SciELO - Scientific Electronic Library Online

 
vol.55 suppl.1Epidemiology of rickettsioses by Rickettsia parkeri andother emerging and reemerging species associated with anthropization in Latin AmericaResúmenes de conferencias magistrales author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

Related links

  • Have no similar articlesSimilars in SciELO

Share


Acta Médica Costarricense

On-line version ISSN 0001-6002Print version ISSN 0001-6012

Acta méd. costarric vol.55  suppl.1 San José Jul. 2013

 

Conferencias Magistrales

Rickettsiosis: pathogenesis, inmunidad y desarrollo de vacunas

Rickettsioses: pathogenesis, immunity, and vaccine development

Gustavo Valbuena


Resumen

Varias especies dentro del género Rickettsia son altamente patogénicas; por ejemplo R. rickettsii (el agente de la fiebre manchada de las Montañas Rocosas) y R. prowazekii (el agente del tifus epidemémico). Muchas de las rickettsiosis son prevalentes a lo largo de América Latina; sin embargo, estas enfermedades son desatendidas porque rara vez son consideradas en el diagnóstico diferencial de enfermedades febriles en los trópicos. Esto se explica parcialmente por el hecho de que todas las infecciones causadas por Rickettsia son difíciles de diagnosticar, debido a la presentación clínica no-específica inicial, sospecha clínica ausente, y la falta de pruebas diagnósticas sensibles y específicas que se pueden utilizar durante la presentación aguda. Además, la confusión diagnóstica con infecciones virales es la regla, y esto es un problema crítico ya que estas infecciones pueden tratarse con antibióticos apropiados. Con esta revisión, esperamos contribuir al conocimiento y la conciencia de estas importantes enfermedades dentro de los profesionales científicos y de salud en América Latina.

Descriptores: Rickettsia, patogénesis, inmunidad, vacuna

Abstract

Several species within the genus Rickettsia are highly pathogenic; for example, R. rickettsii (the agent of Rocky Mountain spotted fever) and R. prowazekii (the agent of epidemic typhus). Many of the rickettsioses are prevalent throughout Latin America; however these diseases are neglected because they are seldom considered in the differential diagnosis of febrile diseases in the tropics. This is partly explained by the fact that all infections caused by Rickettsia are difficult to diagnose due to the initial non- specific clinical presentation, absent clinical suspicion, and the lack of sensitive and specific diagnostic tests that can be deployed during the acute presentation. Furthermore, diagnostic confusion with viral infections is the rule, and this is a critical problem because these infections can be treated with appropriate antibiotics. With this review, we expect to contribute to increase knowledge and awareness of these important diseases among scientists and health care professionals in Latin America.

Keywords: Rickettsia, pathogenesis, immunity, vaccine


Rickettsia are the etiologic agents of two of the most lethal infections known to man, Rocky Mountain spotted fever (Rickettsia rickettsii) and epidemic typhus (R. prowazekii). Moreover, epidemic typhus has shaped History due to the massive epidemics that it produced during times of war until World War I.1 Both agents are select agents because of their potential use as bioweapons.2 On the other hand, several new pathogenic Rickettsia have been discovered in the last few decades; new rickettsioses are certainly emerging and old rickettsioses are re-emerging.3

Members of the genus Rickettsia (family Rickettsiaceae, order Rickettsiales) are α-proteobacteria that share the following general characteristics: 1) they have closely related A/T-rich small genomes, a consequence of evolutionary loss of genes encoding proteins that participate in various biosynthetic pathways;4-6 2) they can only survive in the cytoplasm of eukaryotic cells where they obtain needed metabolic substrates that they cannot synthetize themselves (they are strict obligate intracellular parasites); 3) most of the well-known rickettsiae reside within arthropods. Indeed, hematophagous insects and ticks transmit rickettsiae that are pathogenic to humans and other vertebrates (they are zoonoses); 4) in humans, rickettsiae preferentially target endothelial cells, the cells that line vascular and lymphatic vessels (except for Rickettsia akari, the agent of rickettsialpox, which specially targets monocytes and macrophages).7

The transmission of rickettsia by hematophagous arthropod vectors was established early in the 20th century. In 1906 WW King8 and HT Ricketts9 described their experiments with guinea pigs in which they demonstrated that ticks transmit Rocky Mountain spotted fever (RMSF). At the time, Ricketts and others recognized that the clinical presentation of Rocky Mountain spotted fever closely resembled that of epidemic typhus; however, it was not yet known that closely related organisms caused the two diseases. What was clear then was that the human body louse was the vector of typhus.10 Charles Nicolle received the 1928 Nobel Prize for this discovery.

In 1914, H. Plotz reported the identification of a gram-positive bacillus in the blood of patients with typhus as well as their lice.11 H. da Rocha-Lima confirmed these findings in 1916;12 he named the organism Rickettsia prowazekii in honor of Ricketts and Stanislaus von Prowazek, both of whom died of typhus acquired in the course of their investigations. In 1916, SB Wolbach studied samples from guinea pigs with Rocky Mountain spotted fever and identified very small gram-negative organisms in vascular vessels.13,14 Subsequently, in 1917, he confirmed this finding as well as the vascular nature of the infection in autopsies of human patients with Rocky Mountain spotted fever.15 The integration into a single genus, Rickettsia, would not be proposed until 1943.16 By the late 1960s and early 1970s a more modern conception began to be synthetized.17,18

At the present moment, there are 22 entries for Rickettsia genomes in the database of NCBI. They are R. rickettsii, R. prowazekii,19 R. conorii,20 R. typhi, R. massiliae, R. canadensis, R. slovaca, R. bellii, R. africae, R. sibirica, R. peacockii, R. akari, R. felis, R. montanensis, R. rhipicephali, R. australis, R. parkeri, R. philipii, R. japonica, R. heilongjiangensis, Candidatus Rickettsia amblyommii, and Rickettsia endosymbiont of Ixodes scapularis. Based on the analysis of a subset of these data,21,22 new phylogenetic relationships were proposed. Accordingly, there are four groups: 1) the non-pathogenic ancestral group (R. bellii and R. canadensis), which diverged earlier; 2) typhus group (R. typhi and R. prowazekii); 3) spotted fever group (R. rickettsii, R. parkeri, R. conorii, and several others); and 4) transitional group (R. akari, R. australis, and R. felis). A more recent analysis proposes to split the ancestral group in two with one Rickettsia in each group (i.e., R. bellii and R. canadensis) and to include the transitional group within the spotted fever group (SFG).23 According to this new scheme, the SFG group is divided in four subgropus: 1) the R. rickettsii subgroup (R. rickettsii, R. conorii, R. africae, R. parkeri, R. sibirica, R. slovaca, R. honei, R. japonica, R. heilongjiangensis, and a few others); 2) R. massiliae subgroup (R. massiliae , R. montanensis, R. aeschlimannii and R. rhipicephali, R. raoultii and others); 3) R. helvetica subgroup (R. helvetica, R. asiatica, R. tamurae, R. monacensis); 4) R. akari subgroup (R. akari, R. australis, and R. felis). A phenotypic characteristic of the R. rickettsii subgroup is its susceptibility to rifampin, while the R. massiliae subgroup is resistant to this antibiotic.24 For a long time, the serological response was the main criterion used to classify rickettsiae in only two groups,25,26 spotted fever and typhus; using those criteria, R. canadensis was included in the typhus group at that time. Also, until 1995,27 Orientia tsutsugamushi, the etiologic agent of scrub typhus, was included in the genus Rickettsia (i.e., Rickettsia tsutsugamushi) and considered a third group.

In temperate regions of the globe, the seasonality of SFG rickettsioses is explained by the activity of the tick vectors, particularly the adults, which are more active during the spring and early summer. There is also a periodicity in a timeframe of decades that has not been appropriately explained yet. It is possible that climate change may affect the behavior of tick vectors.28 One of the recent peaks of reporting of Rocky Mountain spotted fever (RMSF) occurred during the early 2000s.29 This may be related to increased disease activity but also to renewed interest not only in the United States but also throughout the Americas (RMSF occurs only in the Americas). The disease has now been documented in almost all countries of Latin America.30-40 Even more importantly, new SFG rickettsioses have been discovered. For instance, R. parkerii, which was considered a non-pathogenic Rickettsia for a very long time, was recently shown to produce a mild spotted fever with an eschar and local lymphadenopathy.41-44 Other recently described Rickettsia associated with eschars and relatively mild disease include Rickettsia 364D45 and R. massiliae.46,47

One of the consequences of the non-specific initial febrile syndrome and the lack of commercially available diagnostic methods that are sensitive and specific during the acute presentation of the rickettsioses is that the disease is frequently underreported and diagnosed as a viral illness.48 In Latin America, the umbrella diagnosis of dengue is frequently applied to cases of rickettsiosis.49

Pathogenesis

Rickettsioses are systemic febrile diseases that affect individuals of any age independently of their immune status.48,50-53 Although the pathogenetic mechanisms are shared, not all rickettsioses are equally severe, which is explained by differences in virulence of the individual species and vector-related factors.

