Skip to main content
Advertisement
  • Loading metrics

An integrated overview of the bacterial flora composition of Hyalomma anatolicum, the main vector of CCHF

  • Nayyereh Choubdar,

    Roles Conceptualization, Formal analysis, Methodology, Software, Visualization, Writing – original draft

    Affiliation Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran

  • Fateh Karimian,

    Roles Formal analysis, Methodology, Software

    Affiliation Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran

  • Mona Koosha,

    Roles Formal analysis, Methodology, Software

    Affiliation Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran

  • Mohammad Ali Oshaghi

    Roles Data curation, Funding acquisition, Investigation, Resources, Supervision, Validation, Writing – review & editing

    moshaghi@sina.tums.ac.ir

    Affiliation Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran

Abstract

The microbial flora associated with Hyalomma anatolicum ticks was investigated using culture-dependent (CD) and independent (next generation sequencing, NGS) methods. The bacterial profiles of different organs, development stages, sexes, and of host cattle skins were analyzed using the CD method. The egg and female gut microbiota were investigated using NGS. Fourteen distinct bacterial strains were identified using the CD method, of which Bacillus subtilis predominated in eggs, larval guts and in adult female and male guts, suggesting probable transovarial transmission. Bacillus velezensis and B. subtilis were identified in cattle skin and tick samples, suggesting that skin is the origin of tick bacteria. H.anatolicum males harbour lower bacterial diversity and composition than females. The NGS analysis revealed five different bacterial phyla across all samples, Proteobacteria contributing to >95% of the bacteria. In all, 56611sequences were generated representing 6,023 OTUs per female gut and 421 OTUs per egg. Francisellaceae family and Francisella make up the vast majority of the OTUs. Our findings are consistent with interference between Francisella and Rickettsia. The CD method identified bacteria, such B. subtilis that are candidates for vector control intervention approaches such paratransgenesis whereas NGS revealed high Francisella spp. prevalence, indicating that integrated methods are more accurate to characterize microbial community and diversity.

Author summary

Crimean-Congo hemorrhagic fever (CCHF) is a viral disease transmitted by hard ticks (Ixodidae: Ixodida) that has a 10–40% fatality. While more than 3 billion people are at risk acquiring the disease, on the order of 10,000–15,000 people are infected and over 500 people die every year from the disease. The main vector and reservoir of the CCHF virus are ticks of the genus Hyalomma. As the microbiota influences vector competence, its manipulation may be used for vector control. In this study, we characterize cultivable and non-cultivable bacteria from H. anatolicum ticks. Field collected samples were processed for aerobic culture and 16s rRNA Next Generation Sequencing (NGS). The cultivable bacterium Bacillus subtilis was found in all samples analysed, including H. anatolicum males, females, larval guts and host skin. NGS revealed Francisella spp as the most common non-cultivable bacteria in H. anatolicum ticks. Bacterial OTUs varied between eggs and female guts, indicating that community structure varies by tick developmental stage. Our results indicate that B. subtilis is be a potential paratransgenesis candidate for vector control intervention. Integrated methods (bacteria culture, NGS) are recommended for more accurate characterization of microbial community and diversity.

Introduction

Tick-borne diseases (TBDs) are imposing a growing burden for human and animal health worldwide. Ticks are obligate blood-feeders and can transmit to humans and animals a wide variety of pathogenic agents such as arboviruses, bacteria, and parasites. Hard ticks (Ixodidae) typically have three life stages (larva, nymph, adult) and feed on distinct host species at each developmental stage, making them important sources of zoonotic diseases [1].

Hyalomma anatolicum (Acari: Ixodidae) is the most important tick involved in transmission of the Crimean-Congo hemorrhagic fever (CCHF) virus in Iran [24]. In addition of CCHF, H. anatolicum is also a vector of a wide variety of agents such as Thogoto virus, Wad Medani virus (WMV), Theileria sp., Ehrlichia sp., Rickettsia sp., Babesia ovis [513], causing transitory lameness [14] and Coxiella burnetii [15].

Symbiotic and commensal microbes can confer numerous unfavourable, neutral, or beneficial effects to their arthropod hosts, and can play several roles in fitness, development, nutritional adaptation, oviposition, egg hatching, larval survival, reproduction, defence against environmental stress, and immunity [1624]. Non-pathogenic microbes may also play a role in transmission of tick-borne pathogens (TBP), with many possible consequences for both animal and human health [25].

The hard tick midgut is colonized by symbiotic, environmentally acquired, and maternally transmitted bacteria. Characterization of H. anatolicum microbiota requires the isolation of the natural bacteria via culture. The cultivable bacteria may be used for vector control interventions such as paratransgenic and RNAi approaches [26,27] that may be explored to render ticks refractory to pathogen infection. On the other hand, non-cultivable bacteria are important components of the tick microbiome. They include endosymbionts beneficial for tick survival such as Coxiella spp., [2830], Rickettsia spp., [3134], Francisella spp., [33,35] and “Candidatus Midichloria mitochondrii” [36], and pathogenic bacteria such as Anaplasma, Borrelia, Ehrlichia, Francisella and Rickettsia [3740]. The introduction of next-generation sequencing (NGS) technologies has permitted the rapid and economic characterization of these microbial communities [37] in contrast to the previously used Sanger sequencing. Recently the number of studies using NGS to investigate the microbial diversity and composition of ticks has expanded [24,37]. There are nine hyper variable regions (V1-V9) of the bacterial 16S ribosomal RNA gene (16S) that can be used to identify bacterial taxa, the V1-V3, V3-V5, V4-V5 regions being the most commonly used. The 454 (Roche) pyrosequencing, Ion Torrent (Thermo Fisher) sequencing by semiconductor ion detection and Illumina MiSeq platforms using fluorescent dye detection have been the most commonly tick microbiome sequencing methods [37,41,42].

