Hostname: page-component-8448b6f56d-qsmjn Total loading time: 0 Render date: 2024-04-23T07:44:41.788Z Has data issue: false hasContentIssue false

Diagnosis of spotted fever group Rickettsia infections: the Asian perspective

Published online by Cambridge University Press:  07 October 2019

Matthew T. Robinson
Affiliation:
Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Mahosot Hospital, Vientiane, Lao People's Democratic Republic Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Churchill Hospital, Oxford, OX3 7FZ, UK
Jaruwan Satjanadumrong
Affiliation:
Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithee Road, 10400 Bangkok, Thailand
Tom Hughes
Affiliation:
EcoHealth Alliance, 460 West 34th Street, 17th floor, New York, NY, USA
John Stenos
Affiliation:
Australian Rickettsial Reference Laboratory, Barwon Health, Geelong VIC 3220, Australia
Stuart D. Blacksell*
Affiliation:
Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Mahosot Hospital, Vientiane, Lao People's Democratic Republic Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Churchill Hospital, Oxford, OX3 7FZ, UK Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithee Road, 10400 Bangkok, Thailand
*
Author for correspondence: Stuart D. Blacksell, E-mail: stuart@tropmedres.ac
Rights & Permissions [Opens in a new window]

Abstract

Spotted fever group rickettsiae (SFG) are a neglected group of bacteria, belonging to the genus Rickettsia, that represent a large number of new and emerging infectious diseases with a worldwide distribution. The diseases are zoonotic and are transmitted by arthropod vectors, mainly ticks, fleas and mites, to hosts such as wild animals. Domesticated animals and humans are accidental hosts. In Asia, local people in endemic areas as well as travellers to these regions are at high risk of infection. In this review we compare SFG molecular and serological diagnostic methods and discuss their limitations. While there is a large range of molecular diagnostics and serological assays, both approaches have limitations and a positive result is dependent on the timing of sample collection. There is an increasing need for less expensive and easy-to-use diagnostic tests. However, despite many tests being available, their lack of suitability for use in resource-limited regions is of concern, as many require technical expertise, expensive equipment and reagents. In addition, many existing diagnostic tests still require rigorous validation in the regions and populations where these tests may be used, in particular to establish coherent and worthwhile cut-offs. It is likely that the best strategy is to use a real-time quantitative polymerase chain reaction (qPCR) and immunofluorescence assay in tandem. If the specimen is collected early enough in the infection there will be no antibodies but there will be a greater chance of a PCR positive result. Conversely, when there are detectable antibodies it is less likely that there will be a positive PCR result. It is therefore extremely important that a complete medical history is provided especially the number of days of fever prior to sample collection. More effort is required to develop and validate SFG diagnostics and those of other rickettsial infections.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s) 2019

Introduction

Rickettsioses are infectious diseases caused by obligate intracellular gram-negative bacteria. They belong to the order Rickettsiales and reside in a wide range of arthropod vectors such as fleas, ticks and mites [Reference Parola1]. These vectors can transmit pathogens to humans at the bite site, who may or may not subsequently develop disease. Rickettsial diseases have been reported to be the second most common cause of non-malarial febrile illness in the Southeast Asia region after dengue infection [Reference Acestor2].

Pathogenic members of the Rickettsia are classified into two major groups: spotted fever group (SFG) and typhus group (TG) rickettsiae and additional, Orientia tsutsugamushi and O. chuto are classified as scrub typhus group (STG) [Reference Bhengsri3]. Although rickettsial diseases have worldwide distribution, there are endemic and hyper-endemic areas. TG and STG rickettsiae are widely diagnosed in Southeast Asia [Reference Parola1, Reference Aung4, Reference Rodkvamtook5]. In Asia, TG infections are mainly caused by Rickettsia typhi [Reference Newton6, Reference Vallee7] which is the etiologic agent of murine typhus (or endemic typhus). Rickettsia prowazekii, also a TG rickettsiae and responsible for louse-borne typhus (epidemic typhus), is rarely detected. Scrub typhus is caused by O. tsutsugamushi along with the related O. chuto [Reference Chikeka and Dumler8, Reference Izzard9]. O. tsutsugamushi is widespread in the Asia-Pacific (and northern Australia) however, when O. chuto is included, the geographical range is extended to include the Middle-east, Africa and South America [Reference Izzard9, Reference Luce-Fedrow10]. The SFG consists of over 30 species that can be found worldwide. The one of the most studied is R. rickettsii which causes Rocky Mountain spotted fever (RMSF) in North America [Reference Kato11]. Other species such as R. australis and R. honei are prevalent in northern Australia [Reference Graves and Stenos12]. Rickettsia conorii is responsible for Mediterranean spotted fever (MSF) in several parts of Europe, Africa and Asia [Reference Niang13Reference Nanayakkara15]. Rickettsia felis (known as the causative agent of flea-borne spotted fever) is seen as an emerging infectious disease. First identified in the USA and now seen worldwide, it is also responsible for many cases of febrile illness in Africa [Reference Brown and Macaluso16].