The entry of Rickettsia into host cells is an active process that requires energy from both the host and the rickettsiae.54 There is evidence that rickettsiae use surface cell antigen 0 (sca0 or rOmpA)55 and sca 156 to attach to target cells (these and the other rickettsial sca proteins are autotransporters). Subsequent to attachment, which is mostly a passive process, endocytosis of rickettsia is actively triggered when the rickettsial outer membrane protein B (rOmpB or sca5) binds to the host cell membrane form of Ku70.57 Since blocking of this interaction only inhibits about 50% of rickettsial entry, other ligands and receptors must be present; sca258 and adr259 appear to be some of those bacterial ligands.

The necessary cytoskeletal rearrangements that produce the zipper-like entry mechanism of Rickettsia spp. involve multiple host pathways that activate the Arp2/3 complex60 with the participation of Cdc42, cofilin, c-Cbl, clathrin, and caveolin 2.61 Rickettsia may also enter phagocytic cells such as monocytes and macrophages (which are a secondary target of most Rickettsia) by antibody-mediated opsonization.62 Within a short period of time after endocytosis, rickettsia escapes into the cytosol. The rickettsial genes pld, which encodes an enzyme with phospholipase D activity,63 and tlyc, which encodes a hemolysin64 are believed to be effectors of this function. This conclusion is based on the ability of the normally vacuolar Salmonella enterica to escape into the cytosol when it expresses rickettsial tlyc or pld.65 In addition, rickettsial proteins with phospholipase A activity were confirmed66, 67 but only in the typhus group Rickettsia. That activity underlies the phenomenon of hemolysis produced by these rickettsiae in vitro.68,69

Once Rickettsia escapes the phagocytic vacuole, it acquires multiple metabolic substrates from the host cytoplasm. The availability of those substrates allowed genome reduction through loss of many genes including, among many others, those for nucleotide synthesis and enzymes for sugar metabolism.70 Multiple transporters of substrates from the host cytoplasm, including ATP,71 compensated for these gene losses.72 The mechanisms of transport are active and include the use of the transmembrane electrical potential.73

Typhus group Rickettsia grow until they burst the host cell74 while spotted fever group Rickettsia rapidly spread from cell to cell75 due to their actin propulsion. Of course, host cells are damaged in the process;76 the mechanisms may involve the production of free radicals77,78 and phospholipase activity.79 On the other hand, there is experimental evidence that rickettsiae can maintain their cellular niche through inhibition of apoptosis,80 and that pathogenic Rickettsia can inhibit autophagy.81

The main target cells of most Rickettsia, with the exception of R. akari are endothelial cells, the cells that line all vascular vessels in the body. These cells have important regulatory functions in angiogenesis, hemostasis, permeability and solute exchange, vascular tone, and inflammation.82-84 Thus, their targeting by rickettsiae explains many of the clinical features of the diseases including systemic involvement and leakage of intravascular fluid. Rickettsial infection of endothelial cells induces cellular damage leading to detachment. Those infected endothelial cells circulate in the blood85,86 and are likely to be the source of new foci of infection once they lodge in distal capillaries.

Several mechanisms are likely to contribute to the increased vascular permeability observed in clinical cases. They include production of vasoactive prostaglandins as a consequence of increased expression of COX-2,87 endothelial production of nitric oxide,88 effects of inflammatory cells and their mediators,89 and endothelial detachment and denudation of vessels. Such damage may be caused by phospholipase activity,79 mechanical damage to the membrane caused by exiting rickettsiae under actin propulsion,90 or lipid peroxidation of the cell membrane.76,77,91,92 The most severe clinical presentations are a consequence of endothelial damage in the lungs and brain and include noncardiogenic pulmonary edema, interstitial pneumonia, adult respiratory distress syndrome, meningoencephalitis, seizures, and coma;93-97 involvement of these organs explains the majority of the mortality, which is observed particularly with Rocky Mountain spotted fever and epidemic typhus (the reported mortality without antibiotics ranges from 10 to 60%). However, it should be emphasized that reliance on serological methods for diagnostic confirmation may lead to underestimation the actual case-fatality rate. This was well illustrated in a recent report of nine fatal cases with negative serological results that were confirmed by immunohistochemical demonstration of the antigen in tissues.51 At the other end of the clinical spectrum are several rickettsioses; murine typhus, with a mortality of less than 2%, is the most important of them because of its global distribution.98

Although multiple coagulation abnormalities have been described during the course of clinical and experimental rickettsiosis,99 disseminated intravascular coagulation occurs only rarely in lethal cases and is not a common feature of rickettsiosis.100

The cells that are infected immediately after inoculation have not been identified. Many of the rickettsiae that result in less severe disease also produce an eschar (area of necrosis with a rich inflammatory infiltrate and local rickettsial proliferation) at the bite site.101 When an eschar is present, another frequent clinical finding is local lymphadenitis, suggesting initial spread through lymphatics. Rocky Mountain spotted fever, the most severe of the spotted fever rickettsioses, does not manifest with an eschar or local lymphadenitis. This could be due to a more rapid hematogenous dissemination.

The recommended antibiotic treatment for all rickettsioses is doxycycline.102 This antibiotic has the advantage of covering other tick-borne bacterial infections. Rickettsiae are resistant to many antibiotics.103 Other antibiotics, including chloramphenicol and fluoroquinolones may be effective, although there is evidence that they may have deleterious effects.104,105 The antibiotic resistance of Rickettsia combined with the non-specific initial clinical presentation and lack of commercially available laboratory tests for confirmatory diagnosis during the acute presentation, lead to delayed diagnosis and inappropriate treatment; the consequence is excessive mortality.104

Rickettsial virulence

Many rickettsial genes have been predicted to participate in virulence based on bioinformatics analyzes;72 several toxin-antitoxin systems are examples. One of them, encoded by the vapB/C genes was shown to be functional; E. coli transformed with rickettsial vapC significantly decrease their growth, while VapB formed a complex with VapC to inhibit its RNase activity.106 More importantly, microinjection of VapC to mammalian cells induced apoptotic death.

A large number of intracellular bacteria use type IV secretion systems to inject proteins into the host in order to produce a favorable niche. Interestingly, genomic analysis showed that multiple genes with the potential to encode a reduced type IV secretion system are conserved in Rickettsia.107 Whether the system is actually functional or not remains to be tested.

The phospholipase D encoded by the gene pld, a likely mediator of phagosomal escape, is a virulence factor as suggested by the milder disease produced in guinea pigs infected with R. prowazekii with a mutated pld.108 This study used homologous recombination for targeted knockout of a rickettsial gene. Previous studies using the difficult techniques of genetic manipulation of Rickettsia, including transposon-mediated mutagenesis, indicated that mutation of the open reading frames (ORFs) 243, 294, and 689 of R. prowazekii do not produce an observable phenotypic difference.109 Thus, these genes may be non-essential genes (at least for growth in mouse cell line in vitro). Also, R. rickettsii mutants lacking expression of sca2, which participates in actin polymerization, do not cause apparent illness in guinea pigs.110

Loss of regulation due to genome decay has also been proposed as a mechanism of increased virulence;111 however, this argument does not explain why R. rickettsii and R. prowazekii are almost equally pathogenic and the radical difference in virulence between the two typhus group rickettsiae, R typhi and R. prowazekii.

In the absence of genetic approaches that work well and consistently for Rickettsia, other methods have been introduced to identify virulence factors. One example is the comparison of the genomes of closely related Rickettsia with different pathogenicity. The Dermacentor andersoni endosymbiont R. peacockii was compared to virulent R. rickettsii; it was found that it had a plasmid, multiple transposons with intact transposase sequences, and many deletions, nonsense mutations, and split genes.112 The authors proposed that some of the absent or mutated genes in R. peacockii might explain the lack of pathogenicity. Those genes include DsbA (a catalyzer of disulfide bond formation), RickA, Sca0, Sca1, a gene encoding Protease II, and a gene encoding a putative phosphoethanolamine transferase that could play a role in the formation of the prominent slime layer found in the pathogenic spotted fever-group rickettsiae. Interestingly, the hypothetical protein A1G_05165 of a virulent strain of R. rickettsii (strain Sheila Smith) is deleted in R. peacockii and it is also not present in other non-pathogenic rickettsiae. This hypothetical protein has ankyrin repeats; similar proteins in other members of this order (i.e., Anaplasma) appear to play a role in virulence through binding of host DNA and altered host gene regulation. A1G_05165 is also mutated in a non-pathogenic strain of R. rickettsii (strain Iowa). In addition, the genomic study that compared the pathogenic strains R and Sheila Smith with strain Iowa also found 23 deletions within predicted ORFs of R. rickettsii Sheila Smith and 24 deletions within predicted ORFs of R. rickettsii Iowa.113 One of the genes deleted in R. rickettsii Iowa is the adhesin rOmpA (sca0). Also, rompB has four single nucleotide polymorphisms (SNPs) that may explain the defective processing of this important membrane protein in strain Iowa.114 Finally, it should be emphasized that there is a good opportunity to understand virulence by comparing the genomes, transcriptomes, and proteomes of the two typhus group Rickettsia since they have very closely related genomes but very different virulence in humans, with R. prowazekii producing a much more severe infection (epidemic typhus) than R. typhi (murine or endemic typhus).