Recently, several reports of microbial composition associated with different development stages, sex, and organs, especially the digestive tract of ticks have been published [37]. However, none addressed the bacterial composition of Hyalomma ticks, the main vector of CCHF virus. Two studies assayed H. asiaicum RNA virus infection [43,44]. The aim of this study is to report characterization of the microbiome of H. anatolicum ticks and their host’s skins using culture-dependent and NGS approaches to identify potential bacterial candidate/s for use with paratransgenesis or RNAi approaches.

Methods

Ethics statement

This study followed the guidelines of the institutional ethical committee (Tehran University of Medical Sciences, TUMS). The protocols were approved by TUMS ethical committee under registry IR.TUMS.SPH.REC.1395.926.

Tick collection and identification

This study was carried out in two closely Crimean-Congo Hemorrhagic Fever (CCHF)-endemic districts (Sarbaz and Chabahar) located in south-east corner (Sistan and Baluchestan Province) of Iran (Fig 1). Sistan and Baluchestan is one of the largest provinces of Iran (181,785 km2) that borders Pakistan and Afghanistan and has subtropical climate. Hyalomma ticks were collected from cattle from this region and kept alive in separate sterile 50 ml Falcon tubes containing a piece of filter paper until their dissection. They were transferred to the laboratory of insect molecular biology, School of Public Health, Tehran University of Medical Sciences (SPH-TUMS). Ticks were identified morphologically to the species level using taxonomic keys [45,46]. About 20% (n = 70) female ticks were selected randomly for NGS analysis. Subsets of engorged females (n = 120) were allowed to lay eggs, and 170 eggs were used for either CD or NGS bacterial analysis (Table 1).

thumbnail
Table 1. Details of the H. anatolicum specimens collected for microbiome analysis.

https://doi.org/10.1371/journal.pntd.0009480.t001

Specimen processing and isolation of midgut

A total 630 H. anatolicum ticks, including adults (n = 503) and larvae (n = 127), were identified and used for bacterial isolation (Table 1). Ticks were individually surface sterilized as described by Portillo et al [47]. Briefly, tick was immersed for 5 min in 70% ethanol and then rinsed with autoclaved double distilled water. Each specimen was then fixed in sterilized paraffin by their legs and UV sterilized under sterile conditions in a Class II biosafety cabinet. Lateral cuts were made with a sterile scalpel and dorsal integument was removed. To understand bacterial diversity, guts and Malpighian tubules (MT) were teased away from other organs using ultra-fine sterile forceps. Between dissections, forceps and scalpel blades were sterilized with bleach (10%) and flame. To reduce laboratory-derived contamination; we used workstations, sterile gloves, pipette tips with filters, and PCR grade RNAse-free water and the experiments performed under laminar flow hoods. Guts and MTs were transferred separately to 100 μL sterile phosphate-buffered saline (PBS) (pH 7.2), and homogenized. Swabbing of the cattle’s ear (the preferred site for tick attachment) was performed after washing with distilled autoclaved water to remove sediment, dirt, and transient bacteria. Swabbing consisted of five strokes along each ear. Swabs were placed directly in Falcon tubes containing brain heart infusion (BHI) broth medium. Guts and MTs were pooled separately according to the sex and development stages, up to 5 individuals per pool. A total of 560 guts or MTs from field-collected H. anatolicum ticks were used for culture-dependent identification and 70 female guts representative of different locations and hosts were analysed by NGS.

A subset of the engorged live female ticks was maintained in the insectarium until oviposition. Engorged females were kept in glass vials at about 70–80% relative humidity and 27–28°C under a photoperiod of 14:10 hours (light: dark) until oviposition. Pools of up to five egg batches were initially washed with distilled water followed by three 70% ethanol washes and then rinsed with distilled water, air-dried and homogenized with glass pestles in 1ml of sterile PBS. The egg homogenates were plated for bacteria. Seventy eggs were also processed for NGS identification. For NGS analysis, due to shortage of funds, we processed only pool female gut and egg samples (Table 1). The final water used for rinsing the cuticles and egg batches were used as negative controls and plated in parallel. To assess the environmental contamination, the cuticles, as an environmental control, was removed from the tick carcass and were subjected to DNA extraction by phenol chloroform method and PCR amplification of 16s rRNA gene. Where the negative control was positive the specimen was eliminated from further analysis. Frequent changes of gloves were used to avoid RNAse-DNAse contamination. Surface sterilization of workstation by bleach (10%) followed by alcohol (70%) was performed prior and after each experiment. Also we have used autoclaved instruments before and after handling each sample, avoid talking, sneezing, and coughing, and touching the area where DNA may exist.

Isolation of bacteria

The culture-dependent method.

A 1 ml of each homogenized pool sample was added to Falcon tubes containing 5 ml of brain heart infusion (BHI) broth and incubated overnight at 37°C and 100 rpm. To obtain single colonies, 100 μl of the overnight cultures were spread onto LB agar plates and incubated at 37°C for 18–24 h. DNA was extracted from individual colonies using either a boiling method (STET buffer) and/or routine phenol/chloroform. Phenol/chloroform DNA extraction method was used for the isolates with hard cell walls that had not yielded proper DNA by the boiling method [48].

