Skip to main content

Chikungunya virus transmission between Aedes albopictus and laboratory mice

Abstract

Background

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus associated with epidemics of acute and chronic arthritic disease in humans. Aedes albopictus has emerged as an important new natural vector for CHIKV transmission; however, mouse models for studying transmission have not been developed.

Methods

Aedes albopictus mosquitoes were infected with CHIKV via membrane feeding and by using infected adult wild-type C57BL/6 mice. Paraffin sections of infected mosquitoes were analysed by immunofluorescent antibody staining using an anti-CHIKV antibody. CHIKV-infected mosquitoes were used to infect adult C57BL/6 and interferon response factor 3 and 7 deficient (IRF3/7-/-) mice.

Results

Feeding mosquitoes on blood meals with CHIKV titres > 5 log10CCID50/ml, either by membrane feeding or feeding on infected mice, resulted in  ≥ 50 % of mosquitoes becoming infected. However, CHIKV titres in blood meals  ≥ 7 log10CCID50/ml were required before salivary glands showed significant levels of immunofluorescent staining with an anti-CHIKV antibody. Mosquitoes fed on blood meals of 7.5 (but not 5.9) log10CCID50/ml were able efficiently to transmit virus to adult C57BL/6 and IRF3/7-/- mice, with the latter mice showing overt signs of arthritis post-infection.

Conclusions

The results provide a simple in vivo model for studying transmission of CHIKV from mosquitoes to mammals and also argue against a resistance barrier to CHIKV infection in adult mice.

Background

Chikungunya virus (CHIKV) belongs to a group of mosquito-borne arthritogenic alphaviruses that include the primarily Australian Ross River and Barmah Forest viruses, the African o’nyong-nyong virus, the Sindbis group of viruses and the South American Mayaro virus [1]. The largest documented outbreak of CHIKV disease ever recorded began in 2004 in Africa and spread across the Indian Ocean to Asia, east to Papua New Guinea and several pacific islands, with small outbreaks also seen in Europe. In late 2013 the epidemic reached the Americas, spreading through the Caribbean, Central and South America, with autochthonous transmission also reported in the USA [2, 3]. Millions of cases have been reported.

The traditional vector for CHIKV has been Aedes aegypti, and this mosquito species was and remains the main vector in East Africa, the Caribbean and South America. However, the recent epidemic was also associated with efficient CHIKV transmission by Aedes albopictus (the so-called Asian tiger mosquito), particularly in the Indian Ocean, West Africa, Europe and Papua New Guinea, with transmission in Asia involving both species. The East/Central/South African (ECSA) genotype of CHIKV developed a mutation in the E1 envelope gene (Alanine 226 to Valine V), which permitted efficient transmission by Aedes albopictus [4, 5], a highly anthropophilic and geographically widespread mosquito species [6].

Herein we explore the requirements for transmission of CHIKV (using a Reunion Island isolate with the A226V mutation) between Aedes albopictus and mice, and provide the parameters required to establish efficient mosquito-mediated transmission to adult wild-type and interferon response factors 3 and 7 deficient (IRF3/7-/-) mice.

Methods

Aedes albopictus mosquitoes

A colony of Aedes albopictus was established from eggs collected on Hammond Island (Torres Strait, Australia) in May 2014, with additional wild-caught mosquitoes included in 2015. Generations 94–98 (counted from 2014) were used in the experiments described herein. The colony was maintained in a climate-controlled insectary at the QIMR Berghofer Medical Research Institute at 27 °C, 70 % relative humidity and 12:12 h light:dark cycling with 30 min crepuscular periods. Eggs were hatched by flooding in rainwater. Larvae were reared in rain water in plastic trays at densities of ≈ 500 larvae per tray. Larvae were fed ground TetraMin Tropical Flakes fish food (Tetra, Melle, Germany) ad libitum. Pupae were collected and placed in a container of rainwater inside a 30 × 30 × 30 cm cage (BugDorm, MegaView Science Education Services Co., Taichung, Taiwan). The cage was provided with 10 % sucrose solution on cotton wool pledgets. Prior to feeding, mosquitoes (5–6 day-old) were deprived of sucrose solution for 24 h. Female mosquitoes were sampled from the cage by placing a bottle of hot water beside one of the cage walls and aspirating females that were probing against the bottle. Female mosquitoes (80–110) were added to each 750 ml plastic containers with gauze lids.

