Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2024 Dec 3;19(12):e0314516. doi: 10.1371/journal.pone.0314516

Evaluation of the immune status of dogs vaccinated against rabies by an enzyme-linked immunosorbent assay using crude preparations of insect cells infected with a recombinant baculovirus encoding the rabies virus glycoprotein gene

A K Santosh 1, Deepak Kumar 2, Charanpreet Kaur 3, Priya Gupta 3,4, Pagala Jasmeen 3,4, L Dilip 1, G Kavitha 1, Suresh Basagoudanavar 5, Madhusudan Hosamani 5, V Balamurugan 6, R Sharada 1, D Rathnamma 1, K M Sunil 1, Nagendra R Hegde 3,4,*, Shrikrishna Isloor 1,*
Editor: Jian Xu7
PMCID: PMC11614288  PMID: 39625902

Abstract

Evaluation of the effectiveness of vaccination of animals against rabies is not routinely implemented. In cases where it is carried out, the rapid fluorescent focus inhibition test (RFFIT) or the fluorescent antibody virus neutralization (FAVN) test are the recommended tests. However, both of these tests require handling of live rabies virus (RABV), and are cumbersome to perform. In view of this, the enzyme-linked immunosorbent assay (ELISA) has been proposed as a surrogate test; however, availability of appropriate antigen is a major impediment for the development of ELISAs to detect anti-rabies antibodies. The most widely used antigen is the RABV glycoprotein (G) purified from cell culture-propagated virus, which requires a biosafety level 3 containment. The alternative is to use recombinantly expressed G, which needs to be to be properly glycosylated and folded to serve as the best antigen. The most suitable system for its production is the baculovirus expression system (BVES). However, purification of RABV G is challenging. We therefore tested partially purified preparations in the form of extracts of insect cells infected with baculovirus expressing RABV G, against sera from vaccinated dogs in an indirect ELISA. The results showed good concordance against RFFIT, with sensitivity and specificity of 90.48% and 80.00%, respectively. The system may be used for quick screening to determine the presence and an approximate level of antibodies, and can be modified to enable monitoring of mass dog vaccination programs, as well as to facilitate certification of dogs intended for international travel and transportation.

Introduction

Rabies has existed for at least 4000 years, and continues to pose a serious public health threat, particularly for children aged <15 years, especially in Asia and Africa [1, 2]. This nearly 100% fatal disease, caused by Lyssavirus rabies (RABV), is completely preventable through vaccination, even after exposure [35]. Dogs account for 54%, terrestrial wildlife (skunks, raccoons, foxes, cats, etc.) for 42%, and bats 4% of the human rabies cases worldwide [6, 7]. However, in many low- and middle-income countries (LMIC) where canine rabies has not been adequately controlled, dogs account for at least 90% of the human rabies cases [1, 8]. Consequently, vaccinating domestic dogs is the most economical and impactful intervention in preventing rabies in humans [5, 7, 9]. It has been estimated that vaccinating >70% of the dogs would be sufficient to create a barrier which would block transmission of RABV to humans [10, 11]. However, whilst recording vaccination is possible for pets, the same is difficult to impossible for free ranging dogs, especially in LMIC, and hence there is no means to verify the success of the vaccination programmes other than keeping a record of the vaccination. Therefore, knowing the immune status by estimating the level of antibodies in vaccinated dogs would be vital, both in the case of vaccination drives, particularly in urban areas, as well as for certification of dogs for international transportation. In addition, monitoring the level of antibodies would be required when clinical trials are conducted for new vaccines or when new vaccination regimen are being standardized.

Presence of neutralising antibodies (NAbs) is the most commonly studied correlate of protection, and hence the most reliable indicator of the success of vaccination against several virus diseases. Detection and quantification of NAbs is typically achieved by employing the virus neutralization (VN) test. For a number of years, the mouse neutralization test (MNT) was the only test used for assessing the presence as well as quantification of NAbs against RABV [12, 13]. Owing to the ethical issues of using mice, the associated expenditure and the time involved, cell culture techniques were later developed. The most widely used techniques are those based on the reduction in the proportion of RABV-infected cells by fluorescence methods, namely, the rapid fluorescent focus inhibition test (RFFIT) and the fluorescent antibody virus neutralization (FAVN) test [14, 15]. These tests are typically used for surveillance in animals (dog, fox) either before or after vaccination. An antibody titre of at least 0.5 international units per mL (IU/mL) of serum is considered to be adequate to confer protection to dogs and other carnivores [4, 1619], although some dogs show protection without NAbs [20].