Another system to study the physiology of Rickettsia in the absence of more efficient genetic systems is the use of E. coli-based assays. For example, to identify proteins transported out of the rickettsial cytoplasm, bioinformatic tools were used to uncover predicted secreted proteins (based on the presence of N-terminal signal peptides). The signal peptides of those proteins from R. typhi were then fused to the E. coli alkaline phosphatase phoA gene (lacking an intrinsic signal peptide sequence) to test if those signal peptides provided information to translocate PhoA into the periplasm of E. coli.115 Eighty-four functional signal peptides were identified suggesting that those rickettsial proteins might be secreted using the rickettsial Sec system. Those proteins include sca1-3, sca5, Pld, and proteins that are believed to be part of a type IV secretion system.

Immunity and vaccines

An often overlooked but critical factor in the pathogenesis of rickettsial diseases is the transmission by arthropod vectors because their saliva is not a passive vehicle for transmission.116-118 In fact, the tick saliva modifies the host environment in order to successfully complete the blood feeding, which occurs during extended periods (several days for nymph and adult ticks). Proteins in the tick saliva modulate host hemostasis, innate and adative immuntiy, complement activation,119 angiogenesis, and extracellular matrix regulation. 120,121. Evidently, all of those factors could determine the final outcome of the infection. Furthermore, tick saliva can modulate the physiology of endothelial cells, the main target cells of Rickettsia. For example, salivary gland extracts from D. andersoni reduce the upregulation of ICAM-1 induced by TNF-α on a mouse endothelial cell line.122 This change could contribute to reduce the migration of leukocytes into tick bite sites.

Endothelial cells are not passive actors in the anti-rickettsial immune response. Upon rickettsial infection, the transcription factor NFκB (a critical stimulating factor of the immune system) becomes activated in endothelial cells.123-125Other critical signaling mediators become activated as well. They include STAT1, STAT3,126 and p38 MAPK.127-129 As a consequence of the activation of these various signaling systems, endothelial cells respond by expressing a variety of chemokines,130,131 cytokines such as IL-1 α, and IL-6,132,133 adhesion molecules such as E-selectin, VCAM-1, ICAM-1,134-136 and αVβ3 integrin,137 and secretion of prostanoids.87,138

NK cells are early producers of IFN-γ after infection with Rickettsia.139,140 This cytokine is important because, together with TNF-α, it activates the bactericidal functions of the endothelium.141,142 Those functions are performed in part through expression of indoleamine-2,3-dioxygenase (IDO), which leads to tryptophan starvation.143 Animal studies have demonstrated the importance of a T helper 1 (Th1) response in effective immunity against rickettsiae144 with a particularly important role for CD8+ T cells.145,146 In fact, T cells are sufficient to mediate protection against a lethal rickettsial challenge, even in the context of a heterologous challenge where anti-typhus group T cells protect against a lethal challenge with SFG Rickettsia and vice versa.147

Despite the fact that rickettsiae are intracellular parasites and that cellular adaptive immunity is critical during a primary infection, there is clear evidence that the humoral immune response is very important in preventing the development of disease during secondary infections or after a lethal challenge following passive serum transfer. In fact, it was Ricketts himself who demonstrated this fact.148 The anti-rickettsial humoral immune response is cross-reactive within rickettsiae of the same group but not across groups (e.g., between typhus and SFG groups).149,150 The most abundant surface protein of Rickettsia is rOmpB (Sca5), which is an autotransporter. It is an immunodominant protein and antibodies against it are protective.151

Inactivated vaccines for R. rickettsii and R. prowazekii were produced early from a variety of sources including their vectors but they were very reactogenic and protection was incomplete. Later on, inactivated vaccines were produced from Rickettsia cultivated in eggs but antigenicity was variable and protection was poor. 152-155 In the 1950s a very effective vaccine for epidemic typhus was produced. It was an attenuated strain denominated Madrid E; 156 however, spontaneous reversion to a virulent phenotype precluded further development and testing. 157,158
We now know that the attenuation is explained, at least in part, by a point mutation in the gene encoding a S-adenosulmethionine-dependent methyltransferase.159 Given the nature of the mutation, it is not surprising that reversion was not an uncommon occurrence. Deletion of the entire gene would permit the production of a safer vaccine. Alternatively, strains with multiple genetic differences could prove to be safe vaccines. In this regard, it is interesting to note that the strain Iowa of R. rickettsii, which is attenuated and has multiple genetic differences when compared with virulent strains, can protect guinea pigs against a challenge with virulent R. rickettsii.113

Other recent efforts have focused on the production of a subunit vaccine. Fragments of rickettsial proteins that may trigger protective immunity were tested. They included rOmpA160,161 and rOmpB162-164 and results were encouraging; however, these approaches are limited and biased because of their focus on proteins that elicit a strong humoral response. A major effort for identification of immunogenic antigens is clearly needed, and the antigen discovery effort will need new tools to identify relevant conserved antigens recognized by T cells.

It will be possible to produce vaccines that cover more than one species of Rickettsia given the evidence of cross-protective immunity within the typhus or spotted fever groups165-171 or even across groups.147 The production of an effective anti-Rickettsia vaccine is a public health priority for several reasons. Firstly, some rickettsioses are highly lethal not only to humans but also to companion animals (i.e., dogs). Secondly, clinical diagnosis of rickettsioses is very difficult due to the non-specific initial clinical presentation. Thirdly, there are no commercially available diagnostic tests that can be used during the acute stage when antibiotic intervention is helpful.

The contemporary development of a vaccine has two initial essential aspects, namely identification of the relevant antigens and definition of immunological correlates of protection to guide the selection of vehicles, vectors, schedules, and adjuvants. In the case of infections caused by Rickettsia, due to the availability of excellent murine models, relevant correlates of protective immunity can be derived from the characterization of experimental infections because animals (as well as humans) that survive the infection become solidly immune to reinfection.

In regard to immunological correlates of protection, the magnitude of a response assessed by a single parameter (e.g., IFN-γ for intracellular pathogens, as frequently reported), is not enough. Now we know that there is functional heterogeneity of the T cell effector responses (including cytokine secretion, cytolytic activity, and development of various memory phenotypes) and that there are particular subsets of T cells, which express unique combinations of effector functions, that are more protective.172-174 We probably should approach the definition of correlates of protective immunity in a way that parallels the complexity of physiological immunity, which is a multifaceted and integrated response that includes many different cells, receptors, ligands, and signaling modules that function in a combinatorial mode. For infections in which cellular immunity plays a predominant role, there is evidence from experimental models that multifunctional T cells are the best correlate of protection described thus far. More importantly, this has been demonstrated in humans as well.175-177

The technologies for understanding the integrated functioning of the immune system are now available and accessible. Those are the tools of Systems Biology, the “omics” methods and bioinformatics tools that permit the analysis of complex interactions in biological systems through the investigation of massively parallel data acquired from each experimental condition.178 The application of Systems Biology to vaccinology is already identifying transcriptional signatures of protective immune responses that include sub-signatures of appropriate innate and adaptive responses.179 Moreover, early predictive signatures of appropriate adaptive immune responses immediately after vaccination have been defined and verified using the Yellow fever (17D) vaccine as a model.180 It is expected that such knowledge will provide paradigms for the development of novel vaccines for which limited data from humans is currently available. That is certainly the case for infections caused by Rickettsia because it is unlikely that we will be able to collect sufficient human samples from clinical cases with diverse outcomes in order to define broad signatures of protective immunity. A promising solution to this problem is to use our current understanding of well-known effective immune responses as guiding principles. The study of the response to two of the most successful human vaccines in history, the yellow fever vaccine177,181 and the smallpox vaccine,176 is likely to yield relevant paradigms that we could use as guiding posts in rickettsiology.

From the perspective of antigen identification for vaccine development, until recently it was almost exclusively biased towards the humoral immune response. This bias was partly due to the effectiveness of antibodies in protection against almost all of the currently approved vaccines for human use, the relative technical simplicity of working with serum and antibodies, and the methodical challenges of working with T-cells. Presently, the barriers to identify potent vaccine antigens recognized by T-cells need to be addressed because most of the vaccines that remain to be produced require a strong T-cell component to afford significant protection. In particular, there is an urgent need to develop appropriate techniques to identify antigens recognized by T-lymphocytes because antigen discovery is the most important aspect of any vaccine development project; without appropriate antigens, a vaccine is unlikely to succeed.

Given the evidence that CD4+T cells and CD8+T cells target different antigens,182 it is clear that antibody-based screening methods are not suitable to identify antigens recognized by CD4+T cells or, particularly, CD8+T cells. Several approaches to more directly identify antigens recognized by T-cells have been used; many of them rely on Reverse Vaccinology, a branch of Systems Biology that analyses entire microbial genomes to predict immunogenic proteins based on predefined rules derived from the analysis of large empirical datasets.183 On the other hand, the predicting power of those immunoinformatic strategies has not been thoroughly tested by direct experimentation. Moreover, at least for bacterial proteins, known protective antigens actually have less predicted epitopes than randomly selected bacterial protein sets used as a control.184

Empirical methods for identification of antigens recognized by T-lymphocytes rely on T-cells from animals or individuals that are immune to the pathogen. Those memory T-cells had been selected during the physiological immune response to persist and recognize a limited number of antigens (i.e., immunodominant antigens). Thus, methods that use memory T-cells for antigen identification are more likely to miss potentially protective subdominant antigens. One strategy for T-cell antigen identification that is not biased towards immunodominant antigens is genomic immunization or Expression Library Immunization (ELI).185 In this technique, pools of eukaryotic expression vectors with cloned pathogen genes are used to directly immunize animals. The animals are then challenged with lethal doses of the microbial pathogen. The gene pools that trigger protection are subsequently deconvoluted by testing each component of the pool one at a time. This method allows the priming of naïve T-cells by the expressed cloned microbial genes regardless of whether they are subdominant or dominant during a natural infection as long as the appropriate T-cell receptors are present. Although ELI has been successfully used,186 it has its own problems as it relies on a DNA immunization strategy; thus, antigen expression is not guaranteed in all cases. Accordingly, it is not possible to know which pathogen genes were not screened validly; a negative response can be due to lack of an immunological response or to failed expression of the microbial gene.