The 16S rRNA gene was amplified using forward primer 16suF 5′-GAGTTTGATCCTGGCTCAG-3′ and reverse primer 16suR 5′-GTTACCTTGTTACGACTT-3′ as reported by Weisburg [49] yielding a 1,500 bp fragment. The PCR amplification was performed using Maxime PCR PreMix Kit (i-Taq) in 20 μl reaction mixtures containing 1 μl of 10 μM both forward and reverse primers and 1–2 μl (~0.1 μg) of extracted genomic DNA. Three no-template controls including PCR grade RNAse-free water, the water used for washing specimens following ethanol sterilization, and the sterilized cuticles were used to detect any bacterial and/or DNA contamination in the culture media and amplification reagents.

The PCR reactions were performed under the following conditions: initial denaturation at 94°C for 1 min, followed by 35 cycles of 95°C for 30S, annealing at 57.5°C for 40 s, 72°C for 30 s and a final extension at 72°C for 8 min. The PCR product were fractionated on a 1% agarose gel and visualized using an UV transilluminator. PCR Purification Kit (Qiagen, Germany) was used for the purification of PCR products before sequencing.

All successfully amplified 16S rRNA amplicons were directionally sequenced using the same amplification primers obtained from Bioneer Company (S. Korea). The sequences were analysed using five databases: EzTaxon-e [http://eztaxon-e.ezbiocloud.net], NCBI (16S rRNA sequences) [http://blast.ncbi.nlm.nih.gov/Blast.cgi], NCBI (Nucleotide collection) [http://blast.ncbi.nlm.nih.gov/Blast.cgi], leBIBI [http://umr5558-sud-str1.univ-lyon1.fr/lebibi/lebibi.cgi], and Blast2Tree [http://bioinfo.unice.fr/blast]. Sequence homology analysis was based on the number and quality of nucleotides in a given sequence and setting defaults of the databases such as cultivable and or non-cultivable, type and non-type specimens. In case of discrepancies among different databases, species identifications were based on either the most common nomenclature among the results of the four databases or on the basis of the highest percentage similarity. Sequences have been submitted to GenBank under Accession Numbers MN399911, MN399915-MN399917, MN399925-MN399926, MN399929-MN399930, MN399941, MN399950-MN399951 and MT355659-MT355661.

The culture-independent method.

DNA was extracted from homogenized gut or egg pools using DNA extraction kit (QiAamp DNA micro kit) following the manufacturer’s recommended protocol. DNA was stored at -20°C until used for sequencing.

The 16S rRNA gene hyper variable V3 region PCR amplified using fusion degenerate primers 341F (5’-CCTACGGGAGGCAGCAG -3’) and 518R (5’- ATTACCGCGGCTGCTGG -3’) and was sequenced on an Illumina Miseq platform. The amplified fragment was approximately 342 bp and raw data were screened and assembled by QIIME. The UCLUST method was used to cluster the sequences into Operational Taxonomic Units (OTUs) at an identity threshold of 97%. Each library pool was sequenced on a Junior+ System Genome Sequencer.

Data analysis

Cytoscape Software (http://www.cytoscape.org) was used to visualize bacterial richness and egg and female gut shared bacteria [50]. GraphPad Prism software v.5.00 for Windows (GraphPad, San Diego, USA) was used for graphical representation.

Results

Bacteria composition using a culture-dependent approach

Using a culture-dependent method, a total of 97 bacterial strains were identified from different developmental stages, organs and sexes of the field-collected H. anatolicum and from the skin of their host (S1 Table). Bacteria were plated on BHI agar and identified based on 16S rRNA sequencing. Eleven bacterial strains were recovered from the H. anatolicum guts and three strains from cattle’s skin (Table 2). Except for one Acintobacteria, all strains belong to the Firmicutes phylum. All cultivable bacteria were Gram positive (G+). A majority (10 out of 14, 71.4%) of the G+ strains from H. anatolicum guts and animal skins belong to the Bacillus genus. G+ Paraclustridium, Enterococcus, and Micrococcus were also recovered. In H.anatolicum guts, the species found were Bacillus subtilis, B. licheniformis, B. velezensis, B. oceanisedimini, Micrococcus aoeverae, Enterococcus lactis, and Paraclustridium benzoelyticum. Only two strains of P. benzoelyticum and B. licheniformis were recovered from eggs, the later one also was recovered from female guts. Micrococcus aoeverae was shared between the guts and MTs of ticks. Of the six bacterial species identified in adults, only B. subtilis was shared between the two sexes (Table 2). B. subtilis has also been found in larval guts and in host skin (Table 2). In addition to B. subtilis the microbiome of cattle’s skin included B. velezensis which is also found in tick guts.

thumbnail
Table 2. Bacterial profile of H. anatolicum ticks and their host skin (ear) revealed by culture dependent method.

https://doi.org/10.1371/journal.pntd.0009480.t002

The NGS method was used to characterize the microbiome of field collected H. anatolicum female guts and eggs. A 346 bp fragment of the hypervariable V3 region of the 16S rRNA gene was PCR amplified from the genomic DNA pools (female gut and egg) using specific universal primers and sequenced using the Illumina-MiSeq platform. A total of 56,611 sequences were generated that were classified into 6,023 operational taxonomic units (OTUs) per female gut and 421 OTUs per egg. The following phyla were identified: Proteobacteria, Actinobacteria, Firmicutes, Deinococcus-Thermus and Fusobacteria. The relative abundance of different female and egg bacterial phyla is summarized in Fig 2. The phylum Proteobacteria makes up nearly all the RPA (relative present abundance) and contributed to 94.9%, and 96.1% of the bacterial sequences in eggs and female guts, respectively. These bacteria belonged to 32 families and 39 genera. A total of 24 and 25 genera were found in female guts and egg samples, respectively (Table 3). Next generation sequencing revealed that Francisella makes up the vast majority of the RPA, making up 96.8 and 92.1% of the female gut and egg bacterial community, respectively. The following nine (out of 40) bacterial genera Kocuria, Propionibacterium, Corynebacterium, Staphylococcus, Ochrobactrum, Acinetobacter, Rhizobium, Pseudomonas and Francisella, were shared between the egg and female gut samples (Table 3, Fig 3).