In spite of their immense utility, both RFFIT and FAVN are time-consuming, expensive, not amenable to high-throughput testing, and require a fluorescent microscope, trained personnel as well as high-level bio-containment (to handle live RABV), not to mention that they are difficult to standardize across laboratories. The read-out also involves observation of at least 20 fields or calculating the proportion of infected cells for each dilution of the antibody, both of which may result in objectivity and varying reproducibility. In addition, poor quality sera (e.g., haemolysed) can cause toxicity to cells used in the assay, making the interpretation of the results difficult [21, 22]. For these reasons, various other techniques have been explored as alternatives. Neutralization of pseudotyped lentiviruses has been shown to be as good as RFFIT [23]. However, the limitations of RFFIT and FAVN, except for the requirement for higher level biocontainment, apply to this test as well. A simpler, user-friendly test is the enzyme immunoassay, a variety of which have been described to quantify the level of antibodies in humans, domestic animals and wildlife species [22, 2448]. Among these, the enzyme-linked immunosorbent assay (ELISA) is easy and rapid (~4 h) to perform, and does not require highly trained workers, nor handling of live virus, and can be performed even with poor quality sera [26, 29, 30, 49]. ELISA has now been accepted for use in the verification of the immune status of dogs and cats as a pre-requisite for international movement, provided that the test is validated as fit for such purposes.

Commercial ELISA kits have been developed for detecting antibodies in dogs and other animal species. However, such kits are cost prohibitive, limiting their routine usage, especially in resource-limited settings. Moreover, most such kits use the RABV glycoprotein (G) purified from bulk propagated infectious virus, a process which is time-consuming and requires biosafety level 3 facilities. These limitations could be overcome by using recombinant G as the antigen. The RABV G has been expressed using various systems, both for use as a vaccine candidate as well as for use as an antigen in diagnostic assays. Full-length and truncated G have been expressed in bacteria and yeast. However, bacterially expressed G is often misfolded, toxic to bacteria, unglycosylated, and poorly immunogenic, and that expressed in yeast appears to undergo improper glycosylation as well as abnormal folding and oligomerization. On the other hand, RABV G expressed in eukaryotic cells of plant or animal origin has been reported to be properly folded and glycosylated (reviewed in [50]).

The best substrates for producing recombinant G are mammalian cells, but they require expensive medium and supplements for their propagation, are mostly anchorage-dependent, do not attain a high density, and are sensitive to hydrodynamic stress [51], making large-scale antigen production through the use of mammalian cells cumbersome and expensive. On the other hand, production of antigen in insect cells through baculoviruses is more economical. Insect cells require relatively milder culture conditions, such as propagation at lower temperatures and no requirement for CO2. In addition, the baculoviruses are safe for handling since their tropism is restricted to insects. The cells and viruses can also be propagated in serum-free media and can easily be scaled up. When mammalian or viral proteins are expressed in insect cells, protein folding and processing are more native than when expressed through prokaryotic systems [52, 53]. The RABV G expressed using baculovirus expression system (BVES) has been shown to be similar structurally and biologically to the native protein and has been used as an immunogen [5457].

Even when a recombinant protein is expressed to reasonable levels, whether in mammalian cells or through BVES, it is often difficult to recover and purify membrane-anchored, glycosylated proteins. Purification of native RABV G from virus propagated in mammalian cells is well-established [58] but requires virus culture. On the other hand, purification of G with the retention of its trimeric, fully glycosylated, and immunogenic structure when expressed in eukaryotic cells is not a trivial task (reviewed in [50]). With the intent to avoid purification processes which are either complicated or result in poor recovery, we used crude preparations of baculovirus-infected insect cells for the development of an indirect ELISA (iELISA) for the detection and semi-quantitative estimation of anti-RABV antibodies in dogs vaccinated against rabies.

Material and methods

Ethics statement

As per the rules and guidelines of the Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA; recently renamed CCSEA), Department of Animal Husbandry and Dairying, Ministry of Fisheries, Animal Husbandry and Dairying, Government of India, no ethical clearance is required for the collection of blood samples of volumes up to 10 mL from animals for academic research purposes in India.