Membrane feeding

Mosquitoes (80–110 per CHIKV dose) were offered defibrinated sheep blood for 1 h (Life Technologies, Mulgrave, VIC, Australia) via a bovine ceacum membrane using an artificial feeding apparatus (kept at 37 °C) as described [7]. The blood meals contained 5-fold serial dilutions of CHIKV stock (LR2006-OPY1; GenBank KT449801 [8] prepared as described [9]) starting at a 1 in 5 dilution. Blood meal titres were determined by CCID50 assays on blood meal samples taken before and after mosquito feeding. Engorged mosquitoes (feeding rate range 15–50 %), anaesthetized with CO2 and placed on a Petri dish on wet ice, were collected and maintained in an environmental chamber (Panasonic, Osaka, Japan) set at 28 °C, 75 % humidity and 12:12 h day:night light schedule with 30 min dawn:dusk periods.

Feeding on CHIKV infected mice

Female C57BL/6 J mice (6–8 weeks) were purchased from Animal Resources Center (Canning Vale, WA, Australia) and were inoculated by needle injection with 2 × 102 or 2 × 104 CCID50 of C6/36-derived Reunion Island isolate of CHIKV (LR2006-OPY1; GenBank KT449801 [8]) s.c. into hind feet as described previously [8, 9]. On days 2, 7 or 10 post-infection, mice (n = 3 per dose and time point) were anesthetized for 30 min with a continuous flow of 3 % isoflurane using a Stinger AAS anesthetic specialist machine (Advanced Anaesthesia Specialists, Gladesville, NSW, Australia) and placed over the gauze of the mosquito containers to allow feeding. Engorged mosquitoes were collected and maintained as above.

Feeding of CHIKV-infected mosquitoes on naïve mice: viraemias, foot measurements and ELISA

Mosquitoes (n = 14–22 per mouse), which had taken a CHIKV-infected blood meal via membrane feeding 7/8 days previously, were allowed to feed on anesthetized naïve female C57BL/6 mice and IRF3/7-/- mice (described previously [10]) (n = 3 per group), with the numbers of engorged and probing mosquitoes noted. Viraemias were determined by CCID50 assays as described [9]. Height and width of feet were measured by digital callipers and expressed as mean of the percentage increases in height x width for each foot as described [8, 10]. Serum anti-CHIKV IgG2c titres were determined by ELISA on day 21 post-infection as described [11].

Mosquito viral titre determination

Viral titres in each individual mosquito were determined 7 days after the blood meal, at which time infection levels reach a plateau [12, 13]. Individual mosquitoes (anesthetized and collected as above) were placed in 2 ml screw cap vials with 4–5 zirconium silica beads and 500 μl of medium [RPMI 1640, 2 % FBS/FGS, 0.25 μg/ml Amphotericin B (Gibco; Thermo Scientific, Waltham, MA, USA) and 10 mM HEPES]. Mosquitoes were homogenized by shaking tubes for 1 min 30 s in a chilled block using a MiniBeadbeater-96 sample homogenizer (Biospec Products, Bartlesville, OK, USA) followed by centrifugation (twice at 17,000× g, 10 min, 4 °C, with tube rotation), and viral titration using CCID50 assays as described [9].

Mosquito immunohistochemistry and staining quantification

Mosquitoes were processed for immunohistochemistry and paraffin sections stained with a mouse anti-CHIKV capsid monoclonal antibody (5.5G9 [14]) and an Alexa Fluor 488 donkey anti-mouse secondary antibody (green), with DNA stained using DAPI (blue). Stained sections were scanned, and staining quantified using Aperio eSlide Manager and ImageScope Viewer software (Aperio). Full details are available in Additional file 1.

Statistics

Statistical analyses were performed using IBM SPSS Statistics (version19). The non-parametric Spearman’s rank correlation test was used to determine the relationship between blood meal titers offered via membrane feeding and the resulting CHIKV titres in the mosquitos. The non-parametric Kolmogorov-Smirnov test was used to compare salivary gland CHIKV staining densities as differences in variance were > 4 [8].