As an alternative, we produced a new in vivo screening platform; the idea is to easily produce antigen presenting cells (APCs) expressing individual open reading frames (ORFs) from any sequenced Rickettsia and use them for immunization of naive mice. Immunization with pooled APCs containing 4 to 5 rickettsial ORFs is followed by challenge with live virulent pathogen and measurement of an indicator of protection such as decreased bacterial load. Once protective pools are identified, each member of the pool is tested individually to identify ORF(s) responsible for a protective immune response. With this platform, one can easily test for cross-protective responses by immunizing with the ORFs of one species of Rickettsia and challenging with another. Importantly, the proposed methodology is not biased by immunodominance because T cells from immune animals are not used to select antigens. This aspect is potentially important for vaccine development because subdominant or cryptic antigens have been shown to elicit protective immune responses in other systems.187-189 The ability of our platform to discover relevant antigens for vaccine development independently of their ranking in the natural hierarchy of immunodominance dramatically expands the universe of possible antigens; thus, this platform offers a possible solution to the identification of protective antigens that are conserved among different strains of a microbe or even different species within a genus.


References

1.Raoult D, Woodward T, Dumler JS. The history of epidemic typhus. Infect Dis Clin North Am 2004; 18:127-140.         [ Links ]

2.Azad AF. Pathogenic rickettsiae as bioterrorism agents. Clin Infect Dis 2007; 45 Suppl 1:S52-55.         [ Links ]

3.Parola P, Paddock CD, Raoult D. Tick-borne rickettsioses around the world: emerging diseases challenging old concepts. Clin Microbiol Rev 2005; 18:719-756.         [ Links ]

4.Fuxelius HH, Darby A, Min CK, Cho NH, Andersson SG. The genomic and metabolic diversity of Rickettsia. Res Microbiol 2007; 158:745-753.         [ Links ]

5.Darby AC, Cho NH, Fuxelius HH, Westberg J, Andersson SG. Intracellular pathogens go extreme: genome evolution in the Rickettsiales. Trends Genet 2007; 23:511-520.         [ Links ]

6.Blanc G, Ogata H, Robert C, Audic S, Suhre K, Vestris G, et al. Reductive genome evolution from the mother of Rickettsia. PLoS Genet 2007; 3:e14.         [ Links ]

7.Valbuena G, Walker DH. Infection of the endothelium by members of the order Rickettsiales. Thromb Haemost 2009; 102:1071-1079.         [ Links ]

8.King WW. Experimental transmission of Rocky Mountain spotted fever by means of the tick. Public Health Rep 1906; 72:863-864.         [ Links ]

9.Ricketts HT. The transmission of Rocky Mountain spotted fever by the bite of the wood-tick (Dermacentor occidentalis). JAMA 1906; 47:358.         [ Links ]

10.Gross L. How Charles Nicolle of the Pasteur Institute discovered that epidemic typhus is transmitted by lice: reminiscences from my years at the Pasteur Institute in Paris. Proc Natl Acad Sci USA 1996; 93:10539-10540.         [ Links ]

11.Plotz H. The etiology of typhus fever (and of Brill’s disease). Preliminary communication. JAMA 1914; 62:1556.         [ Links ]

12.da Rocha-Lima H. On the etiology of typhus fever. En: Hahon N, ed. Selected papers on the pathogenic rickettsiae. Boston: Harvard University Press, 1968: 74-78.         [ Links ]

13.Wolbach SB. The etiology of Rocky Mountain spotted fever. Occurrence of the parasite in the tick. J Med Res 1916; 35:147-150.         [ Links ]

14.Wolbach SB. The etiology of Rocky Mountain spotted fever. A preliminary report. J Med Res 1916; 34:121-126.         [ Links ]

15.Wolbach SB. The etiology and pathology of Rocky Mountain spotted fever. Third preliminary report. J Med Res 1917; 37:499-508.         [ Links ]

16.Philip CB. Nomenclature of the pathogenic rickettsiae. Am J Hygiene 1943; 37:301-309.         [ Links ]

17.Ormsbee RA. Rickettsiae (as organisms). Annu Rev Microbiol 1969; 23:275-292.         [ Links ]

18.Weiss E. Growth and physiology of rickettsiae. Microbiol Mol Biol Rev 1973; 37:259.         [ Links ]

19.Andersson SG, Zomorodipour A, Andersson JO, Sicheritz-Pontén T, Alsmark UC, Podowski RM, et al. The genome sequence of Rickettsia prowazekii and the origin of mitochondria. Nature 1998; 396:133-140.         [ Links ]

20.H, Audic S, Renesto-Audiffren P, Fournier PE, Barbe V, Samson D, et al. Mechanisms of evolution in Rickettsia conorii and R. prowazekii. Science 2001; 293:2093-2098.         [ Links ]

21.Gillespie JJ, Beier MS, Rahman MS, Ammerman NC, Shallom JM, Purkayastha A, et al. Plasmids and rickettsial evolution: insight from Rickettsia felis. PLoS One 2007; 2:e266.         [ Links ]

22.Gillespie JJ, Williams K, Shukla M, Snyder EE, Nordberg EK, Ceraul SM, et. al. Rickettsia phylogenomics: unwinding the intricacies of obligate intracellular life. PLoS One 2008; 3:e2018.         [ Links ]

23.Merhej V, Raoult D. Rickettsial evolution in the light of comparative genomics. Biol Rev Camb Philos Soc 2011; 86:379-405.         [ Links ]

24.Blanc G, Ogata H, Robert C, Audic S, Claverie JM, Raoult D. Lateral gene transfer between obligate intracellular bacteria: evidence from the Rickettsia massiliae genome. Genome Res 2007; 17:1657-1664.         [ Links ]

25.Philip RN, Casper EA, Burgdorfer W, Gerloff RK, Hughes LE, Bell EJ. Serologic typing of rickettsiae of the spotted fever group by microimmunofluorescence. J Immunol 1978; 121:1961-1968.         [ Links ]

26.Ormsbee R, Peacock M, Philip R, Casper E, Plorde J, Gabre-Kidan T, et al. Antigenic relationships between the typhus and spotted fever groups of rickettsiae. Am J Epidemiol 1978;108:53-59.         [ Links ]

27.Tamura A, Ohashi N, Urakami H, Miyamura S. Classification of Rickettsia tsutsugamushi in a new genus, Orientia gen. nov., as Orientia tsutsugamushi comb. nov. Int J Syst Bacteriol 1995; 45:589-591.         [ Links ]

28.Parola P, Socolovschi C, Jeanjean L, Bitam I, Fournier PE, Sotto A, et al. Warmer weather linked to tick attack and emergence of severe rickettsioses. PLoS Negl Trop Dis 2008; 2:e338.         [ Links ]

29.Openshaw JJ, Swerdlow DL, Krebs JW, Holman RC, Mandel E, Harvey A, et al. Rocky mountain spotted fever in the United States, 2000-2007: interpreting contemporary increases in incidence. Am J Trop Med Hyg 2010; 83:174-182.         [ Links ]

30.Peacock MG, Ormsbee RA, Johnson KM. Rickettsioses of Central America. Am J Trop Med Hyg 1971; 20:941-949.         [ Links ]

31.Ripoll CM, Remondegui CE, Ordonez G, Arazamendi R, Fusaro H, Hyman MJ, et al. Evidence of rickettsial spotted fever and ehrlichial infections in a subtropical territory of Jujuy, Argentina. Am J Trop Med Hyg 1999; 61:350-354.         [ Links ]

32.Zavala-Velazquez JE, Ruiz-Sosa J, Vado-Solis I, Billings AN, Walker DH. Serologic study of the prevalence of rickettsiosis in Yucatán: evidence for a prevalent spotted fever group rickettsiosis. Am J Trop Med Hyg 1999; 61:405-408.         [ Links ]

33.de Lemos ER, Alvarenga FB, Cintra ML, Ramos MC, Paddock CD, Ferebee TL, et al. Spotted fever in Brazil: a seroepidemiological study and description of clinical cases in an endemic area in the state of São Paulo. Am J Trop Med Hyg 2001; 65:329-334.         [ Links ]

34.Blair PJ, Schoeler GB, Moron C, Anaya E, Caceda R, Cespedes M, et al. Evidence of rickettsial and leptospira infections in Andean northern Peru. Am J Trop Med Hyg 2004; 70:357-363.         [ Links ]

35.Hidalgo M, Sánchez R, Orejuela L, Hernández J, Walker DH, Valbuena G. Prevalence of antibodies against spotted fever group rickettsiae in a rural area of Colombia. Am J Trop Med Hyg 2007; 77:378-380.         [ Links ]