thumbnail
Fig 2. Relative abundance of H. anatolicum gut and egg bacterial phyla revealed by 16S rRNA gene sequencing on the Illumina MiSeq platform.

https://doi.org/10.1371/journal.pntd.0009480.g002

thumbnail
Fig 3. Network analysis showing the shared and non-shared H. anatolicum female gut and egg bacteria genera revealed by NGS.

https://doi.org/10.1371/journal.pntd.0009480.g003

thumbnail
Table 3. Taxonomic, characters, and the number of operational taxonomic units (OTUs) in H. anatolicum eggs and female guts revealed by NGS.

https://doi.org/10.1371/journal.pntd.0009480.t003

Discussion

Results of NGS analysis revealed the presence of endosymbionts such as Francisella spp. and Candidatus, as well as pathogenic, environmental, and skin-associated bacteria in the gut of H. anatolicum. Francisella spp., important tick-borne pathogens (TBPs) of humans and animals, were the dominant bacteria (more than 92% of OTU reads) associated with H. anatolicum guts and eggs. Francisella and Francisella-like endosymbiotic bacteria (FLEs) are transovarially transmitted and potentially obligate endosymbionts. These bacteria have also been identified in Ornithodoros moubata ovaries and Malphigian tubules [51] and in several hard ticks [16,33,35,52,53]. Interestingly, NGS analysis revealed that none of the H. analoticum ticks harboured other known TBPs such as Ehrlichia, Anaplasma, Babesia, and spotted fever group Rickettsia (SFGR). It is known that Rickettsia and Francisella were negatively correlated in the ticks [54] and that Francisella outcompetes other bacterial genera [33]. However, it is noticeable that previous studies have shown the presence of Ehrlichia, Anaplasma, and Babesia in the ticks of our study region [5557], therefore further studies are needed to confirm the hypothesis that FLEs interfere with the ability of Hyalomma ticks to be infected with Ehrlichia, Anaplasma, Babesia and SFGR.

In this study different microbial communities were found between the H. anatolicum gut and MT and the guts and/or MT with eggs studied. This is in agreement with previous studies indicating microbial variation among anatomical regions within the tick such as the reproductive tract, midgut, and the salivary glands [28,34,5760]. NGS analysis revealed considerable differences in the frequency of bacteria in female guts and eggs (6,023 versus 421 OTUs). However, the diversity between the bacterial community of the guts and eggs was not significant (24 versus 25 with 9 shared genera). Culture dependent method revealed a great variation in frequency and diversity of bacteria among gut, egg, and Malpighian tubule (8 versus 2 versus 1). Micrococcus aoeverae was the only Malpighian tubule bacterium also found in guts, suggesting it is exceptional in its capacity to migrate from midgut to Malpighian tubules, and colonize in this organ. In addition, some bacteria were shared between eggs and guts, indicating possible transovarial transmission from females to eggs and presumably to the next generation.

It seems that the location have effect on the results of tick bacterial community where no tick associated with B. subtilis in Chabahar district, despite this bacterium being found in the skin of cattle in the region. Further field studies are required to verify these preliminary findings. On the other hand, although all of the controls which were used in this study were not environmentally contaminated, the use of 70% ethanol for 5 min, as the only method used in this study, may not be effective enough especially for B. subtilis spores.

Bacillus licheniformis, found in H. anatolicum eggs, produces microbial polysaccharides with multiple bioactivity including antibiofilm activity against Gram-negative (Pseudomonas aeruginosa and Proteus vulgaris) bacteria, Candida albicans, and mosquito larvae [61]. This may partially explain presence of only gram positive bacteria in the culture media of our study. Microbial polysaccharide insect toxicity may play a role in protection of tick eggs against insect predators. These observations deserve further consideration for entomological applications of this bacterium species.

Among environmental and host-related factors that may influence diversity and composition of the H. anatolicum microbiome, we have assessed the effect of sex, organs, and developmental stages. The CD method showed that H. anatolicum male guts harbour lower microbiome diversity and composition than that of females (2 versus 6). Only B. subtilis was shared between the two sexes. It has been reported that females have higher [30,62] or lower [34,54,63] relative bacterial abundance than of males, while other researchers indicated that males and females adults differed only in their community structure, for example, males containing more Rickettsia and females containing more Coxiella [64]. These data suggest that tick microbial community is dynamic.

Our NGS analysis identified pathogenic bacteria associated with H. anatolicum ticks. However, these ticks may harbour additional yet-undiscovered human or animal pathogens and pathogenicity of such bacteria remains to be determined. The fact that H. anatolicum is among the most frequent ticks that come in contact with humans and cattle in Iran [4,65] emphasizes the need to characterize all H. anatolicum-associated microbes to determine the full spectrum of agents that can be transmitted by this tick.

There is current interest in the use of microorganisms as biological control agents of vector borne diseases [66]. Strategies could be developed to manipulate the certain components of the tick microbiome to decrease the vectorial capacity of ticks by hindering pathogen acquisition, development, and horizontal and vertical transmission. Similar microbial management strategies could be developed for ticks which promote the growth of endosymbiotic bacteria to reduce the acquisition of pathogens. Here, we have isolated a strain of the non-pathogenic Bacillus species (B. subtilis) from H. anatolicum, which was previously introduced as a promising candidate for paratransgenic approach [67,68]. We have identified B. subtilis in H. anatolicum eggs, female midguts, males, larvae, as well as cattle’s skin. Moreover, it was shown that H. asiaticum ticks can acquire bacteria from host skin while blood feeding [27]. B. subtilis has been isolated from different arthropods including ticks [69,70], and has potential to be used for control of TBDs. In addition to being non-pathogenic, it is easily cultured and genetically manipulated [67]. The use of symbiotic bacteria expressing dsRNA in a paratransgenic approach is a new method for the control of vector-borne disease [71,72] and has already been used for reducing tick pathogens [7376]. For using B. subtilis for paratransgenic approaches, it will be important to examine its capacity to efficiently colonize the gut, reproductive organs, or salivary glands of Hyalomma spp., and to express enough effector molecules or dsRNA to inhibit the target gene.