Serum samples

Serum samples (n = 245) of vaccinated pet dogs submitted to the Rabies Diagnostic Laboratory, Veterinary College, Karnataka Veterinary & Animal Fisheries Sciences University, Bengaluru, India, were used in the current study. The sera had been collected from ten different states of India, namely, Maharashtra (n = 107), Karnataka (n = 87), Delhi (n = 13), Punjab (n = 10), Haryana (n = 9), Andhra Pradesh, Kerala (n = 6 each), Gujarat, Tamil Nadu, and West Bengal (n = 3 each). The serum samples were stored at -20°C until used for testing.

Cell and virus culture

Baby hamster kidney– 21 (BHK-21; American Type Culture Collection, USA) cells were propagated at 37°C and 5% CO2 in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 5% fetal bovine serum (FBS; Gibco, USA), 100 μg/ml of penicillin and 100 U/ml of streptomycin (Invitrogen, USA). The Spodoptera frugiperda insect-derived Sf21 cells were propagated at 25°C without CO2 in Sf900 II medium (Thermo Fisher Scientific, USA) supplemented with 10% FBS, 100 ug/ml of penicillin and 100 U/ml of streptomycin.

The Larghi strain of RABV was propagated by infecting 75–80% confluent BHK-21cells with a multiplicity of infection (MOI) of 0.1. After 48 hrs, the infected cell supernatant was harvested. The recombinant baculoviruses (generated as outlined below) were propagated by infecting 75–80% confluent Sf21 cells at an MOI of 0.1, and harvesting the cells after 48 hrs. With both the viruses, titres were determined with the relevant cells cultured in 96-well tissue-culture plates, and calculating 50% tissue-culture infective dose (TCID50) by applying the Reed and Muench method [59].

Rapid fluorescent focus inhibition test (RFFIT)

The RFFIT was performed as per the protocol described previously [60]. Briefly, test sera were heat inactivated (56°C, 30 min) and diluted serially two-fold in a 96-well tissue culture plate, and mixed with equal volume (100 μL) of RABV diluted to 100 TCID50. The WHO reference serum was included in each test. The plate was incubated at 37°C for 90 min in a 5% CO2 incubator. Then, 2.5–3.0 x 104 BHK-21 cells were added to each well and incubated further at 37°C under 5% CO2 for 48 hrs. The medium was decanted from the plate and the cells were fixed in 75% ice-cold acetone for 30 min at -20°C. After decanting the acetone and air drying, the fixed monolayer was reacted with fluorescein isothiocyanate (FITC)-conjugated anti-RABV-nucleocapsid protein antibody, and the plate was incubated at 37°C under 5% CO2 for 60 min. The conjugate was decanted, and the plate was washed two times with phosphate buffered saline (PBS, pH 7.0). The plate was observed under a fluorescent microscope. For each serum sample, the highest dilution that neutralized the virus was recorded. The results were compared to the WHO reference serum (2 IU/mL) which at 1:8 dilution completely neutralized RABV. The titre of anti-RABV antibodies in sera was calculated by using the formula:

Neutralizingreciprocaloftestserumshowingcompleteantibodytiterinserum=NeutralizationofvirusinfectivityxUnitageofreferenceserumReciprocalofhighestdilutionofreferenceserumshowingcompleteneutralizationofvirusinfectivity

Generation of recombinant baculovirus encoding RABV G

The RABV G gene (Larghi’s strain), which was available in pET-32 vector, was re-cloned by including a hexa-histidine tag at the C-terminus, using appropriate primers; additional primers were used to confirm the presence of the gene (Table 1). The G gene was inserted into the baculovirus shuttle plasmid pFastbacI using EcoRI and SalI enzymes and T4 DNA ligase. The ligated product was transformed into E. coli DH5α cells and plated on Luria Bertini (LB) agar with 50 μg/mL ampicillin, and colonies were confirmed by PCR with pFastBacI forward and reverse primers (listed in Table 1). The pFastBacI-RABV-G was transformed into E. coli DH10Bac cells and plated on LB agar containing 50 μg/mL of kanamycin, 7 μg/mL of gentamicin, 10 μg/mL of tetracycline, 100 μg/mL of Blue-gal and 40 μg/mL isopropyl-β-thiogalactoside (IPTG). Positive (white) colonies were selected, and the bacmid was amplified and analysed by PCR using M13 forward and reverse primers (listed in Table 1).

Table 1. Primers used in the present study.