The main arthropod vectors of SFG are hard ticks (Ixodidae), although soft ticks (Argasidae) are also implicated in a number of SFG [Reference Parola1, Reference Luce-Fedrow17]. Other SFG such as R. felis, may be transmitted by fleas, whilst more recently, mosquitoes have also been demonstrated to be competent vectors [Reference Dieme18]. Dependent on the vector, transmission of SFG occurs via salivary products produced during feeding or through inoculation with the faeces of infected vectors on the wound, mucosal surfaces and via inhalation. Rickettsial infections occur following infection of the endothelial cell lining of blood vessels (microvascular endothelium in the case of infection by R. conorii and both microvascular and macrovascular endothelium in the case of R. rickettsii) [Reference Colonne, Eremeeva and Sahni19, Reference Rydkina, Turpin and Sahni20]. Symptoms at clinical presentation are variable and are often similar to many other acute febrile illnesses. Careful clinical observation by physicians and reliable laboratory diagnosis can lead to early appropriate administration of antibiotic therapy and patient management and thereby reduce patient mortality or clinical complications. The purpose of this article is to compare and contrast molecular and serological identification methods including limitations for the diagnosis of SFG infections with reference to the Asian perspective especially in low- and middle-income countries (LMICs)

SFG infection and diagnosis

Infection and clinical presentation

The infection cycle of SFG in humans starts with the arthropod bite (normally ticks or fleas) followed by an incubation period of up to fifteen days prior to the onset of clinical symptoms (Fig. 1). Clinical symptoms of SFG infection can vary, ranging from mild to life-threatening. Patients suspected of SFG infection normally present with fever, nausea, vomiting, maculopapular rash and occasionally eschars at the site of inoculation [Reference Gaywee21]. SFG infection can result in variable and often severe clinical symptoms in individual patients and the lack of eschar evidence can lead to misdiagnosis and often delays in commencing antibiotic treatment. For instance, in Hong Kong, treatment was delayed due to misdiagnosis of a case of MSF infection with meningitis symptoms; this resulted in inappropriate treatment and a fatal outcome [Reference Young, Ip and Bassett22]. In addition, some MSF patients have shown complications such as hearing loss [Reference Rossio23], acute myocarditis [Reference Ben Mansour24] and cerebral infarction [Reference Botelho-Nevers25]. In Japan, severe Japanese spotted fever cases (R. japonica) have been reported with acute respiratory failure complications, including acute respiratory distress syndrome [Reference Kodama, Noguchi and Chikahira26, Reference Takiguchi27]. In other cases, complications from SFG infections can include renal failure, purpura fulminans and severe pneumonia [Reference McBride28]. Therefore, the availability of diagnostic techniques for SFG infections is important, as well as SFG-awareness of physicians to enable the early administration of appropriate antibiotic treatment.

Fig. 1. Features and temporal aspects of SFG Rickettsia infection and diagnosis.

SFG laboratory diagnosis

Molecular detection

Polymerase chain reaction (PCR)-based detection is the primary method to detect SFG, especially for the early detection of infection before the development of detectable antibodies [Reference Santibanez29] (Fig. 1). Molecular-based techniques need to be sensitive, and the sample type and assay used determine the success of detection. PCRs are used for both epidemiological and diagnostic purposes and SFG DNA may be isolated from arthropods, animal hosts and human clinical samples including whole blood, buffy coat, serum, tissue biopsies (such as skin), eschar scrapings and swabs [Reference La Scola and Raoult30]. For human clinical diagnostic purposes, whole blood or buffy coat is the preferred sample type, as SFG are intracellular (the cellular component being concentrated in the buffy coat fraction, increasing the sensitivity of detection) and the sample type being easily collected. Concentration of the sample type and maximizing the sensitivity of assays are a very important consideration for the detection of rickettsiae. Although little is known of the quantities of SFG in blood, quantities of other rickettsial organisms in the blood of an infected patient are variable. In RMSF with nonfatal outcomes, R. rickettsii copy numbers ranged from 8.40 × 101 to 3.95 × 105 copies/ml of blood, whilst in patients with fatal infections copy numbers ranged from 1.41 × 103 to 2.05 × 106 copies/ml of blood [Reference Kato, Chung and Paddock31]. By comparison, the STG O. tsutsugamushi may be found at a median density of 13 genome copies/ml of blood (IQR: 0–334) [Reference Sonthayanon32]. Serum or plasma may be used for the PCR, but these samples are less than optimal as there will be fewer patient cells (meaning lower rickettsiae concentration). In addition, serum may have increased concentrations of blood fibrinogen and fibrin materials which can bind to Rickettsia DNA decreasing availability of DNA target for PCRs [Reference Kondo33]. Eschars are a suitable sample type for the PCR (as well as culture techniques) and may be sampled as either scrapings, swabs or biopsied specimen [Reference Bechah, Socolovschi and Raoult34]. As with any sample type, the specimen needs to be maintained at optimal temperatures or preserved prior to detection and/or isolation. Formalin-fixed and paraffin-embedded tissues can also be used, but fixation using formalin causes nucleic acid fragmentation and reduces the quality and quantity of nucleic acids as well as limiting the length of PCR products [Reference Denison35].

Selection of an appropriate gene target is important. Conserved gene targets enable a broad Rickettsia genus- or SFG-level of detection. The use of gene targets such as the citrate synthase (gltA), 16S rRNA and 17 kDa lipoprotein outer membrane antigens (17 kDa) generally confirms the presence of SFG or TG rickettsiae [Reference Ishikura36Reference Roux and Raoult39]. The use of Outer Membrane Protein A (190 kDa) (ompA) and B (ompB) genes appear to be more specific and discriminating for SFG in both patient and animal samples [Reference Santibanez29, Reference Prakash38].With all of these gene targets, down-stream sequencing of the PCR product will discern, in most cases, specific species. It is strongly recommended that multiple gene targets are used to gain an accurate identification.