Conclusions

The culture-dependent approach revealed a bacterial community diversity comprising gram positive bacteria belonging to mostly Firmicutes phyla, among which B. subtilis was the dominant bacterium. Bacillus licheniformis was isolated from eggs and female guts suggesting possible transovarial transmission as well as protective role against insect predators. However, other tick tissues, especially ovaries, should be analysed to support this premise. Presence of B. subtilis in the guts of females, males, and larvae of H. anatolicum ticks as well as their host’s skin suggests that this bacterial species is a potential candidate for paratransgenic and RNAi approaches for prevention of TBPs transmission. High frequency of Francisella and lack of Rickettsia genus is in agreement with that microbe-microbe interactions phenomena and their influence on microbiome composition and interfere with TBP transmission. Finally integration of culture-dependent and culture–independent method provides better understanding and more extensive and accurate results in terms of the microbial community of vector microbiome.

Supporting information

S1 Table. Details of the bacteria isolated from H. anatolicum ticks and their host skin revealed by culture dependent method followed by 16sRNA gene sequencing.

https://doi.org/10.1371/journal.pntd.0009480.s001

(DOCX)

Acknowledgments

Authors would like to express our deep and sincere gratitude to Prof. Marcelo Jacob Lorena from John Hopkins Bloomberg School of Public Health for providing valuable reviews to the manuscript.

