National Academy of Sciences. 2001;98(7):4130-4135
  68. 68. Barbet AF, Blentlinger R, Yi J, Lundgren AM, Blouin EF, Kocan KM. Comparison of surface proteins of Anaplasma marginale grown in tick cell culture, tick salivary glands, and cattle. Infection and Immunity. 1999;67(1):102-107
  69. 69. Palmer GH, McElwain TF. Molecular basis for vaccine development against anaplasmosis and babesiosis. Veterinary Parasitology. 1995;57(1–3):233-253
  70. 70. McGuire TC, Davis WC, Brassfield AL, McElwain TF, Palmer GH. Identification of Anaplasma marginale long-term carrier cattle by detection of serum antibody to isolated MSP-3. Journal of Clinical Microbiology. 1991;29(4):788-793
  71. 71. Alleman AR, Barbet AF. Evaluation of Anaplasma marginale major surface protein 3 (MSP3) as a diagnostic test antigen. Journal of Clinical Microbiology. 1996;34(2):270-276
  72. 72. Oberle SM, Barbet AF. Derivation of the complete msp4 gene sequence of Anaplasma marginale without cloning. Gene. 1993;136(1–2):291-294
  73. 73. Molad T, Brayton KA, Palmer GH, Michaeli S, Shkap V. Molecular conservation of MSP4 and MSP5 in Anaplasma marginale and A. Centrale vaccine strain. Veterinary Microbiology. 2004;100(1–2):55-64
  74. 74. Visser ES, TC MG, Palmer GH, Davis WC, Shkap V, Pipano E, et al. The Anaplasma marginale msp5 gene encodes a 19-kilodalton protein conserved in all recognized Anaplasma species. Infection and Immunity. 1992;60(12):5139-5144
  75. 75. Strik NI, Alleman AR, Barbet AF, Sorenson HL, Wamsley HL, Gaschen FP, et al. Characterization of Anaplasma phagocytophilum major surface protein 5 and the extent of its cross-reactivity with A. marginale. Clinical and Vaccine Immunology. 2007;14(3):262-268
  76. 76. Torioni de Echaide S, Knowles DP, McGuire TC, Palmer GH, Suarez CE, McElwain TF. Detection of cattle naturally infected with Anaplasma marginale in a region of endemicity by nested PCR and a competitive enzyme-linked immunosorbent assay using recombinant major surface protein 5. Journal of Clinical Microbiology. 1998;36(3):777-782
  77. 77. Knowles D, Torioni de Echaide S, Palmer G, McGuire T, Stiller D, McElwain T. Antibody against an Anaplasma marginale MSP5 epitope common to tick and erythrocyte stages identifies persistently infected cattle. Journal of Clinical Microbiology. 1996;34(9):2225-2230
  78. 78. Costa TRD, Felisberto-Rodrigues C, Meir A, Prevost MS, Redzej A, Trokter M, et al. Secretion systems in gram-negative bacteria: Structural and mechanistic insights. Nature Reviews. Microbiology. 2015;13(6):343-359
  79. 79. Alvarez-Martinez CE, Christie PJ. Biological diversity of prokaryotic type IV secretion systems. Microbiology and Molecular Biology Reviews. 2009;73(4):775-808
  80. 80. Lopez JE, Siems WF, Palmer GH, Brayton KA, McGuire TC, Norimine J, et al. Identification of novel antigenic proteins in a complex Anaplasma marginale outer membrane immunogen by mass spectrometry and genomic mapping. Infection and Immunity. 2005;73(12):8109-8118
  81. 81. Morse K, Norimine J, Palmer GH, Sutten EL, Baszler TV, Brown WC. Association and evidence for linked recognition of type IV secretion system proteins VirB9-1, VirB9-2, and VirB10 in Anaplasma marginale. Infection and Immunity. 2012;80(1):215-227
  82. 82. Morse K, Norimine J, Hope JC, Brown WC. Breadth of the CD4+ T cell response to Anaplasma marginale VirB9-1, VirB9-2 and VirB10 and MHC class II DR and DQ restriction elements. Immunogenetics. 2012;64(7):507-523. Available from: http://dx.doi.org/10.1007/s00251-012-0606-4