A variety of different PCR-based assays are available for the diagnosis of SFG infections. Table 1 lists the most commonly used conventional (cPCR), nested (nPCR) and quantitative PCRs (qPCRs) that have been developed. Many of the primer sets have been mixed-and-matched and optimized in different studies to maximize the identification of SFG infections. cPCR assays have targeted most of the key genes discussed. nPCRs (which use two sets of primers) have been developed to increase the detection sensitivity over cPCRs although, as with other PCR assays, there may still be difficulty in differentiating closely-related SFG species, such as R. conorii and R. sibirica [Reference Choi40]. Several real-time or quantitative PCR (qPCR) assays developed for rickettsial detection have largely replaced the use of nPCRs due to a greater sensitivity and shorter run-time (Table 1). Furthermore, multiplexed qPCR is an efficient method that demonstrates greater analytical power, as multiple primers and probes are combined into a single assay, either targeting a number of different species or different gene targets for a single species at the same time [Reference Denison35].

Table 1. Summary of common SFG PCRs used for diagnosis and sequencing for species identification purposes

Species-specific PCRs excluded. PCR type: cPCR, conventional; nPCR, nested (including semi-nested); qPCR, quantitative/real-time PCR. PCR use: VD, diagnostics in arthropod vectors; HD, diagnostics in human samples; S, used for sequencing purposes.

a aka: fD1.

b aka: Rick17 assay.

c aka: Rick17b assay (developed from Rick17 assay).

d aka: 17 kDa-1/Primer1; addition of ACA on 3′ end in references [Reference Jiang42,Reference Taylor45].

e aka: 17 kDa-2/Rr2608Rnew/Primer2; base change G20A in references [Reference Santibanez29,Reference Jiang42,Reference Taylor45]; base change T7C in references [Reference Jiang42,Reference Taylor45]; base change A22C in reference [Reference Prakash38].

f Base change A22C in reference [Reference Prakash38].

g aka: CS1273R; base change G5A in reference [Reference Prakash38].

An alternative to the standard PCR assays, suitable for more field-based diagnosis, is the loop-mediated isothermal amplification assay (LAMP). This has a potential for application as a simple and rapid molecular SFG detection technique, employing an isothermal (constant temperature) nucleic acid technique for the amplification of DNA. The amplification is performed at 60–65 °C and the stop reaction at 80 °C [Reference Pan60]. The LAMP has been reported to be more sensitive than the nPCR to detect ompB from SFG in China, with 73% sensitivity and 100% specificity compared with the nPCR which gave negative results [Reference Pan60].

As with all PCR assays, it is also possible that a vector may carry more than one rickettsial species resulting in multiple PCR products being obtained if using broad range primers (not species-specific). This can cause difficulties in interpreting results, and may even result in misdiagnosing of the actual agent causing disease. Therefore, if the PCR results show positivity for SFG but the assay cannot differentiate between species, the amplicons should be sequenced for further species identification which provides useful epidemiological information.

Serological detection

The genus Rickettsia and Orientia can be characterized into three major antigenic groups: SFG, TG and STG [Reference Paris and Dumler37]. There are serological cross reactions between SFG and TG antibodies [Reference Luce-Fedrow17, Reference La Scola and Raoult30, Reference Paris and Dumler37] as well amongst antibodies of Rickettsia spp. within SFG and therefore serological diagnosis is normally only made to the antigenic (serogroup) group level. Discrimination to the species level within SFG using serological techniques is extremely difficult and only possible following cross-absorption using western blot techniques [Reference La Scola and Raoult30, Reference Raoult and Dasch61].

In many cases, rickettsial infection is confirmed by the use of serological techniques such as indirect immunofluorescence assay (IFA), indirect immunoperoxidase (IIP) test, enzyme-linked immunosorbent assay (ELISA) and the Weil–Felix agglutination test (WFT). Immunoglobulin M (IgM) antibodies are present in the early phase of an infection and reduces afterwards until they are undetectable after a few weeks (Fig. 1). In contrast, IgG increases after the second week of illness and persists at low titers for years in some cases (30% found after one year) [Reference McQuiston62]. The WFT was developed in 1916 and primarily used in mid-1940's for the identification of TG rickettsiae infections [Reference Cruickshank63]. The WFT is an inexpensive test, principally based on the antibodies against Gram-negative Proteus spp. antigens of OX (O-specific polysaccharide chain of outer membrane lipopolysaccharide) which cross-reacts with Rickettsia antigens OX-2 and OX-19 with the former more specific to SFG [Reference Amano64]. However, the WFT demonstrates low sensitivity in the acute phase of infection as demonstrated by 47% sensitivity in RMSF [Reference Kaplan and Schonberger65] and 33% sensitivity for SFG in Sri Lanka [Reference Kularatne and Gawarammana66] and is rarely use nowadays to this shortcoming.