References

  1. 1. Kaufman WR. Ticks: physiological aspects with implications for pathogen transmission. Ticks Tick-Borne Dis 2010; 1(1): 11–22. pmid:21771507
  2. 2. Telmadarraiy Z, Chinikar S, Vatandoost H, Faghihi F, Hosseini-Chegeni A. Vectors of Crimean Congo hemorrhagic fever virus in Iran. J Arthropod Borne Dis 2015; 9(2): 137–47. pmid:26623426
  3. 3. Mohammadian M, Chinikar S, Telmadarraiy Z, Vatandoost H, Oshaghi MA, Hanafi-Bojd AA, et al. Molecular assay on Crimean Congo hemorrhagic fever virus in ticks (Ixodidae) collected from Kermanshah Province, western Iran. J Arthropod Borne Dis 2016; 10(3): 381–91. pmid:27308296
  4. 4. Choubdar N, Oshaghi MA, Rafinejad J, Pourmand MR, Maleki-Ravasan N, Salehi-Vaziri M, et al. Effect of Meteorological Factors on Hyalomma Species Composition and Their Host Preference, Seasonal Prevalence and Infection Status to Crimean-Congo Haemorrhagic Fever in Iran. J Arthropod Borne Dis 2019; 13(3): 268–83. pmid:31879667
  5. 5. Abdigoudarzi M. Detection of naturally infected vector ticks (Acari: Ixodidae) by different species of Babesia and Theileria agents from three different enzootic parts of Iran. J Arthropod Borne Dis 2013; 7(2): 164–72. pmid:24409442
  6. 6. Tavassoli M, Tabatabaei M, Nejad B, Tabatabaei M, Najafabadi A, Pourseyed S. Detection of Theileria annulata by the PCR-RFLP in ticks (Acari, Ixodidae) collected from cattle in West and North-West Iran. Acta Parasitol 2011; 56(1): 8–13.
  7. 7. Nasser H-R, Saeed H, Mohammad A. Molecular detection of Theileria ovis and T. lestoquardi in vector ticks in Lorestan province, Iran. Int J Biosci 2014; 4(12): 78–83.
  8. 8. Špitalská E, Boldiš V, Derdáková M, Selyemová D, Tarageľová VR. Rickettsial infection in Ixodes ricinus ticks in urban and natural habitats of Slovakia. Ticks Tick-Borne Dis 2014; 5(2): 161–5. pmid:24342052
  9. 9. Kayedi MH, Chinikar S, Mostafavi E, Khakifirouz S, Jalali T, Hosseini-Chegeni A, et al. Crimean–Congo hemorrhagic fever virus clade iv (Asia 1) in ticks of western Iran. J Med Entomol 2015; 52(5): 1144–9. pmid:26336221
  10. 10. Sohrabi S, Yakhchali M, Ghashghaei O. PCR-RELP for detecting of Theileria annulata infection in cattle and Hyalomma species in Kermanshah Province, Iran. Archives of Razi Institute 2015; 70(1): 7–12.
  11. 11. Hosseini-Chegeni A, Tavakoli M, Telmadarraiy Z. The updated list of ticks (Acari: Ixodidae & Argasidae) occurring in Iran with a key to the identification of species. Syst Appl Acarol 2019; 24(11): 2133–66.
  12. 12. Zaid T, Ereqat S, Nasereddin A, Al-Jawabreh A, Abdelkader A, Abdeen Z. Molecular characterization of Anaplasma and Ehrlichia in ixodid ticks and reservoir hosts from Palestine: a pilot survey. Vet Med Sci 2019; 5(2): 230–42. pmid:30762295
  13. 13. Abdigoudarzi M, Esmaeilnia K, Shariat N. Laboratory study on biological control of ticks (Acari: Ixodidae) by entomopathogenic indigenous fungi (Beauveria bassiana). J Arthropod Borne Dis 2009; 3(2): 36–43.
  14. 14. Azizi S, Yakhchali M. Transitory lameness in sheep due to Hyalomma spp. infestation in Urmia, Iran. Small Ruminant Res 2006; 63(3): 262–4.
  15. 15. Fard SN, Khalili M. PCR-detection of Coxiella burnetii in ticks collected from sheep and goats in southeast Iran. Iran J Arthropod-borne Dis 2011; 5(1): 1. pmid:22808404
  16. 16. Narasimhan S, Fikrig E. Tick microbiome: the force within. Trends Parasitol 2015; 31(7): 315–23. pmid:25936226
  17. 17. Zhong J, Jasinskas A, Barbour AG. Antibiotic treatment of the tick vector Amblyomma americanum reduced reproductive fitness. PloS One 2007; 2(5). pmid:17476327
  18. 18. McMeniman CJ, Lane RV, Cass BN, Fong AW, Sidhu M, Wang Y-F, et al. Stable introduction of a life-shortening Wolbachia infection into the mosquito Aedes aegypti. Science 2009; 323(5910):141–4. pmid:19119237
  19. 19. Eappen AG, Smith RC, Jacobs-Lorena M. Enterobacter-activated mosquito immune responses to Plasmodium involve activation of SRPN6 in Anopheles stephensi. Plos One 2013; 8(5). pmid:23658788
  20. 20. Minard G, Mavingui P, Moro CV. Diversity and function of bacterial microbiota in the mosquito holobiont. Parasit Vectors 2013; 6(1): 146. pmid:23688194
  21. 21. Narasimhan S, Rajeevan N, Liu L, Zhao YO, Heisig J, Pan J, et al. Gut microbiota of the tick vector Ixodes scapularis modulate colonization of the Lyme disease spirochete. Cell Host Microbe 2014; 15(1): 58–71. pmid:24439898
  22. 22. Jupatanakul N, Sim S, Dimopoulos G. The insect microbiome modulates vector competence for arboviruses. Viruses 2014; 6(11): 4294–4313. pmid:25393895
  23. 23. Coon KL, Vogel KJ, Brown MR, Strand MR. Mosquitoes rely on their gut microbiota for development. Mol Ecol 2014; 23(11): 2727–39. pmid:24766707
  24. 24. Couper LI, Kwan JY, Ma J, Swei A. Drivers and patterns of microbial community assembly in a Lyme disease vector. Ecol Evol 2019; 9(13): 7768–79. pmid:31346439
  25. 25. Bonnet SI, Binetruy F, Hernández-Jarguín AM, Duron O. The tick microbiome: why non-pathogenic microorganisms matter in tick biology and pathogen transmission. Front Cell Infect Microbiol 2017; 7: 236. pmid:28642842
  26. 26. Koosha M, Vatandoost H, Karimian F, Choubdar N, Abai MR, Oshaghi MA. Effect of Serratia AS1 (Enterobacteriaceae: Enterobacteriales) on the Fitness of Culex pipiens (Diptera: Culicidae) for Paratransgenic and RNAi Approaches. J Med Entomol 2019; 56(2): 553–9. pmid:30388221