Results

Infection of mosquitoes via membrane feeding versus infected mice

Whether artificial membrane feeding of mosquitoes (usually involving virus inoculated into anti-coagulated bovine or ovine blood [1517]) accurately recapitulates feeding on viraemic animals (and thus represents a realistic methodology for assessing vector competence) remains a subject for investigation [1820]. Aedes albopictus mosquitoes were fed (i) via membrane feeding using a range of virus titres; and (ii) on mice that had received high and low CHIKV inocula (n = 3 per dose) resulting in mean viraemias on day 2 of 6.5 ± 0.5 and 3.5 ± 1.7 log10CCID50/ml, respectively. The percentage of mosquitoes that became infected increased with the blood meal virus titres, with membrane feeding and feeding on mice providing overlapping and broadly comparable results (Fig. 1a). A threshold effect was evident with titres of > 5 log10CCID50/ml needed before ≥ 50 % of mosquitoes become infected (Fig. 1a).

Fig. 1
figure 1

a Membrane and mouse feeding of Aedes albopictus with different titres of CHIKV. Mosquitoes were fed via artificial membrane with ovine blood containing different titres of CHIKV; the membrane blood meal titres represent the mean (and standard deviation, SD) of before and after feeding titres (i.e. the mean and SD of 2 titre determinations; 30 blood-fed mosquitoes were examined for each CHIKV blood meal titre; limit of detection 2 log10CCID50/ml). A different batch of mosquitoes were fed on CHIKV infected mice on day 2 post-infection; 2 groups of three mice were inoculated with 2 × 102 or 2 × 104 log10CCID50/ml CHIKV, with n = 49/50 fed mosquitoes for each group. The mouse blood meal titres represent the mean (and SD) viraemia on day 2 (n = 3) for each group. b CHIKV titres in the mosquitoes. The CHIKV titers of all CHIKV positive mosquitoes from a are shown; blue lines represent means for each blood meal titre. No CHIKV was detected in any mosquitoes fed with a blood meal titre of 4.75 log10CCID50/ml. Whole mosquitoes were homogenized in 0.5 ml of medium and titres determined by standard CCID50 assays. For membrane fed mosquitoes, a Spearman correlation was performed comparing blood meal titres and mosquito titres, with Rho and p values provided. c Quantification of anti-CHIKV staining density. Mosquitoes were fed as in A and 5–8 fed mosquitoes per blood meal dose were examined by immunohistochemistry for CHIKV using an anti-capsid monoclonal antibody and DAPI staining for DNA. Staining areas were quantified by image analysis and expressed as a ratio of CHIKV staining over DAPI staining for each organ/tissue. Mean background staining density in uninfected mosquitoes was 0.004, range 0–0.15). Statistics by Kolmogorov-Smirnov tests: (i) P = 0.047, comparing salivary gland staining for mosquitoes given blood meals containing 7 log10CCID50/ml (n = 7) with staining for those given 5.25/6.25 log10CCID50/ml (staining data for the latter two doses were combined to provide n = 4) and (ii) P = 0.012, comparing salivary gland staining for mosquitoes given blood meals containing 9.5 log10CCID50/ml (n = 7) with staining for those given 5.25/6.25 log10CCID50/ml (n = 4). d Example of whole body section showing IFA staining in: head (H); midgut (M); and salivary glands (S). e-g High resolution images of IFA staining in head, midgut and salivary glands, respectively

A correlation between blood meal titres and virus levels in mosquitoes

Although a relationship between blood meal titres and the percentage of mosquitoes that become infected is well established [18, 21], the relationship between blood meal virus titres and the resulting virus titres in mosquitoes has, to our knowledge, not been investigated for CHIKV, with a relationship established in some but not other systems [19, 2224]. The viral titre of each positive mosquito from Fig. 1a was determined, with the results illustrating a significant correlation (Spearman’s correlation, rho = 0.38, P < 0.001, n = 101) between the blood meal titres and the CHIKV titres in the mosquito, although a 4.25 log10CCID50/ml rise in the former only resulted in a mean ≈ 1 log10CCID50/ml rise in the latter.