In many Asian countries, IFA and IIP are recognized as standard methods for routine diagnosis. The sensitivity of IIP and IFA often depends on the timing of serum collection due to the lack of antibodies in the first week of illness prior to complete seroconversion [Reference Tay and Rohani67] although it can demonstrate high sensitivity (83–100%) and specificity (99–100%) (Table 2) [Reference Paris and Dumler37]. A seroconversion or a four-fold difference in antibodies from acute to convalescent phase IgM and IgG antibodies is considered to be significant [Reference McQuiston62, Reference Premaratna68] however, the diagnostic accuracy of such tests is also dependent on the cut-offs applied and therefore a understanding of the background immunity in endemic and non-endemic populations is essential with higher cut-offs often used in endemic settings. Furthermore, IFA and IIP results are dependent on appropriate antigenic types (often R. honei and R. conorii in Asia), well-trained personnel for the interpretation of the results, and requires a fluorescence microscope, which is often expensive and difficult to maintain.

Table 2. The comparison of sensitivity and specificity of serological techniques for SFG infection

NA, not available; ST, scrub typhus; MT, murine typhus; Sens, sensitivity; Spec, specificity; WF, Weil–Felix; CF, complement fixation; IFA, immunofluorescence assay; RMSF, Rocky Mountain spotted fever; SFG, spotted fever group Rickettsia; STG, scrub typhus group; TG, typhus group.

Enzyme-linked immunosorbent assays (ELISAs) are widely used and demonstrate high sensitivity and specificity. In an Indian study, researchers used a commercial R. conorii ELISA IgG/IgM kit that demonstrated 85% sensitivity and 100% specificity [Reference Kalal73] (Table 2). The advantage of ELISA methodologies is that they allow screening of large batches of samples, which is less time-consuming, and provide more objectivity compared to the subjective IFA, as there is no reader-bias due to the use of optical density (OD) by ELISA readers. A diagnostic cut-off OD of ⩾0.5 has been applied previously to the SFG ELISA developed by the US Naval Medical Research Center [Reference Trung74, Reference Khan75], however, there is no independent validation of the use of this cut-off. However, ELISAs may not recognize all Rickettsia pathogens and may be negative at the acute stage of an infection depending on the isotype (IgM or IgG) (Fig. 1), and results can only be obtained after seroconversion (around 5–14-days post onset of illness) [Reference Clements71,Reference Rathi72]. Collecting only a single sample from patients can often make diagnosis difficult [Reference Tsai76] and it is recommended that both acute and convalescent samples are collected whereby a significant rise in antibody levels is then considered as indicative of an active infection.

Need to improve diagnosis and clinical awareness of SFG

There is an urgent need to improve diagnosis and awareness of SFG in Asia. Generally, SFG infections are significantly neglected and under recognized in Asia. Spotted fever rickettsiae diseases often have non-specific clinical symptoms and may be overlooked by physicians [Reference Raoult and Dasch61, Reference Salgado Lynn77], although awareness is increasing with improved diagnostics [Reference Luce-Fedrow17, Reference Kondo33, Reference Paris and Dumler37] and publication of findings. SFG are also under recognized by laboratory staff as SFG are obligate intracellular bacteria whose culture in vitro is complicated, lengthy, expensive and often reserved to specialized laboratories equipped with BSL3 level containment facilities [Reference Ammerman, Beier-Sexton and Azad78].

SFG may be misidentified as TG Rickettsia due to serological cross-reactions in which the rOmpB protein appears to play an important role [Reference Kantso79Reference Ormsbee81]. Moreover, sequencing data indicates that some 50 amino acids in rOmpB of R. japonica (SFG) are identical to that of R. typhi (TG) [Reference Uchiyama82]. Antibodies for R. typhi have demonstrated greater cross-reaction with R. conorii than R. rickettsii and higher cross-reactivity with IgM than IgG antibodies. However, there may be enough difference between end point titers to permit identity between TG and SFG [Reference Hechemy83]. The cross-reacting antibodies may also depend on the immunogenic responses of each patient [Reference Uchiyama82] and multiple infections and re-infections also make it difficult to distinguish between species due to the broadness of the immune response.

In the case of molecular diagnostic tests, adoption of PCR technologies is limited due to cost, lack of expertise and technical issues. Although often highly sensitive and specific, PCR techniques have limitations as false-negative results may result from low quantities of SFG and the transient nature of these pathogens present in the circulating blood [Reference Kidd84]. In reality, the major limiting factor for the use of PCR methods is the need for expensive thermocycler equipment, reagents and specific primers. Despite this, these techniques should be encouraged as PCR assays are highly useful, they allow accurate SFG identification, enable the discovery of novel species, are increasingly affordable, reproducible and less-time consuming with high specificity and sensitivity especially in the early phase of infection [Reference Znazen85].

Diagnostic cutoffs in seroprevalence studies

Seroprevalence studies provide important information regarding the distribution of SFG in community- or hospital-based settings. The choice of diagnostic cut off may greatly influence the seroprevalence results, with a low cut off often giving high sensitivity but low specificity, whereas a high cut off will give low sensitivity but high specificity [Reference Newhouse69]. Seroprevalence studies of SFG in Asia have used IFA, IIP and ELISA techniques (Table 1) with IFA considered to be the diagnostic gold standard for the quantitative detection of rickettsial antibodies [Reference La Scola and Raoult30]. However, there appears to be a lack of consistency in the application of the cut-off titer criteria depending on the geographic location (Table 2). In the case of seroprevalence studies, the application of a consistent regional cut off will more readily reflect true endemicity and enable comparability between countries and regions. In Southeast, South and East Asia, studies have used reasonably consistent cut-off titers of ⩾1:64 in Philippines [Reference Camer86], Sri Lanka [Reference Premaratna87], Thailand [Reference Bhengsri3] and Bangladesh [Reference Faruque88] and in South Korea and Taiwan, a cut-off of 1:40 is considered as positive [Reference Jang89, Reference Lai90]. However, different cut-off values have been reported for IgM and IgG isotypes, such as ⩾1:128 for IgG and ⩾1:64 for IgM in Thailand [Reference Bhengsri3]. Interestingly, Denmark has used a cutoff as high as 1:512 [Reference Kantso79]. There is a need to standardize diagnostic cut-offs for seroprevalence studies to allow easier comparability of results.