36.Paddock CD, Fernandez S, Echenique GA, Sumner JW, Reeves WK, Zaki SR, et al. Rocky Mountain spotted fever in Argentina. Am J Trop Med Hyg 2008; 78:687-692.         [ Links ]

37.Zavala-Castro JE, Dzul-Rosado KR, León JJ, Walker DH, Zavala-Velázquez JE. An increase in human cases of spotted fever rickettsiosis in Yucatan, Mexico, involving children. Am J Trop Med Hyg 2008; 79:907-910.         [ Links ]

38.Argüello AP, Hun L, Rivera P, Taylor L. A fatal urban case of rocky mountain spotted Fever presenting an eschar in San Jose, Costa Rica. Am J Trop Med Hyg 2012; 87:345-348.         [ Links ]

39.Troyo A, Alvarez D, Taylor L, Abdalla G, Calderón-Arguedas O, Zambrano ML, et al. Rickettsia felis in Ctenocephalides felis from Guatemala and Costa Rica. Am J Trop Med Hyg 2012; 86:1054-1056.         [ Links ]

40.Hidalgo M, Montoya V, Martínez A, Mercado M, De la Ossa A, Vélez C, et al. Flea-Borne Rickettsioses in the North of Caldas Province, Colombia. Vector Borne Zoonotic Dis 2013; 13:289-294.         [ Links ]

41.Paddock CD, Sumner JW, Comer JA, Zaki SR, Goldsmith CS, Goddard J, et al. Rickettsia parkeri: a newly recognized cause of spotted fever rickettsiosis in the United States. Clin Infect Dis 2004; 38:805-811.         [ Links ]

42.Paddock CD. Rickettsia parkeri as a paradigm for multiple causes of tick-borne spotted fever in the western hemisphere. Ann N Y Acad Sci 2005; 1063:315-326.         [ Links ]

43.Whitman TJ, Richards AL, Paddock CD, Tamminga CL, Sniezek PJ, Jiang J, et al. Rickettsia parkeri infection after tick bite, Virginia. Emerg Infect Dis 2007; 13:334-336.         [ Links ]

44.Paddock CD, Finley RW, Wright CS, Robinson HN, Schrodt BJ, Lane CC, et al. Rickettsia parkeri rickettsiosis and its clinical distinction from Rocky Mountain spotted fever. Clin Infect Dis 2008; 47:1188-1196.         [ Links ]

45.Shapiro MR, Fritz CL, Tait K, Paddock CD, Nicholson WL, Abramowicz KF, et al. Rickettsia 364D: a newly recognized cause of eschar-associated illness in California. Clin Infect Dis 2010; 50:541-548.         [ Links ]

46.Vitale G, Mansuelo S, Rolain JM, Raoult D. Rickettsia massiliae human isolation. Emerg Infect Dis 2006; 12:174-175.         [ Links ]

47.García-García JC, Portillo A, Núñez MJ, Santibáñez S, Castro B, Oteo JA. A patient from Argentina infected with Rickettsia massiliae. Am J Trop Med Hyg 2010; 82:691-692.         [ Links ]

48.Paddock CD, Holman RC, Krebs JW, Childs JE. Assessing the magnitude of fatal Rocky Mountain spotted fever in the United States: comparison of two national data sources. Am J Trop Med Hyg 2002; 67:349-354.         [ Links ]

49.Zavala-Velazquez JE, Yu XJ, Walker DH. Unrecognized spotted fever group rickettsiosis masquerading as dengue fever in Mexico. Am J Trop Med Hyg 1996; 55:157-159.         [ Links ]

50.Centers for Disease Control and Prevention (CDC). Fatal cases of Rocky Mountain spotted fever in family clusters--three states, 2003. MMWR Morb Mortal Wkly Rep 2004; 53:407-410.         [ Links ]

51.Paddock CD, Greer PW, Ferebee TL, Singleton J, McKechnie DB, Treadwell TA, et al. Hidden mortality attributable to Rocky Mountain spotted fever: immunohistochemical detection of fatal, serologically unconfirmed disease. J Infect Dis 1999; 179:1469-1476.         [ Links ]

52.Lee N, Ip M, Wong B, Lui G, Tsang OT, Lai JY, et al. Risk factors associated with life-threatening rickettsial infections. Am J Trop Med Hyg 2008; 78:973-978.         [ Links ]

53.Lee CS, Hwang JH, Lee HB, Kwon KS. Risk factors leading to fatal outcome in scrub typhus patients. Am J Trop Med Hyg 2009; 81:484-488.         [ Links ]

54.Walker TS, Winkler HH. Penetration of cultured mouse fibroblasts (L cells) by Rickettsia prowazeki. Infect Immun 1978; 22:200-208.         [ Links ]

55.Li H, Walker DH. rOmpA is a critical protein for the adhesion of Rickettsia rickettsii to host cells. Microb Pathog 1998; 24:289-298.         [ Links ]

56.Riley SP, Goh KC, Hermanas TM, Cardwell MM, Chan YG, Martinez JJ. The Rickettsia conorii autotransporter protein Sca1 promotes adherence to nonphagocytic mammalian cells. Infect Immun 2010; 78:1895-1904.         [ Links ]

57.Martinez JJ, Seveau S, Veiga E, Matsuyama S, Cossart P. Ku70, a component of DNA-dependent protein kinase, is a mammalian receptor for Rickettsia conorii. Cell 2005; 123:1013-1023.         [ Links ]

58.Cardwell MM, Martinez JJ. The Sca2 autotransporter protein from Rickettsia conorii is sufficient to mediate adherence to and invasion of cultured mammalian cells. Infect Immun 2009; 77:5272-5280.         [ Links ]

59.Vellaiswamy M, Kowalczewska M, Merhej V, Nappez C, Vincentelli R, Renesto P, et al. Characterization of rickettsial adhesin Adr2 belonging to a new group of adhesins in α-proteobacteria. Microb Pathog 2011; 50:233-242.         [ Links ]

60.Martinez JJ, Cossart P. Early signaling events involved in the entry of Rickettsia conorii into mammalian cells. J Cell Sci 2004; 117:5097-5106.         [ Links ]

61.Chan YG, Cardwell MM, Hermanas TM, Uchiyama T, Martinez JJ. Rickettsial outer-membrane protein B (rOmpB) mediates bacterial invasion through Ku70 in an actin, c-Cbl, clathrin and caveolin 2-dependent manner. Cell Microbiol 2009; 11:629-644.         [ Links ]

62.Feng HM, Whitworth T, Popov V, Walker DH. Effect of antibody on the rickettsia-host cell interaction. Infect Immun 2004; 72:3524-3530.         [ Links ]

63.P, Dehoux P, Gouin E, Touqui L, Cossart P, Raoult D. Identification and characterization of a phospholipase D-superfamily gene in rickettsiae. J Infect Dis 2003; 188:1276-1283.         [ Links ]

64.Radulovic S, Troyer JM, Beier MS, Lau AO, Azad AF. Identification and molecular analysis of the gene encoding Rickettsia typhi hemolysin. Infect Immun 1999; 67:6104-6108.         [ Links ]

65.Whitworth T, Popov VL, Yu XJ, Walker DH, Bouyer DH. Expression of the Rickettsia prowazekii pld or tlyC gene in Salmonella enterica serovar Typhimurium mediates phagosomal escape. Infect Immun 2005; 73:6668-6673.         [ Links ]

66.Housley NA, Winkler HH, Audia JP. The Rickettsia prowazekii ExoU Homologue Possesses Phospholipase A1 (PLA1), PLA2, and lyso-PLA2 Activities and can Function in the Absence of Any Eukaryotic Co-Factors in vitro. J Bacteriol 2011; 193:4634-4642.         [ Links ]

67.Rahman MS, Ammerman NC, Sears KT, Ceraul SM, Azad AF. Functional characterization of a phospholipase A(2) homolog from Rickettsia typhi. J Bacteriol 2010; 192:3294-3303.         [ Links ]

68.Clarke DH, Fox JP. The phenomenon of in vitro hemolysis produced by the rickettsiae of typhus fever, with a note on the mechanism of rickettsial toxicity in mice. J Exp Med 1948; 88:25-41.         [ Links ]

69.Ramm LE, Winkler HH. Rickettsial hemolysis: adsorption of rickettsiae to erythrocytes. Infect Immun 1973; 7:93-99.         [ Links ]

70.Walker DH, Yu XJ. Progress in rickettsial genome analysis from pioneering of Rickettsia prowazekii to the recent Rickettsia typhi. Ann N Y Acad Sci 2005; 1063:13-25.         [ Links ]

71.Winkler HH. Rickettsial permeability. An ADP-ATP transport system. J Biol Chem 1976; 251:389-396.         [ Links ]

72.McLeod MP, Qin X, Karpathy SE, Gioia J, Highlander SK, Fox GE, et al. Complete genome sequence of Rickettsia typhi and comparison with sequences of other rickettsiae. J Bacteriol 2004; 186:5842-5855.         [ Links ]

73.Zahorchak RJ, Winkler HH. Transmembrane electrical potential in Rickettsia prowazekii and its relationship to lysine transport. J Bacteriol 1983; 153:665-671.         [ Links ]