Membrane feeding with 6.25 log10CCID50/ml and mouse feeding with 6.5 log10CCID50/ml also produced similar virus titres in the mosquitoes (Fig. 1b), supporting the contention that membrane and mouse feeding provide similar results.

Immunofluorescent antibody staining of CHIKV in mosquitoes

Using a recently developed monoclonal antibody recognizing the CHIKV capsid protein [14], a group of mosquitoes fed by membrane feeding (as in Fig. 1a, b) were analysed by immunofluorescent antibody staining. The percentage of mosquitoes showing staining above background in at least some area(s) of the different organs/tissues was determined, with broadly similar results for each organ/tissue (Additional file 1: Figure S1). These data correlated well with the data in Fig. 1a and b. However, quantification of the CHIKV staining density (relative to nuclear DNA staining) across the whole organs/tissues, illustrated that pronounced (and significantly increased) staining densities were only observed in mosquitoes fed with blood meals containing viral titres of ≥ 7 log10CCID50/ml (Fig. 1d). In addition, high staining densities were observed in nearly all salivary glands examined in such mosquitoes (Fig. 1d). High staining densities in salivary glands are perhaps consistent with a recent report of replication of CHIKV in the salivary gland of Aedes albopictus [25]. An example of staining of a whole mounted mosquito (Fig. 1e) and the different organs/tissues are shown (Fig. 1f-g).

No infection of mosquitoes with tissue-associated virus post-viraemia

Infection of mosquitoes by arboviruses in the absence of a detectable viraemia has been reported [26]. After the end of the 4–5 day viraemic period, high titres of replication competent CHIKV persist in mouse foot tissues until day 7 [9], with viral RNA persisting for up to 100 days [8]. Mosquitoes were thus allowed to feed on the feet of mice day 7 post-infection, with the feet of anesthetized mice accessible via the mesh in the lid of the mosquito container. The feet were placed through holes in a piece of paper preventing feeding on the mouse body. Although the mean feet tissue titres on day 7 were 6.1 ± 0.9 log10CCID50/mg (n = 3 mice), none of the 85 fed mosquitoes were infected (data not shown). A repeated experiment day 10 post-infection also resulted in none of the 86 fed mosquitoes becoming infected (data not shown).

Mosquito to mouse transmission

Transmission of CHIKV from mosquitoes to mice has, to our knowledge, only been reported for wild-type suckling mice [13, 27]. Mosquito-mediated infection of interferon receptor 3 and 7 deficient (IRF3/7-/-) mice has been reported for dengue virus [28], with IRF3/7-/- mice also highly susceptible to CHIKV infection due to their inability effectively to generate type I interferon responses [10]. Mosquitoes were membrane fed on blood meals (with a CHIKV titre of 7.5 ± 0.35) and left for 8 days (and allowed to lay eggs) and were then fed on the shaved belly area of wild-type C57BL/6 and IRF3/7-/- mice (n = 3 per strain). Whole body CHIKV titres in 10 of these mosquitoes was determined (as in Fig. 1b) to be 6.15 ± 0.58 (SD) log10CCID50/ml, with all 10 mosquitoes CHIKV positive.

Only a small number of mosquitoes took a detectable second blood meal (Table 1), although 1–4 mosquitoes per mouse were seen to probe, with probing previously reported to result in arbovirus inoculation [29]. All mice became viraemic within 2 days and developed CHIKV-specific IgG responses (confirming infection) (Table 1). IRF3/7-/- also developed swollen feet (Table 1; Additional file 1: Figure S2), with 2/3 mice requiring euthanasia (as described previously [10]).

Table 1 Transmission of CHIKV from mosquitoes to mice

In a second experiment, mosquitoes fed with a blood meal of 5.9 ± 0.9 (SD) log10CCID50/ml, after 7 days were allowed to feed on three naïve C57BL/6 mice. Although more mosquitoes were used in this experiment and 10–17 mosquitoes per mouse took a detectable second blood meal, no infection of C57BL/6 mice was detected (data not shown). This is consistent with the data in Fig. 1d showing that mosquitoes fed on a blood meal containing ≤ 6.25 log10CCID50/ml of CHIKV failed to show significant levels of CHIKV in salivary glands.