Conclusions

SFG Rickettsia causes a large number of emerging and re-emerging infectious diseases with a worldwide distribution. Its occurrence in LMICs exacerbates an already problematic diagnostic issue. With a limited range of suitable sensitive and specific tests available, the additional compounding factor of the need for cheap and easy to use diagnostic tests inputs additional burden on the populations exposed to these pathogens. Despite many tests being available, their lack of suitability for use in resource-limited regions is of concern, as many require technical expertise, expensive equipment and reagents. In addition, many existing diagnostic tests still require rigorous validation in the regions and populations where these tests may be used, in particular to establish coherent and worthwhile cut-offs. Possibly the best strategy would be to use a qPCR and IFA in tandem whereby, if the specimen is collected early enough in the infection where there will be no antibodies, there is a greater chance of a PCR-positive result. Conversely, if there are detectable antibodies, it is less likely that there will be a positive PCR result. We are in an age when more and more novel diagnostic tests are coming onto the market, and we need to ensure that these tests are suitable and appropriate for the diagnosis of rickettsial diseases, especially in low-income countries.

Acknowledgements

Stuart D. Blacksell and Matthew T. Robinson are supported by the Wellcome Trust of the United Kingdom.

References

1.Parola, P et al. (2013) Update on tick-borne rickettsioses around the world: a geographic approach. Clinical Microbiology Reviews 26, 657702.Google Scholar
2.Acestor, N et al. (2012) Mapping the aetiology of non-malarial febrile illness in Southeast Asia through a systematic review – terra incognita impairing treatment policies. PLoS One 7, e44269.Google Scholar
3.Bhengsri, S et al. (2016) Sennetsu neorickettsiosis, spotted fever group, and typhus group rickettsioses in three provinces in Thailand. American Journal of Tropical Medicine and Hygiene 95, 4349.Google Scholar
4.Aung, AK et al. (2014) Rickettsial infections in Southeast Asia: implications for local populace and febrile returned travellers. American Journal of Tropical Medicine and Hygiene 91, 451460.Google Scholar
5.Rodkvamtook, W et al. (2013) Scrub typhus outbreak, northern Thailand, 2006–2007. Emerging Infectious Diseases 19, 774777.Google Scholar
6.Newton, PN et al. (2019) A prospective, open-label, randomized trial of doxycycline versus azithromycin for the treatment of uncomplicated murine typhus. Clinical Infectious Diseases 68, 738747.Google Scholar
7.Vallee, J et al. (2010) Contrasting spatial distribution and risk factors for past infection with scrub typhus and murine typhus in Vientiane City, Lao PDR. PLoS Neglected Tropical Diseases 4, e909.Google Scholar
8.Chikeka, I and Dumler, JS (2015) Neglected bacterial zoonoses. Clinical Microbiology and Infection 21, 404415.Google Scholar
9.Izzard, L et al. (2010) Isolation of a novel Orientia species (O. chuto sp. nov.) from a patient infected in Dubai. Journal of Clinical Microbiology 48, 44044409.Google Scholar
10.Luce-Fedrow, A et al. (2018) A review of scrub typhus (Orientia tsutsugamushi and related organisms): then, now, and tomorrow. Tropical Medicine and Infectious Diseases 3, 8. doi: 10.3390/tropicalmed3010008.Google Scholar
11.Kato, CY et al. (2013) Assessment of real-time PCR assay for detection of Rickettsia spp. and Rickettsia rickettsii in banked clinical samples. Journal of Clinical Microbiology 51, 314317.Google Scholar
12.Graves, S and Stenos, J (2009) Rickettsioses in Australia. Annals of the New York Acadamy of Sciences 1166, 151155.Google Scholar
13.Niang, M et al. (1998) Prevalence of antibodies to Rickettsia conorii, Rickettsia africae, Rickettsia typhi and Coxiella burnetii in Mauritania. European Journal of Epidemiology 14, 817818.Google Scholar
14.Parola, P (2004) Tick-borne rickettsial diseases: emerging risks in Europe. Comparative Immunology, Microbiology and Infectious Diseases 27, 297304.Google Scholar
15.Nanayakkara, DM et al. (2013) Serological evidence for exposure of dogs to Rickettsia conorii, Rickettsia typhi, and Orientia tsutsugamushi in Sri Lanka. Vector Borne and Zoonotic Diseases 13, 545549.Google Scholar
16.Brown, LD and Macaluso, KR (2016) Rickettsia felis, an emerging flea-borne rickettsiosis. Current Tropical Medicine Reports 3, 2739.Google Scholar
17.Luce-Fedrow, A et al. (2015) Strategies for detecting rickettsiae and diagnosing rickettsial diseases. Future Microbiology 10, 537564.Google Scholar
18.Dieme, C et al. (2015) Transmission potential of Rickettsia felis infection by Anopheles gambiae mosquitoes. Proceedings of the National Academy of Sciences. 112, 80888093.Google Scholar