Primer Sequences (5’-3’) Amplicon size (bp) Reference
VF (EcoRI) GCG GAA TTC ATG GTT CCT CAG GTT CTT TTG T 1611 bp [61]
VR (NotI) CCCGG GCG GCC GC TCA CAG TCT GGT CTC ACC CCC
VR (SalI) RAB His RER TATTGAGTCGACTTATCAGTGATGGTGATGGTGATGACTGCC 1634 This report
pFastBacTM For GGATTATTCATACCGTCCCA 1784 [62]
pFastBacTM Rev CAAATGTGGTATGGCTGATT 1784 [62]
pUC/M13 Forward CCCAGTCACGACGTTGTAAAACG 3914 [62]
pUC/M13 Reverse AGCGGATAACAATTTCACACAGG 3914 [62]

Note: The underlined sequences represent restriction enzyme recognition sites.

The recombinant bacmid was transfected into Sf21 cells using the CellfectinTM reagent as per the manufacturer’s instructions. The resultant baculovirus (Passage 1 or P1) was stored at 4°C, and TCID50 was determined as described above. Further propagation to generate P2 and P3 was carried out by infecting Sf21 cells at an MOI of 0.1, and the preparations were titrated and stored at -80°C for further use.

Reverse transcription polymerase chain reaction (RT-PCR)

Approximately 0.8 × 106 Sf21 cells were seeded into each well of a 6-well plate and incubated at 28°C. The following day, the cells were infected with the recombinant baculovirus or a control baculovirus at an MOI of 0.1 and further incubated at 28°C for 48 hrs. The cells were harvested, and the supernatant and the pellet were processed separately using Trizol (Invitrogen, USA) to extract RNA. The RNA (1 μg) was utilized as a template for cDNA synthesis using the PrimeScript First Strand cDNA synthesis kit (Takara, Japan). The PCR was performed using gene-specific primers, involving denaturation at 98°C for 30 sec, annealing at 58°C for 45 sec, and extension at 72°C for 90 sec, followed by a final extension at 72°C for 10 min. The amplified products were analyzed by agarose gel electrophoresis, stained with ethidium bromide, and visualized under UV transillumination in a gel documentation system.

Western blotting

For confirming the expression of the G protein, supernatants and extracts from baculovirus-infected Sf21 cells were subjected to sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting. For this, 2X106 Sf21 cells were seeded in a 60-mm cell culture dish, incubated at 25°C without CO2. After an overnight culturing, the cells were infected at an MOI of 10 with the baculovirus encoding RABV G. After 48 hrs, the cells were harvested and centrifuged at 1000 X g. The cell pellet was solubilised with lysis buffer [25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM ethylene diamine tetraacetic acid (EDTA), 2.0% Triton X-100, 5% glycerol, 1 mM phenyl methyl sulfonyl fluoride (PMSF) and 8 M urea]. Protein was estimated in various samples by bicinchoninic acid (BCA) assay (Pierce, USA), and equal amounts of protein were electrophoresed on a 12% SDS-PAG, followed by transfer to a polyvinylidene difluoride (PVDF) membrane. The membranes were blocked with 5% bovine serum albumin in PBS, and immunoblotted using a 1:10,000 dilution of anti-His antibody (Sigma Aldrich, USA) or a 1:300 dilution of a dog serum with an estimated titre of 16 IU of anti-RABV antibodies. Then, the blot was incubated with a 1:5000 dilution of horse radish peroxidase (HRP) conjugated anti-mouse or anti-canine IgG (both from Sigma Aldrich, USA). Between each step, the membranes were washed thrice with PBS containing 0.05% Tween-20. Finally, bands were visualized by using the enhanced chemiluminescence reagent (Pierce, USA) and exposure to an X-ray film or capture through a gel documentation system.

Enzyme-linked immunosorbent assay

A crude preparation of RABV G was prepared by infecting Sf21 cells with an MOI of 10 of the recombinant baculovirus and harvesting at 96 hrs post-infection. The infected cells were pelleted, and extracts were prepared by one cycle of freeze-thaw. The total protein was estimated by Bradford assay (Bio-Rad, USA), before using it for the iELISA. A checker-board titration was performed and an antigen concentration of 500 ng/well, a serum dilution of 1:100 and a dilution of 1:15,000 for anti-dog IgG-HRP conjugate were determined to be optimum for use in the iELISA.