Discussion

Herein we show for CHIKV and Aedes albopictus that provision of blood meals via membrane feeder or via viraemic mice provided overlapping and broadly comparable results, supporting the view that membrane feeding represents a credible method for assessing vector competence [19, 20]. Furthermore, only blood-borne virus appeared able to transmit to mosquitoes, with (post-viraemic) tissue-associated virus unable to transmit, perhaps because it is not efficiently imbibed and/or because neutralising antibodies (present day 7 post-infection [8]) prevent infection of mosquitoes.

Blood meal titres needed to be > 5 log10CCID50/ml before more than ≥ 50 % of mosquitoes become infected. However, only blood meal titres of ≥ 7 log10CCID50/ml resulted in significant levels of virus in salivary glands, with direct evidence for CHIKV replication in salivary glands recently provided [25]. Virus in the salivary glands is clearly a key requirement for onward transmission to vertebrate hosts, and our observations are consistent with the notion of a dose-dependent barrier to salivary gland infection [30]. Although comparisons are complicated by different methods for quantifying CHIKV titres, the requirement for high titres blood meal (107 pfu/ml) for infecting a high percentage of Aedes albopictus mosquitoes with CHIKV has been reported previously [13, 21], with 107.5 pfu/ml used in another study [12]. Such high titre blood meals were also used to infect mosquitoes that were subsequently used to infect suckling mice [13]. CHIKV viraemias do reach high levels in both mice and humans, albeit only for a few days [10, 31, 32]. However, the full spectrum of inter-relationships between blood meal titres and overt salivary gland infection, and the influence of inter alia time post-feeding, temperature and the presence of other infection(s) in the mosquito, remain to be explored.

This paper represents the first report of infection of adult wild-type mice and IRF3/7-/- mice by CHIKV-infected mosquitoes, providing a convenient new model for studying transmission of CHIKV from mosquitoes to mammalian hosts [3337]. Mosquito-mediated infection of IRF3/7-/- mice with CHIKV also resulted in joint swelling, an arthritic manifestation often seen in symptomatic human CHIKV infections [1]. CHIKV disease manifestations are often more severe in the elderly and the very young [1], populations with compromised type I interferon and/or IRF7 responses [3842]. The rapid appearance of the CHIKV viraemia (within 2 days), in both wild-type and IRF3/7-/- mice post-mosquito feeding, recapitulates the often short incubation period seen for CHIKV infections in humans [1]. The results also argue that the main barrier to transmission is the presence of significant levels of virus in the mosquito salivary glands, rather than the existence of a resistance barrier in adult mice [43, 44].

Conclusion

Feeding Aedes albopictus mosquitoes CHIKV infected blood meals, via a membrane feeder or via infected mice, did not result in marked differences in mosquito infection rates, supporting the view that membrane feeding is a credible method for assessing vector competence. For mosquito salivary glands to become clearly infected, the blood meal titres needed to be ≈ 1–2 logs higher than the titres required simply to infect the mosquitoes. Mosquitoes fed the high titre blood meals were able efficiently to transmit CHIKV to adult mice. The results argue against the presence of a resistance barrier in adult mice and provide a laboratory model for studying transmission of CHIKV from mosquitoes to mammals.

Abbreviations

CCID:

Cell culture infective dose

CHIKV:

Chikungunya virus

IRF:

Interferon response factor

References

  1. Suhrbier A, Jaffar-Bandjee MC, Gasque P. Arthritogenic alphaviruses - an overview. Nat Rev Rheumatol. 2012;8:420–9.

    Article  CAS  PubMed  Google Scholar 

  2. Weaver SC, Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med. 2015;372:1231–9.

    Article  CAS  PubMed  Google Scholar 

  3. Nunes MR, Faria NR, de Vasconcelos JM, Golding N, Kraemer MU, de Oliveira LF, et al. Emergence and potential for spread of chikungunya virus in Brazil. BMC Med. 2015;13:102.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Tsetsarkin KA, McGee CE, Volk SM, Vanlandingham DL, Weaver SC, Higgs S. Epistatic roles of E2 glycoprotein mutations in adaption of chikungunya virus to Aedes albopictus and Ae. aegypti mosquitoes. PLoS One. 2009;4:e6835.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Tsetsarkin KA, Chen R, Yun R, Rossi SL, Plante KS, Guerbois M, et al. Multi-peaked adaptive landscape for chikungunya virus evolution predicts continued fitness optimization in Aedes albopictus mosquitoes. Nat Commun. 2014;5:4084.