19.Colonne, PM, Eremeeva, ME and Sahni, SK (2011) Beta interferon-mediated activation of signal transducer and activator of transcription protein 1 interferes with Rickettsia conorii replication in human endothelial cells. Infection and Immunity 79, 37333743.Google Scholar
20.Rydkina, E, Turpin, LC and Sahni, SK (2010) Rickettsia rickettsii infection of human macrovascular and microvascular endothelial cells reveals activation of both common and cell type-specific host response mechanisms. Infection and Immunity 78, 25992606.Google Scholar
21.Gaywee, J et al. (2007) Human infection with Rickettsia sp. related to R. japonica, Thailand. Emerging Infectious Diseases 13, 671673.Google Scholar
22.Young, RP, Ip, M and Bassett, DC (1995) Fatal rickettsial meningitis in Hong Kong: a need for rapid laboratory diagnosis. Scandinavian Journal of Infectious Diseases 27, 527528.Google Scholar
23.Rossio, R et al. (2015) Mediterranean spotted fever and hearing impairment: a rare complication. International Journal of Infectious Diseases 35, 3436.Google Scholar
24.Ben Mansour, N et al. (2014) [Acute myocarditis complicating Mediterranean spotted fever. A case report]. Annales de Cardiologie et d'Angéiologie 63, 5557.Google Scholar
25.Botelho-Nevers, E et al. (2005) Cerebral infarction: an unusual complication of Mediterranean spotted fever. European Journal of Internal Medicine 16, 525527.Google Scholar
26.Kodama, K, Noguchi, T and Chikahira, Y (2000) [Japanese spotted fever complicated by acute respiratory failure]. Kansenshogaku Zasshi 74, 162165.Google Scholar
27.Takiguchi, J et al. (2016) [Severe Japanese spotted fever complicated by acute respiratory failure in Kobe City]. Kansenshogaku Zasshi 90, 120124.Google Scholar
28.McBride, WJ et al. (2007) Severe spotted fever group rickettsiosis, Australia. Emerging Infectious Diseases 13, 17421744.Google Scholar
29.Santibanez, S et al. (2013) Usefulness of rickettsial PCR assays for the molecular diagnosis of human rickettsioses. Enfermedades Infecciosas y Microbiología Clínica 31, 283288.Google Scholar
30.La Scola, B and Raoult, D (1997) Laboratory diagnosis of rickettsioses: current approaches to diagnosis of old and new rickettsial diseases. Journal of Clinical Microbiology 35, 27152727.Google Scholar
31.Kato, C, Chung, I and Paddock, C (2016) Estimation of Rickettsia rickettsii copy number in the blood of patients with Rocky Mountain spotted fever suggests cyclic diurnal trends in bacteraemia. Clinical Microbiology and Infection 22, 394396.Google Scholar
32.Sonthayanon, P et al. (2009) Association of high Orientia tsutsugamushi DNA loads with disease of greater severity in adults with scrub typhus. Journal of Clinical Microbiology 47, 430434.Google Scholar
33.Kondo, M et al. (2015) Improvement in early diagnosis of Japanese spotted fever by using a novel Rick PCR system. Journal of Dermatology 42, 10661071.Google Scholar
34.Bechah, Y, Socolovschi, C and Raoult, D (2011) Identification of rickettsial infections by using cutaneous swab specimens and PCR. Emerging Infectious Diseases 17, 8386.Google Scholar
35.Denison, AM et al. (2014) Detection of Rickettsia rickettsii, Rickettsia parkeri, and Rickettsia akari in skin biopsy specimens using a multiplex real-time polymerase chain reaction assay. Clinical Infectious Diseases 59, 635642.Google Scholar
36.Ishikura, M et al. (2003) Phylogenetic analysis of spotted fever group rickettsiae based on gltA, 17-kDa, and rOmpA genes amplified by nested PCR from ticks in Japan. Microbiology and Immunology 47, 823832.Google Scholar
37.Paris, DH and Dumler, JS (2016) State of the art of diagnosis of rickettsial diseases: the use of blood specimens for diagnosis of scrub typhus, spotted fever group rickettsiosis, and murine typhus. Current Opinions in Infectious Diseases 29, 433439.Google Scholar
38.Prakash, JA et al. (2012) Molecular detection and analysis of spotted fever group Rickettsia in patients with fever and rash at a tertiary care centre in Tamil Nadu, India. Pathogens and Global Health 106, 4045.Google Scholar
39.Roux, V and Raoult, D (2000) Phylogenetic analysis of members of the genus Rickettsia using the gene encoding the outer-membrane protein rOmpB (ompB). International Journal of Systematic Evolution and Microbiology 50, 14491455.Google Scholar
40.Choi, YJ et al. (2005) Evaluation of PCR-based assay for diagnosis of spotted fever group rickettsiosis in human serum samples. Clinical Diagnostic Laboratory Immunology 12, 759763.Google Scholar
41.Marquez, FJ et al. (1998) Genotypic identification of an undescribed spotted fever group Rickettsia in Ixodes ricinus from southwestern Spain. American Journal of Tropical Medicine and Hygiene 58, 570577.Google Scholar
42.Jiang, J et al. (2013) Molecular detection of Rickettsia felis and Candidatus Rickettsia asemboensis in fleas from human habitats, Asembo, Kenya. Vector Borne and Zoonotic Diseases 13, 550558.Google Scholar
43.Jiang, J et al. (2004) Development of a quantitative real-time polymerase chain reaction assay specific for Orientia tsutsugamushi. American Journal of Tropical Medicine and Hygiene 70, 351356.Google Scholar