  83. 83. Araújo FR, Costa CM, Ramos CAN, Farias TA, De Souza IIF, Melo ESP, et al. IgG and IgG2 antibodies from cattle naturally infected with Anaplasma marginale recognize the recombinant vaccine candidate antigens VirB9, VirB10, and elongation factor-Tu. Memórias do Instituto Oswaldo Cruz. 2008;103(2):186-190
  84. 84. Zhang JZ, Guo H, Winslow GM, Yu XJ. Expression of members of the 28-Kilodalton major outer membrane protein family of Ehrlichia chaffeensis during persistent infection. Infection and Immunity. 2004;72(8):4336-4343
  85. 85. Callaghan MJ, Buckee CO, Jolley KA, Kriz P, Maiden MCJ, Gupta S. The effect of immune selection on the structure of the meningococcal Opa protein repertoire. Guttman DS, editor. PLoS Pathogens. 2008;4(3):e1000020
  86. 86. Baldo L, Desjardins CA, Russell JA, Stahlhut JK, Werren JH. Accelerated microevolution in an outer membrane protein (OMP) of the intracellular bacteria Wolbachia. BMC Evolutionary Biology. 2010;10:48
  87. 87. Lin J, Huang S, Zhang Q. Outer membrane proteins: Key players for bacterial adaptation in host niches. Microbes and Infection. 2002;4(3):325-331
  88. 88. Wang Y, Chen Z, Qiao F, Zhong Z, Xu J, Wang Z, et al. and the outer membrane properties of Brucella melitensis. 2010;303:92-100
  89. 89. Brattig NW, Bazzocchi C, Kirschning CJ, Reiling N, Büttner DW, Ceciliani F, et al. The major surface protein of Wolbachia endosymbionts in filarial nematodes elicits immune responses through TLR2 and TLR4. Journal of Immunology. 2004;173(1):437-445
  90. 90. Oliveira TL, Grassmann AA, Schuch RA, Clair A. Evaluation of the Leptospira interrogans outer membrane protein OmpL37 as a vaccine candidate. PLoS ONE. 2015;10(11): e0142821. Available from: https://doi.org/10.1371/journal.pone.0142821
  91. 91. Pan J, Li C, Ye Z. Immunoproteomic approach for screening vaccine candidates from bacterial outer membrane proteins. In: Thomas S, editor. Vaccine Design. Methods in Molecular Biology. 2016;1404:519-528. Vaccines for Veterinary Diseases, Volume 2. New York, NY: Humana Press; 2016
  92. 92. Singh R, Garg N, Shukla G, Capalash N, Sharma P, Fernandez MM. Immunoprotective eefficacy of Acinetobacter baumannii outer membrane protein, FilF, predicted in silico as a potential vaccine candidate. Frontiers in Microbiology. 2016;7(February):1-9
  93. 93. Noh SM, Brayton KA, Knowles DP, Agnes JT, Dark MJ, Brown WC, et al. Differential expression and sequence conservation of the Anaplasma marginale msp2 gene superfamily outer membrane proteins. Infection and Immunity. 2006;74(6):3471-3479
  94. 94. Crosby FL, Wamsley HL, Pate MG, Lundgren AM, Noh SM, Munderloh UG, et al. Knockout of an outer membrane protein operon of Anaplasma marginale by transposon mutagenesis. BMC Genomics. 2014;15(1):278. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24725301
  95. 95. Haake DA, Mazel MK, McCoy AM, Milward F, Chao G, Matsunaga J, et al. Leptospiral outer membrane proteins OmpL1 and LipL41 exhibit synergistic immunoprotection. Infection and Immunity. 1999;67(12):6572-6582
  96. 96. Wen Z, Boddicker MA, Kaufhold RM, Khandelwal P, Durr E, Qiu P, et al. Recombinant expression of chlamydia trachomatis major outer membrane protein in E. coli outer membrane as a substrate for vaccine research. BMC Microbiology. 2016:1-13
  97. 97. Nuñez PA, Moretta R, Ruybal P, Wilkowsky S, Farber MD. Immunogenicity of hypothetical highly conserved proteins as novel antigens in Anaplasma marginale. Current Microbiology. 2013;68(3):269-277. Available from: http://dx.doi.org/10.1007/s00284-013-0475-6

Written By

Itzel Amaro-Estrada and Sergio D. Rodríguez-Camarillo

Submitted: 25 September 2017 Reviewed: 22 November 2017 Published: 21 March 2018