  27. 27. Koosha M, Vatandoost H, Karimian F, Choubdar N, Oshaghi MA. Delivery of a genetically marked Serratia AS1 to medically important arthropods for use in RNAi and paratransgenic control strategies. Microb Ecol 2019; 78(1): 185–94. pmid:30460544
  28. 28. Andreotti R, de León AAP, Dowd SE, Guerrero FD, Bendele KG, Scoles GA. Assessment of bacterial diversity in the cattle tick Rhipicephalus (Boophilus) microplusthrough tag-encoded pyrosequencing. BMC Microbiol 2011; 11(1): 6. pmid:21211038
  29. 29. Qiu Y, Nakao R, Ohnuma A, Kawamori F, Sugimoto C. Microbial population analysis of the salivary glands of ticks; a possible strategy for the surveillance of bacterial pathogens. PloS one 2014; 9(8). e103961. pmid:25089898
  30. 30. Lalzar I, Harrus S, Mumcuoglu KY, Gottlieb Y. Composition and seasonal variation of Rhipicephalus turanicus and Rhipicephalus sanguineus bacterial communities. Appl Environ Microbiol 2012; 78(12): 4110–6. pmid:22467507
  31. 31. Carpi G, Cagnacci F, Wittekindt NE, Zhao F, Qi J, Tomsho LP, et al. Metagenomic profile of the bacterial communities associated with Ixodes ricinus ticks. PloS one 2011; 6(10). pmid:22022422
  32. 32. Zhang X-C, Yang Z-N, Lu B, Ma X-F, Zhang C-X, Xu H-J. The composition and transmission of microbiome in hard tick, Ixodes persulcatus, during blood meal. Ticks Tick-Borne Dis 2014; 5(6): 864–70. pmid:25150725
  33. 33. Budachetri K, Browning RE, Adamson SW, Dowd SE, Chao C-C, Ching W-M, et al. An insight into the microbiome of the Amblyomma maculatum (Acari: Ixodidae). J Med Entomol 2014; 51(1): 119–29. pmid:24605461
  34. 34. Ponnusamy L, Gonzalez A, Van Treuren W, Weiss S, Parobek CM, Juliano JJ, et al. Diversity of Rickettsiales in the microbiome of the lone star tick, Amblyomma americanum. Appl Environ Microbiol 2014; 80(1): 354–9. pmid:24162580
  35. 35. Hawlena H, Rynkiewicz E, Toh E, Alfred A, Durden LA, Hastriter MW, et al. The arthropod, but not the vertebrate host or its environment, dictates bacterial community composition of fleas and ticks. ISME J 2013; 7(1): 221–3. pmid:22739493
  36. 36. Gofton AW, Oskam CL, Lo N, Beninati T, Wei H, McCarl V, et al. Inhibition of the endosymbiont “Candidatus Midichloria mitochondrii” during 16S rRNA gene profiling reveals potential pathogens in Ixodes ticks from Australia. Parasites Vectors 2015; 8(1): 345. pmid:26108374
  37. 37. Greay TL, Gofton AW, Paparini A, Ryan UM, Oskam CL, Irwin PJ. Recent insights into the tick microbiome gained through next-generation sequencing. Parasites Vectors 2018; 11(1): 12. pmid:29301588
  38. 38. Rafinejad J, Choubdar N, Oshaghi M, Piazak N, Satvat T, Mohtarami F, et al. Detection of Borrelia persica infection in Ornithodoros tholozani using PCR targeting rrs gene and xenodiagnosis. Iran J Public Health 2011; 40(4): 138–45. pmid:23113113
  39. 39. Oshaghi MA, Rafinejad J, Choubdar N, Piazak N, Vatandoost H, Telmadarraiy Z, et al. Discrimination of relapsing fever Borrelia persica and Borrelia microtti by diagnostic species-specific primers and polymerase chain reaction–restriction fragment length polymorphism. Vector Borne Zoonotic Dis 2011; 11(3): 201–7. pmid:20586604
  40. 40. Hosseini-Vasoukolaei N, Oshaghi MA, Shayan P, Vatandoost H, Babamahmoudi F, Yaghoobi-Ershadi MR, et al. Anaplasma infection in ticks, livestock and human in Ghaemshahr, Mazandaran Province, Iran J Arthropod Borne Dis 2014; 8(2): 204–11.
  41. 41. Fadrosh DW, Ma B, Gajer P, Sengamalay N, Ott S, Brotman RM, et al. An improved dual-indexing approach for multiplexed 16S rRNA gene sequencing on the Illumina MiSeq platform. Microbiome 2014; 2(1): 6. pmid:24558975
  42. 42. Akorli J, Gendrin M, Pels NAP, Yeboah-Manu D, Christophides GK, Wilson MD. Seasonality and locality affect the diversity of Anopheles gambiae and Anopheles coluzzii midgut microbiota from Ghana. PloS One 2016; 11(6). pmid:27322614
  43. 43. Li C-X, Shi M, Tian J-H, Lin X-D, Kang Y-J, Chen L-J, et al. Unprecedented genomic diversity of RNA viruses in arthropods reveals the ancestry of negative-sense RNA viruses. elife. 2015; 4: e05378. pmid:25633976
  44. 44. Shi M, Lin X-D, Tian J-H, Chen L-J, Chen X, Li C-X, et al. Redefining the invertebrate RNA virosphere. Nature 2016; 540(7634): 539–43. pmid:27880757
  45. 45. Walker AR. Ticks of domestic animals in Africa: a guide to identification of species: Bioscience Reports Edinburgh; 2003.
  46. 46. Estrada-Peña A, Bouattour A, Camicas J, Walker AR. Tick of domestic animals in Mediterranean region. A guide to identification of species. 2004.
  47. 47. Portillo A, Palomar AM, de Toro M, Santibáñez S, Santibáñez P, Oteo JA. Exploring the bacteriome in anthropophilic ticks: To investigate the vectors for diagnosis. PLoS ONE 2019, 14(3): e0213384. pmid:30889229
  48. 48. Andreou LV. Isolation of plasmid DNA from bacteria. Methods Enzymol 2013; 2(8): 135–42. pmid:24011041
  49. 49. Weisburg WG, Barns SM, Pelletier DA, Lane DJ. 16S ribosomal DNA amplification for phylogenetic study. J Bacteriol 1991; 173(2): 697–703. pmid:1987160
  50. 50. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Research 2003;13(11): 2498–504. pmid:14597658
  51. 51. Burgdorfer W, Owen CR. Experimental studies on argasid ticks as possible vectors of tularemia. Int J Infect Dis 1956: 67–74. pmid:13295627
  52. 52. Scoles GA. Phylogenetic analysis of the Francisella-like endosymbionts of Dermacentor ticks. J Med Entomol 2004; 41(3): 277–86. pmid:15185926
  53. 53. Kugeler KJ, Gurfield N, Creek JG, Mahoney KS, Versage JL, Petersen JM. Discrimination between Francisella tularensis and Francisella-like endosymbionts when screening ticks by PCR. Appl Environ Microbiol 2005; 71(11): 7594–7. pmid:16269811