44.Jiang, J, Stromdahl, EY and Richards, AL (2012) Detection of Rickettsia parkeri and Candidatus Rickettsia andeanae in Amblyomma maculatum Gulf Coast ticks collected from humans in the United States. Vector Borne and Zoonotic Diseases 12, 175182.Google Scholar
45.Taylor, AJ et al. (2016) Large-scale survey for tickborne bacteria, Khammouan Province, Laos. Emerging Infectious Diseases 22, 16351639.Google Scholar
46.Maina, AN et al. (2017) Molecular characterization of novel mosquito-borne Rickettsia spp. from mosquitoes collected at the Demilitarized Zone of the Republic of Korea. PLoS One 12, e0188327.Google Scholar
47.Massung, RF et al. (2001) Epidemic Typhus Meningitis in the Southwestern United States. Clinical Infectious Diseases 32, 979982.Google Scholar
48.Tzianabos, T, Anderson, BE and McDade, JE (1989) Detection of Rickettsia rickettsii DNA in clinical specimens by using polymerase chain reaction technology. Journal of Clinical Microbiology 27, 28662868.Google Scholar
49.Paddock, CD et al. (2004) Rickettsia parkeri: a newly recognized cause of spotted fever rickettsiosis in the United States. Clinical Infectious Diseases 38, 805811.Google Scholar
50.Webb, L et al. (1990) Detection of murine typhus infection in fleas by using the polymerase chain reaction. Journal of Clinical Microbiology 28, 530534.Google Scholar
51.Roux, V et al. (1997) Citrate synthase gene comparison, a new tool for phylogenetic analysis, and its application for the rickettsiae. International Journal of Systematic Bacteriology l 47, 252261.Google Scholar
52.Jiang, J et al. (2005) Human Infection with Rickettsia honei, Thailand. Emerging Infectious Diseases 11, 14731475.Google Scholar
53.Regnery, RL, Spruill, CL and Plikaytis, BD (1991) Genotypic identification of rickettsiae and estimation of intraspecies sequence divergence for portions of two rickettsial genes. Journal of Bacteriology 173, 15761589.Google Scholar
54.Labruna, MB et al. (2004) Rickettsia species infecting Amblyomma cooperi ticks from an area in the state of Saõ Paulo, Brazil, where Brazilian spotted fever is endemic. Journal of Clinical Microbiology 42, 9098.Google Scholar
55.Stenos, J, Graves, SR and Unsworth, NB (2005) A highly sensitive and specific real-time PCR assay for the detection of spotted fever and typhus group Rickettsiae. American Journal of Tropical Medicine and Hygiene 73, 10831085.Google Scholar
56.Fournier, PE and Raoult, D (2009) Current knowledge on phylogeny and taxonomy of Rickettsia spp. Annals of the New York Acadamy of Sciences 1166, 111.Google Scholar
57.Eremeeva, ME, Dasch, GA and Silverman, DJ (2003) Evaluation of a PCR assay for quantitation of Rickettsia rickettsii and closely related spotted fever group rickettsiae. Journal of Clinical Microbiology 41, 54665472.Google Scholar
58.Wikswo, ME et al. (2008) Detection and identification of spotted fever group rickettsiae in Dermacentor species from southern California. Journal of Medical Entomology 45, 509516.Google Scholar
59.Jiang, J et al. (2005) Phylogenetic analysis of a novel molecular isolate of spotted fever group Rickettsiae from northern Peru: Candidatus Rickettsia andeanae. Annals of the New York Acadamy of Sciences 1063, 337342.Google Scholar
60.Pan, L et al. (2012) Rapid, simple, and sensitive detection of the ompB gene of spotted fever group rickettsiae by loop-mediated isothermal amplification. BMC Infectious Diseases 12, 254.Google Scholar
61.Raoult, D and Dasch, GA (1995) Immunoblot cross-reactions among Rickettsia, Proteus spp. and Legionella spp. in patients with Mediterranean spotted fever. FEMS Immunology and Medical Microbiology 11, 1318.Google Scholar
62.McQuiston, JH et al. (2014) Inadequacy of IgM antibody tests for diagnosis of Rocky Mountain Spotted Fever. American Journal of Tropical Medicine and Hygiene 91, 767770.Google Scholar
63.Cruickshank, R (1927) The Weil-Felix reaction in typhus fever. Journal of Hygiene (Lond) 27, 6469.Google Scholar
64.Amano, K et al. (1992) Serological studies of antigenic similarity between Japanese spotted fever rickettsiae and Weil-Felix test antigens. Journal of Clinical Microbiology 30, 24412446.Google Scholar
65.Kaplan, JE and Schonberger, LB (1986) The sensitivity of various serologic tests in the diagnosis of Rocky Mountain spotted fever. American Journal of Tropical Medicine and Hygiene 35, 840844.Google Scholar
66.Kularatne, SA and Gawarammana, IB (2009) Validity of the Weil-Felix test in the diagnosis of acute rickettsial infections in Sri Lanka. Transactions of the Royal Society of Tropical Medicine and Hygiene 103, 423424.Google Scholar
67.Tay, ST and Rohani, MY (2002) The use of the indirect immunoperoxidase test for the serodiagnosis of rickettsial diseases in Malaysia. Southeast Asian Journal of Tropical Medicine and Public Health 33, 314320.Google Scholar