For testing the sera by iELISA, antigen (in the form of crude extract) was coated in carbonate-bicarbonate buffer (pH 9.6) at 4°C overnight. The content of the wells was discarded, and the plates were washed two times with PBS. The test sera were diluted 1:100 and added in duplicate wells. The appropriate controls viz., positive control serum (RFFIT titer of 8 IU/mL when undiluted and diluted 1:2 to 1:64 to achieve 4.0 to 0.125 IU/mL, respectively) negative control serum (from an unvaccinated healthy dog), conjugate controls (no sera added) and blank wells were maintained in each plate. The plate was then incubated at 37°C for 2 hrs. The plate was washed with PBS containing 0.05% Tween-20 (PBST) and horse radish peroxidase (HRP)-conjugated anti-dog IgG (Sigma Aldrich, USA) diluted 1:15,000 in PBST was added to each well and incubated at 37°C for 60 min. The content of the wells was discarded, and the plates were washed two times with PBST. Then, freshly prepared solution containing o-phenylene diamine (OPD) and 3% H2O2 (4 μL/mL of OPD) was added. The plate was incubated at room temperature for 15 min followed by addition of 2.5 N HCl to stop the reaction. Absorbance values were recorded at 492 nm wavelength using an ELISA reader. Percentage Positivity (PP) was calculated by the formula [61].

PP=ODofthetestsampleODofpositivecontrolX100

The positive cut-off PP value was determined by comparing test sera against different dilutions of pooled known vaccinated dog serum (control) having a titer of 8.0 IU/mL in each ELISA plate. The process was repeated 13 times on different days. The cut-off PP value was then derived by analyzing results of iELISA against RFFIT using Michaelis-Menton (2 parms) regression model which uses the general equation. Statistical analysis was performed by using R Studio 2022.12.0 Build 353 with R base version 4.2.2 using packages dplyr 1.0.10, ggplot2 3.4.0 and drc 3.0–1

y=axb+x

Statistical analyses

The Graphpad prism software version 5.0 was used to calculate the mean, standard deviation, and standard error, as well as for curve fitting using non-linear polynomial quadratic equation. Spearman rank correlation was employed to compare the RFFIT and the ELISA. One-way ANOVA was employed to discern the effect of breed, gender, vaccine brand and association between time intervals of blood sampling and antibody titer, using GraphPad Prism V5 software. A p value of < 0.05 was considered statistically significant. Sensitivity and specificity of the ELISA was calculated as described previously [63], and by using the formulae as follows:

Sensitivity=aa+cX100
Specificity=db+dX100

where

a = Number of samples positive by both standard test and test to be compared

b = Number of samples positive by test to be compared but negative by the standard test

c = Number of samples negative by test to be compared but positive by the standard test

d = Number of samples negative by both the tests.

Kappa value was also calculated as described previously [64].

Results

The RABV G gene was amplified by PCR, and the resultant product was used in a second PCR to include an in-frame hexa-histidine (His) tag at the C terminus of the G protein. The product was inserted into pFastBac, and the cloning was confirmed by PCR. The construct pFastBac-RABV-G was used to transform DH10Bac cells, and the transposition of the G gene into the baculovirus genome was confirmed by blue-white colony screening, followed by PCR, and nucleotide sequencing.

Three different clones of pFastBac-RABV-G produced baculoviruses upon transfection of the Sf21 cells. When extracts and culture supernatants from uninfected cells and cells infected with one of the clones were subjected to PCR, both the pellet and the supernatant fractions from infected cells produced a band corresponding to the size (~ 1.6 kbp) of RABV G gene. As expected, the band intensity was higher with the infected cell pellet than the supernatant (Fig 1A; compare third and fourth lanes), reflecting the fact that much more DNA was present during virus replication in cells than that contained in the encapsidated virus released into the medium. On the other hand, only the pellet fraction of infected cells produced a band (Fig 1A, last two lanes), confirming that the mRNA would be present only in the fraction from cells where the virus would be expressing its genes, and not in the supernatant which is expected to contain mostly just the baculovirus. All the three different clones were then used to infect Sf21 cells, and the cell extracts were subjected to SDS-PAGE. A band of ~60 kDa was observed with all the three clones (Fig 1B, second to fourth lanes). When one of the clones was subjected to Western blotting with anti-His antibodies, a single band of ~60 kDa was observed with cells infected with the baculovirus encoding the G gene, and not with a control baculovirus which did not carry any insert (Fig 1C, compare the second lane against the first lane). These data together confirm the generation of the recombinant baculovirus and the expression of RABV G in Sf21 insect cells.

Fig 1. Expression of G by recombinant baculovirus.