    Article  CAS  PubMed  Google Scholar 

  6. Higgs S, Vanlandingham D. Chikungunya virus and its mosquito vectors. Vector Borne Zoonotic Dis. 2015;15:231–40.

    Article  PubMed  Google Scholar 

  7. Rutledge LC, Ward RA, Gould DJ. Studies on the feeding response of mosquitoes to nutritive solutions in a new membrane feeder. Mosq News. 1964;24:407–19.

    Google Scholar 

  8. Poo YS, Rudd PA, Gardner J, Wilson JA, Larcher T, Colle MA, et al. Multiple immune factors are involved in controlling acute and chronic chikungunya virus infection. PLoS Negl Trop Dis. 2014;8:e3354.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Gardner J, Anraku I, Le TT, Larcher T, Major L, Roques P, et al. Chikungunya virus arthritis in adult wild-type mice. J Virol. 2010;84:8021–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rudd PA, Wilson J, Gardner J, Larcher T, Babarit C, Le TT, et al. Interferon response factors 3 and 7 protect against chikungunya virus hemorrhagic fever and shock. J Virol. 2012;86:9888–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang D, Suhrbier A, Penn-Nicholson A, Woraratanadharm J, Gardner J, Luo M, et al. A complex adenovirus vaccine against chikungunya virus provides complete protection against viraemia and arthritis. Vaccine. 2011;29:2803–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dubrulle M, Mousson L, Moutailler S, Vazeille M, Failloux AB. Chikungunya virus and Aedes mosquitoes: saliva is infectious as soon as two days after oral infection. PLoS One. 2009;4:e5895.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Tsetsarkin KA, Vanlandingham DL, McGee CE, Higgs S. A single mutation in chikungunya virus affects vector specificity and epidemic potential. PLoS Pathog. 2007;3:e201.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Goh LY, Hobson-Peters J, Prow NA, Gardner J, Bielefeldt-Ohmann H, Suhrbier A, et al. Monoclonal antibodies specific for the capsid protein of chikungunya virus suitable for multiple applications. J Gen Virol. 2015;96:507–12.

    Article  CAS  PubMed  Google Scholar 

  15. van den Hurk AF, Hall-Mendelin S, Pyke AT, Smith GA, Mackenzie JS. Vector competence of Australian mosquitoes for chikungunya virus. Vector Borne Zoonotic Dis. 2010;10:489–95.

    Article  PubMed  Google Scholar 

  16. Hunt GJ, McKinnon CN. Evaluation of membranes for feeding Culicoides variipennis (Diptera: Ceratopogonidae) with an improved artificial blood-feeding apparatus. J Med Entomol. 1990;27:934–7.

    Article  CAS  PubMed  Google Scholar 

  17. Balenghien T, Vazeille M, Grandadam M, Schaffner F, Zeller H, Reiter P, et al. Vector competence of some French Culex and Aedes mosquitoes for West Nile virus. Vector Borne Zoonotic Dis. 2008;8:589–95.

  18. Pesko K, Westbrook CJ, Mores CN, Lounibos LP, Reiskind MH. Effects of infectious virus dose and bloodmeal delivery method on susceptibility of Aedes aegypti and Aedes albopictus to chikungunya virus. J Med Entomol. 2009;46:395–9.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Smith DR, Carrara AS, Aguilar PV, Weaver SC. Evaluation of methods to assess transmission potential of Venezuelan equine encephalitis virus by mosquitoes and estimation of mosquito saliva titers. Am J Trop Med Hyg. 2005;73:33–9.