74.Wisseman CL, Waddell AD, Silverman DJ. In vitro studies on Rickettsia-host cell interactions: lag phase in intracellular growth cycle as a function of stage of growth of infecting Rickettsia prowazeki, with preliminary observations on inhibition of rickettsial uptake by host cell fragments. Infect Immun 1976; 13:1749-1760.         [ Links ]

75.Wisseman CL, Edlinger EA, Waddell AD, Jones MR. Infection cycle of Rickettsia rickettsii in chicken embryo and L-929 cells in culture. Infect Immun 1976; 14:1052-1064.         [ Links ]

76.Silverman DJ. Rickettsia rickettsii-induced cellular injury of human vascular endothelium in vitro. Infect Immun 1984; 44:545-553.         [ Links ]

77.Silverman DJ, Santucci LA. Potential for free radical-induced lipid peroxidation as a cause of endothelial cell injury in Rocky Mountain spotted fever. Infect Immun 1988; 56:3110-3115.         [ Links ]

78.ME, Silverman DJ. Effects of the antioxidant alpha-lipoic acid on human umbilical vein endothelial cells infected with Rickettsia rickettsii. Infect Immun 1998; 66:2290-2299.         [ Links ]

79.Walker DH, Firth WT, Ballard JG, Hegarty BC. Role of phospholipase-associated penetration mechanism in cell injury by Rickettsia rickettsii. Infect Immun 1983; 40:840-842.         [ Links ]

80.Bechelli JR, Rydkina E, Colonne PM, Sahni SK. Rickettsia rickettsii infection protects human microvascular endothelial cells against staurosporine-induced apoptosis by a cIAP(2)-independent mechanism. J Infect Dis 2009;1 99:1389-398.         [ Links ]

81.Uchiyama T, Kishi M, Ogawa M. Restriction of the growth of a nonpathogenic spotted fever group rickettsia. FEMS Immunol Med Microbiol 2012; 64:42-47.         [ Links ]

82.Michiels C. Endothelial cell functions. J Cell Physiol 2003; 196:430-443.         [ Links ]

83.Danese S, Dejana E, Fiocchi C. Immune regulation by microvascular endothelial cells: directing innate and adaptive immunity, coagulation, and inflammation. J Immunol 2007; 178:6017-6022.         [ Links ]

84.Pober JS, Min W, Bradley JR. Mechanisms of endothelial dysfunction, injury, and death. Annu Rev Pathol 2009; 4:71-95.         [ Links ]

85.George F, Brouqui P, Boffa MC, Mutin M, Drancourt M, Brisson C, et al. Demonstration of Rickettsia conorii-induced endothelial injury in vivo by measuring circulating endothelial cells, thrombomodulin, and von Willebrand factor in patients with Mediterranean spotted fever. Blood 1993; 82:2109-2016.         [ Links ]

86.La Scola B, Raoult D. Diagnosis of Mediterranean spotted fever by cultivation of Rickettsia conorii from blood and skin samples using the centrifugation-shell vial technique and by detection of R. conorii in circulating endothelial cells: a 6-year follow-up. J Clin Microbiol 1996; 34:2722-2727.         [ Links ]

87.Rydkina E, Sahni A, Baggs RB, Silverman DJ, Sahni SK. Infection of human endothelial cells with spotted Fever group rickettsiae stimulates cyclooxygenase 2 expression and release of vasoactive prostaglandins. Infect Immun 2006; 74:5067-5074.         [ Links ]

88.Woods ME, Wen G, Olano JP. Nitric oxide as a mediator of increased microvascular permeability during acute rickettsioses. Ann N Y Acad Sci 2005; 1063:239-245.         [ Links ]

89.Woods ME, Olano JP. Host defenses to Rickettsia rickettsii infection contribute to increased microvascular permeability in human cerebral endothelial cells. J Clin Immunol 2008; 28:174-185.         [ Links ]

90.Walker DH, Cain BG. The rickettsial plaque. Evidence for direct cytopathic effect of Rickettsia rickettsii. Lab Invest 1980; 43:388-396.         [ Links ]

91.Hong JE, Santucci LA, Tian X, Silverman DJ. Superoxide dismutase-dependent, catalase-sensitive peroxides in human endothelial cells infected by Rickettsia rickettsii. Infect Immun 1998; 66:1293-1298.         [ Links ]

92.Devamanoharan PS, Santucci LA, Hong JE, Tian X, Silverman DJ. Infection of human endothelial cells by Rickettsia rickettsii causes a significant reduction in the levels of key enzymes involved in protection against oxidative injury. Infect Immun 1994; 62:2619-2621.         [ Links ]

93.Walker DH, Valbuena GA, Olano JP. Pathogenic mechanisms of diseases caused by Rickettsia. Ann N Y Acad Sci 2003; 990:1-11.         [ Links ]

94.Rizzo M, Mansueto P, Di Lorenzo G, Morselli S, Mansueto S, Rini GB. Rickettsial disease: classical and modern aspects. New Microbiol 2004; 27:87-103.         [ Links ]

95.Bechah Y, Capo C, Mege JL, Raoult D. Epidemic typhus. Lancet Infect Dis 2008; 8:417-426.         [ Links ]

96.Chen LF, Sexton DJ. What’s new in Rocky Mountain spotted fever? Infect Dis Clin North Am 2008; 22:415-432.         [ Links ]

97.Demeester R, Claus M, Hildebrand M, Vlieghe E, Bottieau E. Diversity of life-threatening complications due to Mediterranean spotted fever in returning travelers. J Travel Med 2010; 17:100-104.         [ Links ]

98.Civen R, Ngo V. Murine typhus: an unrecognized suburban vectorborne disease. Clin Infect Dis 2008; 46:913-918.         [ Links ]

99.Sahni SK. Endothelial cell infection and hemostasis. Thromb Res 2007; 119:531-549.         [ Links ]

100.Schmaier AH, Srikanth S, Elghetany MT, Normolle D, Gokhale S, Feng HM, et al. Hemostatic/fibrinolytic protein changes in C3H/HeN mice infected with Rickettsia conorii--a model for Rocky Mountain spotted fever. Thromb Haemost 2001; 86:871-879.         [ Links ]

101.Walker DH, Occhino C, Tringali GR, Di Rosa S, Mansueto S. Pathogenesis of rickettsial eschars: the tache noire of boutonneuse fever. Hum Pathol 1988; 19:1449-1454.         [ Links ]

102.Chapman AS, Bakken JS, Folk SM, Paddock CD, Bloch KC, Krusell A, et al. Diagnosis and management of tickborne rickettsial diseases: Rocky Mountain spotted fever, ehrlichioses, and anaplasmosis--United States: a practical guide for physicians and other health-care and public health professionals. MMWR Recomm Rep 2006; 55:1-27.         [ Links ]

103.Rolain JM, Maurin M, Vestris G, Raoult D. In vitro susceptibilities of 27 rickettsiae to 13 antimicrobials. Antimicrob Agents Chemother 1998; 42:1537-1541.         [ Links ]

104.Holman RC, Paddock CD, Curns AT, Krebs JW, McQuiston JH, Childs JE. Analysis of risk factors for fatal Rocky Mountain Spotted Fever: evidence for superiority of tetracyclines for therapy. J Infect Dis 2001; 184:1437-1444.         [ Links ]

105.-Nevers E, Rovery C, Richet H, Raoult D. Analysis of risk factors for malignant Mediterranean spotted fever indicates that fluoroquinolone treatment has a deleterious effect. J Antimicrob Chemother 2011; 66:1821-1830.         [ Links ]

106.Audoly G, Vincentelli R, Edouard S, Georgiades K, Mediannikov O, Gimenez G, et al. Effect of rickettsial toxin VapC on its eukaryotic host. PLoS One 2011; 6:e26528.         [ Links ]

107.Gillespie JJ, Ammerman NC, Dreher-Lesnick SM, Rahman MS, Worley MJ, Setubal JC, et al. An anomalous type IV secretion system in Rickettsia is evolutionarily conserved. PLoS One 2009; 4:e4833.         [ Links ]

108.Driskell LO, Yu XJ, Zhang L, Liu Y, Popov VL, Walker DH, et al. Directed mutagenesis of the Rickettsia prowazekii pld gene encoding phospholipase D. Infect Immun 2009;7 7:3244-3248.         [ Links ]

109.Qin A, Tucker AM, Hines A, Wood DO. Transposon mutagenesis of the obligate intracellular pathogen Rickettsia prowazekii. Appl Environ Microbiol 2004; 70:2816-2822.         [ Links ]

110.Kleba B, Clark TR, Lutter EI, Ellison DW, Hackstadt T. Disruption of the Rickettsia rickettsii Sca2 autotransporter inhibits actin-based motility. Infect Immun 2010; 78:2240-2247.         [ Links ]

111.Fournier PE, El Karkouri K, Leroy Q, Robert C, Giumelli B, Renesto P, et al. Analysis of the Rickettsia africae genome reveals that virulence acquisition in Rickettsia species may be explained by genome reduction. BMC Genomics 2009; 10:166.         [ Links ]

112.Felsheim RF, Kurtti TJ, Munderloh UG. Genome sequence of the endosymbiont Rickettsia peacockii and comparison with virulent Rickettsia rickettsii: identification of virulence factors. PLoS One 2009; 4:e8361.         [ Links ]