Fig 1

A. Uninfected (Uninf.) or infected (Inf.) Sf-21 cell cultures were separated into cells and supernatants and subjected to PCR or RT-PCR using primers for full-length G gene. B. Cell extracts of uninfected (Mock) and infected (three different clones designated 1.4, 4.4 and 5.17) were subjected to SDS-PAGE and Coomassie blue staining. The position of protein markers is shown on the right. C. Extracts from control (untransfected) and transfection control (Cellfectin only) as well as cells infected with a control baculovirus (Bac-Null) or a baculovirus encoding RABV G (Bac-RABV-G) were subjected to immunoblotting using anti-His antibodies. The position of protein markers is shown on the right.

Efforts were then made to purify G. Various lysis buffers and other conditions were initially attempted, and the procedure described in the Materials and Methods was finalized. Cell extracts were subjected to Ni-NTA chromatography, and a single band of expected size was observed in the elution fractions by SDS-PAGE followed by Coomassie blue staining (Fig 2A), as well as in pooled fractions by Western blotting using anti-His antibodies (Fig 2B). Furthermore, when pooled fractions were tested against sera from vaccinated and unvaccinated dogs, faint bands of expected size were observed only with the immune sera, and not with the non-immune sera (Fig 2C).

Fig 2. Purification of G expressed through baculovirus.

Fig 2

A. Extracts of Sf21 cells infected with baculovirus encoding the RABV G gene were passed over a Ni-NTA column, and fractions (numbered 1 through 8) were subjected to SDS-PAGE and Coomassie blue staining. The position of protein markers is shown on the right. B. Pooled fractions (2–4 and 5–8) were subjected to immunoblotting using anti-His antibodies. C. The pooled fraction 5–8 was subjected to immunoblotting using sera from dogs which were either naïve (non-immune) or vaccinated (immune) against rabies. The expected position of the G protein is shown on the left.

However, whilst G was consistently being expressed through the BVES, as evidenced through Western blotting of cell extracts using anti-His antibodies, the success rate of obtaining purified protein was poor. We frequently failed to observe any binding of the His-tagged protein to the Ni-NTA resin. Even when bound, often the yield of the purified protein was low, and insufficient to develop an ELISA. This was observed at two of the collaborating laboratories. Considering this, infected cell extracts were directly tested against some canine sera in an ELISA using anti-His antibodies. Following encouraging results, such extracts were used to develop the in-house iELISA. Total protein in the infected cell extract was estimated and following checker-board analysis, a concentration of 500 ng per well, test serum dilution of 1:100, and anti-dog-HRP conjugate of 1:15,000 were optimized. All the test serum samples were then subjected to iELISA as per the optimized protocol.

As evaluated against the reference standard serum by RFFIT, 210 out of the 245 serum samples (85.71%) showed an antibody titre of ≥ 0.5 IU/mL whereas the rest (n = 35; 14.29%) had a titre of < 0.5 IU/mL (S1 Table). When all the samples were tested in duplicate by iELISA, the PP values showed a certain range for each of the group of samples with a certain unitage. The number of samples falling under each of the titres by RFFIT as well as the corresponding range of PP values by iELISA are shown in Table 2.

Table 2. Distribution of samples and their respective PP values under each of the titres.

Unitage (IU/mL) No. of samples by RFFIT Range of PP values by ELISA
16.0 2 48.64–116.30
8.0 8 48.64–100.00
4.0 40 28.70–92.65
2.0 52 15.74–98.09
1.0 51 25.78–85.95
0.5 57 20.87–93.24
<0.5 35 11.06–60.76

The average PP values plotted against corresponding units of antibody titres generated a perfect hyperbola (Fig 3A), indicating concordance between the two tests. Spearman rank correlation analysis for all the 245 samples (Fig 3B) showed a coefficient (r) of 0.44 (P<0.0001) indicating a significant correlation between the two tests. The regression model fitted the results significantly with a = 114.572435 (Standard Error 1.910154), b = 1.255603 (Standard Error 0.062057) with very significant p value ≤ 2.2 x 10−16 (p < 0.0001). From the regression analysis, the positive cut-off value was found to be 32.64 (IU/mL) for the iELISA (equivalent to 0.5 IU /mL by RFFIT). The derived regression model was also used to convert the test results of iELISA values into predicted RFFIT values. The predicted and actual RFFIT values were plotted into Area Under the Curve (AUC) plot, which is shown in Fig 4.