    PubMed  Google Scholar 

  20. Tan CH, Wong PS, Li MZ, Yang HT, Chong CS, Lee LK, et al. Membrane feeding of dengue patient’s blood as a substitute for direct skin feeding in studying Aedes-dengue virus interaction. Parasit Vectors. 2016;9:211.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Vazeille M, Moutailler S, Coudrier D, Rousseaux C, Khun H, Huerre M, et al. Two chikungunya isolates from the outbreak of La Reunion (Indian Ocean) exhibit different patterns of infection in the mosquito, Aedes albopictus. PLoS One. 2007;2:e1168.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Anderson SL, Richards SL, Tabachnick WJ, Smartt CT. Effects of West Nile virus dose and extrinsic incubation temperature on temporal progression of vector competence in Culex pipiens quinquefasciatus. J Am Mosq Control Assoc. 2010;26:103–7.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Weaver SC, Scherer WF, Cupp EW, Castello DA. Barriers to dissemination of Venezuelan encephalitis viruses in the Middle American enzootic vector mosquito, Culex (Melanoconion) taeniopus. Am J Trop Med Hyg. 1984;33:953–60.

    CAS  PubMed  Google Scholar 

  24. Kramer LD, Hardy JL, Presser SB, Houk EJ. Dissemination barriers for western equine encephalomyelitis virus in Culex tarsalis infected after ingestion of low viral doses. Am J Trop Med Hyg. 1981;30:190–7.

    CAS  PubMed  Google Scholar 

  25. Vega-Rua A, Schmitt C, Bonne I, Krijnse Locker J, Failloux AB. Chikungunya virus replication in salivary glands of the mosquito Aedes albopictus. Viruses. 2015;7:5902–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. van den Hurk AF, Smith CS, Field HE, Smith IL, Northill JA, Taylor CT, et al. Transmission of Japanese encephalitis virus from the black flying fox, Pteropus alecto, to Culex annulirostris mosquitoes, despite the absence of detectable viremia. Am J Trop Med Hyg. 2009;81:457–62.

    PubMed  Google Scholar 

  27. Mangiafico JA. Chikungunya virus infection and transmission in five species of mosquito. Am J Trop Med Hyg. 1971;20:642–5.

    CAS  PubMed  Google Scholar 

  28. Christofferson RC, McCracken MK, Johnson AM, Chisenhall DM, Mores CN. Development of a transmission model for dengue virus. Virol J. 2013;10:127.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Styer LM, Kent KA, Albright RG, Bennett CJ, Kramer LD, Bernard KA. Mosquitoes inoculate high doses of West Nile virus as they probe and feed on live hosts. PLoS Pathog. 2007;3:1262–70.

    Article  CAS  PubMed  Google Scholar 

  30. Franz AW, Kantor AM, Passarelli AL, Clem RJ. Tissue barriers to arbovirus infection in mosquitoes. Viruses. 2015;7:3741–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Laurent P, Le Roux K, Grivard P, Bertil G, Naze F, Picard M, et al. Development of a sensitive real-time reverse transcriptase PCR assay with an internal control to detect and quantify chikungunya virus. Clin Chem. 2007;53:1408–14.

    Article  CAS  PubMed  Google Scholar 

  32. Appassakij H, Khuntikij P, Kemapunmanus M, Wutthanarungsan R, Silpapojakul K. Viremic profiles in asymptomatic and symptomatic chikungunya fever: a blood transfusion threat? Transfusion. 2013;53:2567–74.

    Article  PubMed  Google Scholar 

  33. Schneider BS, Higgs S. The enhancement of arbovirus transmission and disease by mosquito saliva is associated with modulation of the host immune response. Trans R Soc Trop Med Hyg. 2008;102:400–8.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Styer LM, Lim PY, Louie KL, Albright RG, Kramer LD, Bernard KA. Mosquito saliva causes enhancement of West Nile virus infection in mice. J Virol. 2011;85:1517–27.