113.Ellison DW, Clark TR, Sturdevant DE, Virtaneva K, Porcella SF, Hackstadt T. Genomic comparison of virulent Rickettsia rickettsii Sheila Smith and avirulent Rickettsia rickettsii Iowa. Infect Immun 2008; 76:542-550.         [ Links ]

114.Hackstadt T, Messer R, Cieplak W, Peacock MG. Evidence for proteolytic cleavage of the 120-kilodalton outer membrane protein of rickettsiae: identification of an avirulent mutant deficient in processing. Infect Immun 1992; 60:159-165.         [ Links ]

115.Ammerman NC, Rahman MS, Azad AF. Characterization of Sec-translocon-dependent extracytoplasmic proteins of Rickettsia typhi. J Bacteriol 2008; 190:6234-6242.         [ Links ]

116.Brossard M, Wikel SK. Tick immunobiology. Parasitology. 2004; 129 Suppl:S161-76.         [ Links ]

117.Wikel SK. Tick modulation of host immunity: an important factor in pathogen transmission. Int J Parasitol 1999; 29:851-859.         [ Links ]

118.Francischetti IM, Sa-Nunes A, Mans BJ, Santos IM, Ribeiro JM. The role of saliva in tick feeding. Front Biosci 2009; 14:2051-2088.         [ Links ]

119.Tyson KR, Elkins C, de Silva AM. A novel mechanism of complement inhibition unmasked by a tick salivary protein that binds to properdin. J Immunol 2008; 180:3964-3968.         [ Links ]

120.Steen NA, Barker SC, Alewood PF. Proteins in the saliva of the Ixodida (ticks): pharmacological features and biological significance. Toxicon 2006; 47:1-20.         [ Links ]

121.Ribeiro JM, Francischetti IM. Role of arthropod saliva in blood feeding: sialome and post-sialome perspectives. Annu Rev Entomol 2003; 48:73-88.         [ Links ]

122.Maxwell SS, Stoklasek TA, Dash Y, Macaluso KR, Wikel SK. Tick modulation of the in-vitro expression of adhesion molecules by skin-derived endothelial cells. Ann Trop Med Parasitol 2005; 99:661-672.         [ Links ]

123.Sporn LA, Sahni SK, Lerner NB, Marder VJ, Silverman DJ, Turpin LC, et al. Rickettsia rickettsii infection of cultured human endothelial cells induces NF-kappaB activation. Infect Immun 1997; 65:2786-2791.         [ Links ]

124.Sahni SK, Van Antwerp DJ, Eremeeva ME, Silverman DJ, Marder VJ, Sporn LA. Proteasome-independent activation of nuclear factor kappaB in cytoplasmic extracts from human endothelial cells by Rickettsia rickettsii. Infect Immun 1998; 66:1827-1833.         [ Links ]

125.Sahni SK, Rydkina E, Joshi SG, Sporn LA, Silverman DJ. Interactions of Rickettsia rickettsii with endothelial nuclear factor-kappaB in a “cell-free” system. Ann NY Acad Sci 2003; 990:635-641.         [ Links ]

126.Sahni SK, Kiriakidi S, Colonne MP, Sahni A, Silverman DJ. Selective activation of signal transducer and activator of transcription (STAT) proteins STAT1 and STAT3 in human endothelial cells infected with Rickettsia rickettsii. Clin Microbiol Infect 2009; 15 Suppl 2:303-304.         [ Links ]

127.Rydkina E, Silverman DJ, Sahni SK. Activation of p38 stress-activated protein kinase during Rickettsia rickettsii infection of human endothelial cells: role in the induction of chemokine response. Cell Microbiol 2005; 7:1519-1530.         [ Links ]

128.Rydkina E, Turpin LC, Sahni SK. Activation of p38 mitogen-activated protein kinase module facilitates in vitro host cell invasion by Rickettsia rickettsii. J Med Microbiol 2008; 57:1172-1175.         [ Links ]

129.Rydkina E, Turpin LC, Sahni SK. Rickettsia rickettsii infection of human macrovascular and microvascular endothelial cells reveals activation of both common and cell type-specific host response mechanisms. Infect Immun 2010; 78:2599-2606.         [ Links ]

130.Valbuena G, Bradford W, Walker DH. Expression analysis of the T-cell-targeting chemokines CXCL9 and CXCL10 in mice and humans with endothelial infections caused by rickettsiae of the spotted fever group. Am J Pathol 2003; 163:1357-1369.         [ Links ]

131.Valbuena G, Walker DH. Effect of blocking the CXCL9/10-CXCR3 chemokine system in the outcome of endothelial-target rickettsial infections. Am J Trop Med Hyg 2004; 71:393-399.         [ Links ]

132.Kaplanski G, Teysseire N, Farnarier C, Kaplanski S, Lissitzky JC, Durand JM, et al. IL-6 and IL-8 production from cultured human endothelial cells stimulated by infection with Rickettsia conorii via a cell-associated IL-1 alpha-dependent pathway. J Clin Invest 1995; 96:2839-2844.         [ Links ]

133.Sporn LA, Marder VJ. Interleukin-1 alpha production during Rickettsia rickettsii infection of cultured endothelial cells: potential role in autocrine cell stimulation. Infect Immun 1996; 64:1609-1613.         [ Links ]

134.Sporn LA, Lawrence SO, Silverman DJ, Marder VJ. E-selectin-dependent neutrophil adhesion to Rickettsia rickettsii-infected endothelial cells. Blood 1993; 81:2406-2412.         [ Links ]

135.Dignat-George F, Teysseire N, Mutin M, Bardin N, Lesaule G, Raoult D, et al. Rickettsia conorii infection enhances vascular cell adhesion molecule-1- and intercellular adhesion molecule-1-dependent mononuclear cell adherence to endothelial cells. J Infect Dis 1997; 175:1142-1152.         [ Links ]

136.Damås JK, Davì G, Jensenius M, Santilli F, Otterdal K, Ueland T, et al. Relative chemokine and adhesion molecule expression in Mediterranean spotted fever and African tick bite fever. J Infect 2009; 58:68-75.         [ Links ]

137.Bechah Y, Capo C, Grau G, Raoult D, Mege JL. Rickettsia prowazekii infection of endothelial cells increases leukocyte adhesion through alphavbeta3 integrin engagement. Clin Microbiol Infect 2009; 15 Suppl 2:249-250.         [ Links ]

138.Rydkina E, Turpin LC, Silverman DJ, Sahni SK. Rickettsia rickettsii infection of human pulmonary microvascular endothelial cells: modulation of cyclooxygenase-2 expression. Clin Microbiol Infect 2009; 15 Suppl 2:300-302.         [ Links ]

139.Billings AN, Feng HM, Olano JP, Walker DH. Rickettsial infection in murine models activates an early anti-rickettsial effect mediated by NK cells and associated with production of gamma interferon. Am J Trop Med Hyg 2001; 65:52-56.         [ Links ]

140.Jordan JM, Woods ME, Soong L, Walker DH. Rickettsiae stimulate dendritic cells through toll-like receptor 4, leading to enhanced NK cell activation in vivo. J Infect Dis 2009; 199:236-242.         [ Links ]

141.Feng HM, Popov VL, Walker DH. Depletion of gamma interferon and tumor necrosis factor alpha in mice with Rickettsia conorii-infected endothelium: impairment of rickettsicidal nitric oxide production resulting in fatal, overwhelming rickettsial disease. Infect Immun 1994; 62:1952-1960.         [ Links ]

142.Walker DH, Popov VL, Crocquet-Valdes PA, Welsh CJ, Feng HM. Cytokine-induced, nitric oxide-dependent, intracellular antirickettsial activity of mouse endothelial cells. Lab Invest 1997; 76:129-138.         [ Links ]

143.Feng HM, Walker DH. Mechanisms of intracellular killing of Rickettsia conorii in infected human endothelial cells, hepatocytes, and macrophages. Infect Immun 2000; 68:6729-6736.         [ Links ]

144.Mansueto P, Vitale G, Di Lorenzo G, Arcoleo F, Mansueto S, Cillari E. Immunology of human rickettsial diseases. J Biol Regul Homeost Agents 2008; 22:131-139.         [ Links ]

145.Feng H, Popov VL, Yuoh G, Walker DH. Role of T lymphocyte subsets in immunity to spotted fever group Rickettsiae. J Immunol 1997; 158:5314-5320.         [ Links ]

146.Walker DH, Olano JP, Feng HM. Critical role of cytotoxic T lymphocytes in immune clearance of rickettsial infection. Infect Immun 2001; 69:1841-1846.         [ Links ]

147.G, Jordan JM, Walker DH. T cells mediate cross-protective immunity between spotted fever group rickettsiae and typhus group rickettsiae. J Infect Dis 2004; 190:1221-1227.         [ Links ]

148.Ricketts HT, Gomez L. Studies on immunity in Rocky Mountain spotted fever. First communication. J Infect Dis 1908; 5:221-244.         [ Links ]

149.Shirai A, Dietel JW, Osterman JV. Indirect hemagglutination test for human antibody to typhus and spotted fever group rickettsiae. J Clin Microbiol 1975; 2:430-437.         [ Links ]