Fig 3. Non-linear regression analysis of RFFIT vs iELISA.

Fig 3

A. Dog sera with an antibody titre of 8.0 IU/mL as estimated by RFFIT were pooled and serially diluted, and plotted against positive predictive values obtained by iELISA for the test samples. B. Values in IU/mL for the iELISA, derived based on the non-linear regression analysis from Fig 3, were plotted against values in IU/mL of RFFIT.

Fig 4. Area under the curve for RFFIT vs iELISA.

Fig 4

The distribution of the frequency of the number of samples at each of the dilutions is plotted for RFFIT and iELISA. The dashed vertical line represents cut-off between positive and negative results at 0.5 IU/mL for RFFIT.

When compared to RFFIT, the sensitivity and specificity of the ELISA were 90.48% and 80.00%, respectively (Table 3). Furthermore, the results of our in-house ELISA showed good agreement (κ value 0.647) with RFFIT (Table 3).

Table 3. Sensitivity and specificity of the in-house iELISA.

Test RFFIT Total Sensitivity (%) Specificity (%) Kappa value
Positive Negative
In-house ELISA Positive 190 7 197 90.48 80.00 0.647
Negative 20 28 48
Total 210 35 245

Note: Kappa (κ) value of 0.61–0.80 is considered as a substantial agreement between the two tests [29].

Discussion

Detection and quantitation of antibodies in the sera of vaccinated humans and animals is essential for the assessing the success of vaccination and in turn the control of rabies. Owing to several constraints in performing RFFIT and FAVN for this purpose, ELISAs have been promulgated as alternatives. However, the development and application of a universally acceptable ELISA has been difficult. One of the impediments is the requirement for an appropriate antigen. Besides biosafety concerns for its production when requiring purification from whole virus, the G antigen may produce false positive results due to the presence of other viral antigens which may cross-react with antibodies to closely related viruses (unpublished observations). Hence, production of recombinant G is a safe and better alternative. We used baculovirus-infected insect cell extracts as antigen in an iELISA. The results showed good correlation with RFFIT.

For any diagnostic test, one requires a continuous supply of sufficient quantities of an immunologically functional, native protein. However, the expression of viral glycoproteins is challenging due to their glycosylation and structural complexity. The RABV G is a type I membrane protein that assembles into trimers [65, 66]. Appropriate glycosylation and assembly of G is important for its proper expression, folding, oligomerization, and function [6772]. The membrane anchorage of G on the virus surface and its trimeric structure are critical for the induction of NAbs [7378]. The purification of native G in sufficient quantities depends on a number of variables such as the cell type, the extraction volume, the buffer used and its ionic strength, the concentration of salts, the detergent used and its concentration, the incubation temperatures and duration, and the propensity of the protein to refold [55, 73, 7982]. Affinity tags (e.g. 6X His) fused to G have been reported but have produced variable recovery [55, 78, 80, 81], and may require additional or alternative methods such as gel filtration, ion exchange or lectin chromatography [35, 79, 82]. Indeed, we found it problematic to consistently purify His-tagged G from baculovirus-infected cell extracts. Other than the often-observed variations in the expression levels as well as the suitability of a lysis buffer to extract the protein, the major difficulty was the frequent inability to capture the protein on a nickel column even when expressed well. These were the reasons for us to use baculovirus-infected Sf21 cell extracts as antigen in the iELISA reported here.

The ELISA is a simple, rapid, affordable, and automatable test. The non-requirement for live virus makes ELISA a complementary or practical alternative test to neutralization methods for rabies. Several studies have reported the development of ELISAs for detecting antibodies to RABV, in human and animal sera [15, 19, 26, 28, 3235, 37, 38, 40, 49, 8386]. Indirect ELISA (iELISA) is the easiest of the ELISA methods to perform and standardize. However, its concordance with neutralization assays for anti-RABV antibodies has ranged from poor to excellent [22, 24, 26, 2830, 34, 35, 40, 41, 45, 83, 8691]. The variable performance of ELISAs could be due to the type of the antigen or the conjugated antibodies used, the species from which the serum is tested, the gold standard (MNT, RFFIT, FVNA or other neutralization tests) used for comparison, as well as inter-laboratory variations. Inconsistent relationship between neutralization (RFFIT) and antigen-binding (iELISA) either in the degree of agreement or in the direction of response has been noted for individual subjects [18, 19, 92]. Despite the limitations, the iELISA is the most amenable assay for the detection and quantitation of antibodies against RABV. A commercial iELISA is available, and has been tested with human as well as animal sera [33, 34, 49, 86, 93], although some results which are discrepant on sensitivity, specificity, accuracy, and seroconversion threshold in dogs vaccinated in the field as well as incompatible with clinical utility index have been noted [34, 47, 49, 94].