68.Premaratna, R et al. (2008) Rickettsial infections and their clinical presentations in the Western Province of Sri Lanka: a hospital-based study. International Journal of Infectious Diseases s 12, 198202.Google Scholar
69.Newhouse, VF et al. (1979) A comparison of the complement fixation, indirect fluorescent antibody, and microagglutination tests for the serological diagnosis of rickettsial diseases. American Journal of Tropical Medicine and Hygiene 28, 387395.Google Scholar
70.Shirai, A, Dietel, JW and Osterman, JV (1975) Indirect hemagglutination test for human antibody to typhus and spotted fever group rickettsiae. Journal of Clinical Microbiology 2, 430437.Google Scholar
71.Clements, ML et al. (1983) Serodiagnosis of Rocky Mountain spotted fever: comparison of IgM and IgG enzyme-linked immunosorbent assays and indirect fluorescent antibody test. Journal of Infectious Diseases 148, 876880.Google Scholar
72.Rathi, NB et al. (2011) Rickettsial diseases in central India: proposed clinical scoring system for early detection of spotted fever. Indian Pediatrics 48, 867872.Google Scholar
73.Kalal, BS et al. (2016) Scrub typhus and spotted fever among hospitalised children in South India: Clinical profile and serological epidemiology. Indian Journal of Medical Microbiology 34, 293298.Google Scholar
74.Trung, NV et al. (2017) Seroprevalence of scrub typhus, typhus, and spotted fever among rural and urban populations of Northern Vietnam. American Journal of Tropical Medicine and Hygiene 96, 10841087.Google Scholar
75.Khan, SA et al. (2016) Seroepidemiology of rickettsial infections in Northeast India. Transactions of the Royal Society of Tropical Medicine and Hygiene 110, 487494.Google Scholar
76.Tsai, KH et al. (2009) African tick bite Fever in a Taiwanese traveller returning from South Africa: molecular and serologic studies. American Journal of Tropical Medicine and Hygiene 81, 735739.Google Scholar
77.Salgado Lynn, M et al. (2018) Spotted fever rickettsiosis in a wildlife researcher in Sabah, Malaysia: a case study. Tropical Medicine and Infectious Diseases 3, 2936.Google Scholar
78.Ammerman, NC, Beier-Sexton, M and Azad, AF (2008) Laboratory maintenance of Rickettsia rickettsii. Current Protocols in Microbiology 11, 1:3A.5.13A.5.21.Google Scholar
79.Kantso, B et al. (2009) Evaluation of serological tests for the diagnosis of rickettsiosis in Denmark. Journal of Microbiological Methods 76, 285288.Google Scholar
80.Keysary, A and Strenger, C (1997) Use of enzyme-linked immunosorbent assay techniques with cross-reacting human sera in diagnosis of murine typhus and spotted fever. Journal of Clinical Microbiology 35, 10341035.Google Scholar
81.Ormsbee, R et al. (1978) Antigenic relationships between the typhus and spotted fever groups of rickettsiae. American Journal of Epidemiology 108, 5359.Google Scholar
82.Uchiyama, T et al. (1995) Cross-reactivity of Rickettsia japonica and Rickettsia typhi demonstrated by immunofluorescence and Western immunoblotting. Microbiology and Immunology 39, 951957.Google Scholar
83.Hechemy, KE et al. (1989) Cross-reaction of immune sera from patients with rickettsial diseases. Journal of Medical Microbiology 29, 199202.Google Scholar
84.Kidd, L et al. (2008) Evaluation of conventional and real-time PCR assays for detection and differentiation of Spotted Fever Group Rickettsia in dog blood. Veterinary Microbiology 129, 294303.Google Scholar
85.Znazen, A et al. (2015) Comparison of two quantitative real time PCR assays for Rickettsia detection in patients from Tunisia. PLoS Neglected Tropical Diseases 9, e0003487.Google Scholar
86.Camer, GA et al. (2003) Detection of antibodies against spotted fever group Rickettsia (SFG), typhus group Rickettsia (TGR), and Coxiella burnetii in human febrile patients in the Philippines. Jpn Journal of Infectious Diseases 56, 2628.Google Scholar
87.Premaratna, R et al. (2014) Rickettsial infection among military personnel deployed in Northern Sri Lanka. BMC Infectious Diseases 14, 3864.Google Scholar
88.Faruque, LI et al. (2017) Prevalence and clinical presentation of Rickettsia, Coxiella, Leptospira, Bartonella and chikungunya virus infections among hospital-based febrile patients from December 2008 to November 2009 in Bangladesh. BMC Infectious Diseases 17, 141.Google Scholar
89.Jang, WJ et al. (2005) Seroepidemiology of spotted fever group and typhus group rickettsioses in humans, South Korea. Microbiology and Immunology 49, 1724.Google Scholar
90.Lai, CH et al. (2014) Human spotted fever group rickettsioses are underappreciated in southern Taiwan, particularly for the species closely-related to Rickettsia felis. PLoS One 9, e95810.Google Scholar
Figure 0

Fig. 1. Features and temporal aspects of SFG Rickettsia infection and diagnosis.

Figure 1

Table 1. Summary of common SFG PCRs used for diagnosis and sequencing for species identification purposes

Figure 2

Table 2. The comparison of sensitivity and specificity of serological techniques for SFG infection