    Article  CAS  PubMed  Google Scholar 

  35. Thangamani S, Higgs S, Ziegler S, Vanlandingham D, Tesh R, Wikel S. Host immune response to mosquito-transmitted chikungunya virus differs from that elicited by needle inoculated virus. PLoS One. 2010;5:e12137.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Mores CN, Christofferson RC, Davidson SA. The role of the mosquito in a dengue human infection model. J Infect Dis. 2014;209 Suppl 2:S71–8.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Schmid MA, Glasner DR, Shah S, Michlmayr D, Kramer LD, Harris E. Mosquito saliva increases endothelial permeability in the skin, immune cell migration, and dengue pathogenesis during antibody-dependent enhancement. PLoS Pathog. 2016;12:e1005676.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Thakar J, Mohanty S, West AP, Joshi SR, Ueda I, Wilson J, et al. Aging-dependent alterations in gene expression and a mitochondrial signature of responsiveness to human influenza vaccination. Aging (Albany NY). 2015;7:38–52.

    Article  Google Scholar 

  39. Qian F, Wang X, Zhang L, Lin A, Zhao H, Fikrig E, et al. Impaired interferon signaling in dendritic cells from older donors infected in vitro with West Nile virus. J Infect Dis. 2011;203:1415–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Stout-Delgado HW, Yang X, Walker WE, Tesar BM, Goldstein DR. Aging impairs IFN regulatory factor 7 up-regulation in plasmacytoid dendritic cells during TLR9 activation. J Immunol. 2008;181:6747–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Danis B, George TC, Goriely S, Dutta B, Renneson J, Gatto L, et al. Interferon regulatory factor 7-mediated responses are defective in cord blood plasmacytoid dendritic cells. Eur J Immunol. 2008;38:507–17.

    Article  CAS  PubMed  Google Scholar 

  42. Levy O. Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat Rev Immunol. 2007;7:379–90.

    Article  CAS  PubMed  Google Scholar 

  43. Judith D, Mostowy S, Bourai M, Gangneux N, Lelek M, Lucas-Hourani M, et al. Species-specific impact of the autophagy machinery on chikungunya virus infection. EMBO Rep. 2013;14:534–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kuno G, Chang GJ. Biological transmission of arboviruses: reexamination of and new insights into components, mechanisms, and unique traits as well as their evolutionary trends. Clin Microbiol Rev. 2005;18:608V37.

    Article  Google Scholar 

Download references

Acknowledgements

We thank Clay Winterford and his staff (Histotechnology Facility QIMR B) and animal house staff at QIMR B for their excellent support, and to Elise Kho (Mosquito Control Laboratory QIMR B) for assistance with mosquito rearing. We would also like to thank Dr Roy Hall (University of Queensland) for supply of the 5.5G9 monoclonal antibody.

Funding

The work was funded by a project grant from the National Health and Medical Research Council, Australia (APP1078468), a Perpetual JT Wilson Fellowship (Leon Hugo), and donations from John and Elizabeth Hunter, and Ed Westaway, Royal Australian Air Force Association.

Availability of data and material

The data supporting the conclusions of this article are included within the article. Raw data and materials are available from the corresponding author upon request.

Authors’ contributions

LH, NP, GD, AS conceived and designed the study; LH, NP, BT undertook the experiments; LH, NP, AS analysed the data; AS wrote the paper. All authors read, reviewed and approved the final manuscript.

Authors’ information

LH and NAP should be considered joint first authors.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethics approval

All mouse work was conducted in accordance with the “Australian code for the care and use of animals for scientific purposes” as defined by the National Health and Medical Research Council of Australia. Mouse work was approved by the QIMR Berghofer Medical Research Institute animal ethics committee (approval number A0108-062 M) and was conducted in biosafety level 3 facility at the QIMR Berghofer. Mice were euthanized using carbon dioxide.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Andreas Suhrbier.

Additional file

Additional file 1: Figure S1.

The percentage of mosquitoes where some positive staining for the indicated organs/tissues was evident is shown; from the experiment described in Fig. 1c-g. Quantification of staining density for this experiment is shown in Fig. 1c. Figure S2. Image of foot swelling in IRF3/7-/- mice. Detailed methods: mosquito immunohistochemistry and quantification. (PDF 71 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hugo, L.E., Prow, N.A., Tang, B. et al. Chikungunya virus transmission between Aedes albopictus and laboratory mice. Parasites Vectors 9, 555 (2016). https://doi.org/10.1186/s13071-016-1838-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13071-016-1838-1

Keywords