  54. 54. Gurfield N, Grewal S, Cua LS, Torres PJ, Kelley ST. Endosymbiont interference and microbial diversity of the Pacific coast tick, Dermacentor occidentalis, in San Diego County, California. Peer J 2017; 5: e3202. pmid:28503372
  55. 55. Ranjbar-Bahadori S, Eckert B, Omidian Z, Shirazi NS, Shayan P. Babesia ovis as the main causative agent of sheep babesiosis in Iran. Parasitol Res 2012; 110(4): 1531–6. pmid:21975684
  56. 56. Jafarbekloo A, Bakhshi H, Faghihi F, Telmadarraiy Z, Khazeni A, Oshaghi MA, et al. Molecular detection of Anaplasma and Ehrlichia infection in ticks in borderline of Iran-Afghanistan. J Biomed Sci Eng 2014; 7(11): 919–26.
  57. 57. Choubdar N, Karimian F, Koosha M, Nejati J. Oshaghi M. Hyalomma spp. (Acari: Ixodida) ticks and associated Anaplasma/Ehrlichia spp. in the border region between Iran and Pakistan. Parasites Vectors 2021. (Forthcoming)
  58. 58. Gall CA, Reif KE, Scoles GA, Mason KL, Mousel M, Noh SM, et al. The bacterial microbiome of Dermacentor andersoni ticks influences pathogen susceptibility. ISME J. 2016; 10(8):1846–55. pmid:26882265
  59. 59. Clayton KA, Gall CA, Mason KL, Scoles GA, Brayton KA. The characterization and manipulation of the bacterial microbiome of the Rocky Mountain wood tick, Dermacentor andersoni. Parasites Vectors 2015; 8(1): 632. pmid:26653035
  60. 60. Zolnik CP, Prill RJ, Falco RC, Daniels TJ, Kolokotronis SO. Microbiome changes through ontogeny of a tick pathogen vector. Mol Ecol 2016; 25(19): 4963–77. pmid:27588381
  61. 61. Abinaya M, Vaseeharan B, Divya M, Vijayakumar S, Govindarajan M, Alharbi NS, et al. Structural characterization of Bacillus licheniformis Dahb1 exopolysaccharide—antimicrobial potential and larvicidal activity on malaria and Zika virus mosquito vectors. Environ Sci Pollut Res Int 2018; 25(19): 18604–19. pmid:29704178
  62. 62. Trout Fryxell R, DeBruyn J. The Microbiome of Ehrlichia-Infected and Uninfected Lone Star Ticks (Amblyomma americanum). PLoS One 2016; 11(1).
  63. 63. Van Treuren W, Ponnusamy L, Brinkerhoff RJ, Gonzalez A, Parobek CM, Juliano JJ, et al. Variation in the microbiota of Ixodes ticks with regard to geography, species, and sex. Appl Environ Microbiol 2015; 81(18): 6200–9. pmid:26150449
  64. 64. Williams-Newkirk AJ, Rowe LA, Mixson-Hayden TR, Dasch GA. Characterization of the bacterial communities of life stages of free living lone star ticks (Amblyomma americanum). PloS one. 2014; 9(7).
  65. 65. Biglari P, Bakhshi H, Chinikar S, Belqeiszadeh H, Ghaffari M, Javaherizadeh S, Faghihi F, Telmadarraiy Z. Hyalomma anatolicum as the Main Infesting Tick in an Important Livestock Rearing Region, Central Area of Iran. Iran J Public Health, 2018; 47(5):742–749. pmid:29922618
  66. 66. Thomas MB. Biological control of human disease vectors: a perspective on challenges and opportunities. Biol Control 2018; 63(1): 61–9. pmid:29391855
  67. 67. Hurwitz I, Hillesland H, Fieck A, Das P, Durvasula R. The paratransgenic sand fly: a platform for control of Leishmania transmission. Parasites Vectors. 2011; 4(1): 82. pmid:21595907
  68. 68. Fraihi W, Fares W, Perrin P, Dorkeld F, Sereno D, Barhoumi W, et al. An integrated overview of the midgut bacterial flora composition of Phlebotomus perniciosus, a vector of zoonotic visceral leishmaniasis in the Western Mediterranean Basin. PLoS Negl Trop Dis 2017; 11(3): e0005484. pmid:28355207
  69. 69. Karimian F, Vatandoost H, Rassi Y, Maleki-Ravasan N, Mohebali M, Shirazi MH, et al. Aerobic midgut microbiota of sand fly vectors of zoonotic visceral leishmaniasis from northern Iran, a step toward finding potential paratransgenic candidates. Parasites Vectors 2019; 12(1):1–12. pmid:30606222
  70. 70. Xu X-L, Cheng T-Y, Yang H, Yan F. Identification of intestinal bacterial flora in Rhipicephalus microplus ticks by conventional methods and PCR–DGGE analysis. Exp Appl Acarol 2015; 66(2): 257–68. pmid:25784070
  71. 71. Taracena ML, Oliveira PL, Almendares O, Umaña C, Lowenberger C, Dotson EM, et al. Genetically modifying the insect gut microbiota to control Chagas disease vectors through systemic RNAi. PLoS Negl Trop Dis 2015; 9(2). pmid:25675102
  72. 72. Airs PM, Bartholomay LC. RNA Interference for mosquito and mosquito-borne disease control. Insects 2017; 8(1): 4. pmid:28067782
  73. 73. e la Fuente J, Blouin EF, Manzano-Roman R, Naranjo V, Almazán C, de la Lastra JMP, et al. Functional genomic studies of tick cells in response to infection with the cattle pathogen, Anaplasma marginale. Genomics 2007; 90(6): 712–22. pmid:17964755
  74. 74. Narasimhan S, Sukumaran B, Bozdogan U, Thomas V, Liang X, DePonte K, et al. A tick antioxidant facilitates the Lyme disease agent’s successful migration from the mammalian host to the arthropod vector. Cell Host Microbe 2007; 2(1): 7–18. pmid:18005713
  75. 75. Adamson SW, Browning RE, Budachetri K, Ribeiro JM, Karim S. Knockdown of selenocysteine-specific elongation factor in Amblyomma maculatum alters the pathogen burden of Rickettsia parkeri with epigenetic control by the Sin3 histone deacetylase corepressor complex. PLoS One 2013; 8(11). pmid:24282621
  76. 76. Budachetri K, Karim S. An insight into the functional role of thioredoxin reductase, a selenoprotein, in maintaining normal native microbiota in the Gulf Coast tick (Amblyomma maculatum). Insect Mol Biol 2015; 24(5): 570–81. pmid:26184979