150.Vishwanath S. Antigenic relationships among the rickettsiae of the spotted fever and typhus groups. FEMS Microbiol Lett 1991; 65:341-344.         [ Links ]

151.Anacker RL, McDonald GA, List RH, Mann RE. Neutralizing activity of monoclonal antibodies to heat-sensitive and heat-resistant epitopes of Rickettsia rickettsii surface proteins. Infect Immun 1987; 55:825-827.         [ Links ]

152.Mason RA, Wenzel RP, Seligmann EB, Ginn RK. A reference, inactivated, epidemic typhus vaccine: clinical trials in man. J Biol Stand 1976; 4:217-224.         [ Links ]

153.DuPont HL, Hornick RB, Dawkins AT, Heiner GG, Fabrikant IB, Wisseman CL, et al. Rocky Mountain spotted fever: a comparative study of the active immunity induced by inactivated and viable pathogenic Rickettsia rickettsii. J Infect Dis 1973; 128:340-344.         [ Links ]

154.Clements ML, Wisseman CL, Woodward TE, Fiset P, Dumler JS, McNamee W, et al. Reactogenicity, immunogenicity, and efficacy of a chick embryo cell-derived vaccine for Rocky Mountain spotted fever. J Infect Dis 1983; 148:922-930.         [ Links ]

155.Woodward TE. Rickettsial vaccines with emphasis on epidemic typhus. Initial report of an old vaccine trial. S Afr Med J 1986; Suppl:73-76.         [ Links ]

156.Fox JP, Everritt MG, Robinson TA, Conwell DP. Immunization of man against epidemic typhus by infection with avirulent Rickettsia prowazeki (strain E); observations as to post-vaccination reactions, the relation of serologic response to size and route of infecting dose, and the resistance to challenge with virulent typhus strains. Am J Hyg 1954; 59:74.         [ Links ]

157.Balayeva NM, Nikolskaya VN. Analysis of lung culture of Rickettsia prowazekii E strain with regard to its capacity of increasing virulence in passages on the lungs of white mice. J Hyg Epidemiol Microbiol Immunol 1973; 17:294-303.         [ Links ]

158.Nikolskaya VN, Balayeva NM. Homogeneity of Rickettsia prowazekii E strain egg culture as to the capacity to increase virulence in passages on white mouse lungs. J Hyg Epidemiol Microbiol Immunol 1973; 17:505-506.         [ Links ]

159.Zhang JZ, Hao JF, Walker DH, Yu XJ. A mutation inactivating the methyltransferase gene in avirulent Madrid E strain of Rickettsia prowazekii reverted to wild type in the virulent revertant strain Evir. Vaccine 2006; 24:2317-2323.         [ Links ]

160.Crocquet-Valdes PA, Díaz-Montero CM, Feng HM, Li H, Barrett AD, Walker DH. Immunization with a portion of rickettsial outer membrane protein A stimulates protective immunity against spotted fever rickettsiosis. Vaccine 2001; 20:979-988.         [ Links ]

161.Sumner JW, Sims KG, Jones DC, Anderson BE. Protection of guinea-pigs from experimental Rocky Mountain spotted fever by immunization with baculovirus-expressed Rickettsia rickettsii rOmpA protein. Vaccine 1995; 13:29-35.         [ Links ]

162.Churilla A, Ching WM, Dasch GA, Carl M. Human T lymphocyte recognition of cyanogen bromide fragments of the surface protein of Rickettsia typhi. Ann NY Acad Sci 1990; 590:215-220.         [ Links ]

163.Li Z, Díaz-Montero CM, Valbuena G, Yu XJ, Olano JP, Feng HM, et al. Identification of CD8 T-lymphocyte epitopes in OmpB of Rickettsia conorii. Infect Immun 2003; 71:3920-3926.         [ Links ]

164.Chan YG, Riley SP, Chen E, Martinez JJ. Molecular Basis of Immunity to Rickettsial Infection Conferred through Outer Membrane Protein B. Infect Immun 2011; 79:2303-2313.         [ Links ]

165.Zinsser H, Castaneda MR. On the isolation from a case of Brill’s disease of a typhus strain resembling the European type. N Engl J Med 1933; 209:815-819.         [ Links ]

166.Zinsser H. The rickettsial diseases: variety, epidemiology and geographical distribution. Am J Hyg 1937; 25:430-463.         [ Links ]

167.WC, Waner JL. Serological cross-reaction and cross-protection in guinea pigs infected with Rickettsia rickettsii and Rickettsia montana. Infect Immun 1980; 28:627-629.         [ Links ]

168.Eisemann CS, Nypaver MJ, Osterman JV. Susceptibility of inbred mice to rickettsiae of the spotted fever group. Infect Immun 1984; 43:143-148.         [ Links ]

169.TR, Jarboe DL, Eisemann CS. Cross-reactive lymphocyte responses and protective immunity against other spotted fever group rickettsiae in mice immunized with Rickettsia conorii. Infect Immun 1986; 51:832-837.         [ Links ]

170.Gage KL, Jerrells TR. Demonstration and partial characterization of antigens of Rickettsia rhipicephali that induce cross-reactive cellular and humoral immune responses to Rickettsia rickettsii. Infect Immun 1992; 60:5099-5106.         [ Links ]

171.Feng HM, Walker DH. Cross-protection between distantly related spotted fever group rickettsiae. Vaccine 2003; 21:3901-3905.         [ Links ]

172.Wu CY, Kirman JR, Rotte MJ, Davey DF, Perfetto SP, Rhee EG, et al. Distinct lineages of T(H)1 cells have differential capacities for memory cell generation in vivo. Nat Immunol 2002; 3:852-858.         [ Links ]

173.Darrah PA, Patel DT, De Luca PM, Lindsay RW, Davey DF, Flynn BJ, et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat Med 2007; 13:843-850.         [ Links ]

174.Lindenstrøm T, Agger EM, Korsholm KS, Darrah PA, Aagaard C, Seder RA, et al. Tuberculosis subunit vaccination provides long-term protective immunity characterized by multifunctional CD4 memory T cells. J Immunol 2009; 182:8047-8055.         [ Links ]

175.Tenzer S, Wee E, Burgevin A, Stewart-Jones G, Friis L, Lamberth K, et al. Antigen processing influences HIV-specific cytotoxic T lymphocyte immunodominance. Nat Immunol 2009; 10:636-646.         [ Links ]

176.Miller JD, van der Most RG, Akondy RS, Glidewell JT, Albott S, Masopust D, et al. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity 2008; 28:710-722.         [ Links ]

177.Akondy RS, Monson ND, Miller JD, Edupuganti S, Teuwen D, Wu H, et al. The yellow fever virus vaccine induces a broad and polyfunctional human memory CD8+ T cell response. J Immunol 2009; 183:7919-7930.         [ Links ]

178.Rinaudo CD, Telford JL, Rappuoli R, Seib KL. Vaccinology in the genome era. J Clin Invest 2009; 119:2515-2525.         [ Links ]

179.Haining WN, Wherry EJ. Integrating genomic signatures for immunologic discovery. Immunity 2010; 32:152-161.         [ Links ]

180.Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol 2009; 10:116-125.         [ Links ]

181.Gaucher D, Therrien R, Kettaf N, Angermann BR, Boucher G, Filali-Mouhim A, et al. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J Exp Med 2008; 205:3119-3131.         [ Links ]

182.Moutaftsi M, Bui HH, Peters B, Sidney J, Salek-Ardakani S, Oseroff C, et al. Vaccinia virus-specific CD4+ T cell responses target a set of antigens largely distinct from those targeted by CD8+ T cell responses. J Immunol 2007; 178:6814-6820.         [ Links ]

183.Sette A, Rappuoli R. Reverse vaccinology: developing vaccines in the era of genomics. Immunity 2010; 33:530-541.         [ Links ]

184.Halling-Brown M, Sansom CE, Davies M, Titball RW, Moss DS. Are bacterial vaccine antigens T-cell epitope depleted? Trends Immunol 2008; 29:374-379.         [ Links ]

185.Sykes K. Progress in the development of genetic immunization. Expert Rev Vaccines 2008; 7:1395-1404.         [ Links ]

186.Stemke-Hale K, Kaltenboeck B, DeGraves FJ, Sykes KF, Huang J, Bu CH, et al. Screening the whole genome of a pathogen in vivo for individual protective antigens. Vaccine 2005; 23:3016-3025.         [ Links ]

187.Riedl P, Wieland A, Lamberth K, Buus S, Lemonnier F, Reifenberg K, et al. Elimination of immunodominant epitopes from multispecific DNA-based vaccines allows induction of CD8 T cells that have a striking antiviral potential. J Immunol 2009; 183:370-380.         [ Links ]

188.Ruckwardt TJ, Luongo C, Malloy AM, Liu J, Chen M, Collins PL, et al. Responses against a subdominant CD8+ T cell epitope protect against immunopathology caused by a dominant epitope. J Immunol 2010; 185:4673-4680.         [ Links ]

189.Im EJ, Hong JP, Roshorm Y, Bridgeman A, Létourneau S, Liljeström P, et al. Protective efficacy of serially up-ranked subdominant CD8 T cell epitopes against virus challenges. PLoS Pathog 2011; 7:e1002041.         [ Links ]

Creative Commons License All the contents of this journal, except where otherwise noted, is licensed under a Creative Commons Attribution License