Rabies has the highest case fatality rate of any infectious disease. Hence, any assay which determines a cut-off value as a correlate of protection needs to be robust, particularly in eliminating false positive results. A serum antibody titre cut-off of 0.5 IU/mL is recognized as proof of adequate response deemed to be protective against rabies in cats, dogs, and wild carnivores [4, 16, 17, 19, 95]. However, this was originally determined based on neutralization methods such as MNT and RFFIT, for testing sera collected around one month after vaccination. To extrapolate this value to other methods requires extensive investigation and standardization, while also keeping in mind that the methods measure different kinds and characteristics of antibodies [18, 19]. In addition, the relationship between the measurement of binding antibody and neutralizing function is expected to vary between individuals and between different tests, and the comparisons may not be equal or sometimes even interpretable [18, 92]. The interpretation of the result therefore depends on the method applied, what it measures, the cut-off value and equivalency determination for the purpose of testing, and various sample considerations including quality, time of collection and the presence of toxic substances [18, 92].

Finally, several factors influence the development of any robust assay. These include (a) the fit for purpose, required sensitivity, specificity, limit of detection, limit of quantitation in the linear range, (b) the design of the experiment, (c) the type and the purity of the antigen, (c) the type, source and quality of the test sample, (d) the type of capture and/or detection antibody (species, poly/monoclonal), (d) the conjugate and the detection system [94]. In terms of purity of the antigen used for coating, it has been observed that homogenous (purified) antigens are better for evaluating NAbs [96]. One of the major issues with ELISA is its reliability to faithfully discriminate sera with sufficient versus insufficient neutralization capacities. The fact that only a fraction of the anti-G MAbs can neutralize RABV suggests that conventional ELISAs may be inadequate to predict the quality or quantity of NAbs. Hence, even though several ELISAs have been developed, none of them have replaced the VNTs [97]. Therefore, ELISAs could be used as initial screening tests before applying the recommended tests for confirmation [98].

Our iELISA showed a sensitivity of 90.48% and a specificity of 80.00% against RFFIT, with a κ value of 0.647 with a sample size of 245 (dog sera). A few other studies have evaluated the application of different forms of ELISA in the context of monitoring antibodies in dogs. The earliest study tested an iEILSA using virus-derived G as the antigen against sera from wild and captive unvaccinated and vaccinated foxes [29, 30]. High correlation was observed, with r = 0.91 in one study and 0.88 in another, and agreement of 93.0% and 89.9%, respectively, against FAVN [29, 30]. Parallel testing at five different laboratories found that there was substantial to almost perfect agreement among the results between the tests as well as between ELISA and FAVN [29]. The test was later adapted with inactivated virus as antigen for testing 2360 sera from dogs and cats in three independent studies, where a specificity of 96.9% and sensitivity of 86.4% was reported [28]. A recently reported iELISA used detergent lysed virus from concentrated culture supernatant as antigen, and the test performed equally or better than two commercial iELISAs [34]. A competitive ELISA using virus-infected mammalian cell lysate as antigen, and 4350 vaccinated and unvaccinated dog sera, showed a correlation of 96.2% against FAVN [99]. A commercial blocking ELISA was tested in another study against 233 cat sera (46 unvaccinated, 187 vaccinated) and 765 dog sera (269 naïve and 496 vaccinated). The test showed high specificity (>97%) against a commercial iELISA, and 86.2% overall agreement with FAVN [22]. The sensitivity for various groups of sera, as categorized by different ranges of IU/mL, varied from 50% to 94.42%, and most of the discrepancy was with sera nearer to the cut-off value of 0.5 IU/mL [22]. The versatility of the blocking ELISA was demonstrated by applying a single test for sera from dogs, cats, cattle and humans, although the total number of samples (n = 46) was low; this test uses virus-like particles (VLP) obtained from mammalian cells [100]. Contrary to other studies where high specificity and lower sensitivity was observed, our results showed higher sensitivity and lower specificity. The reasons for this are unknown. It may be noted that all the above studies used different antigens (G purified from virus, inactivated virus, lysate of mammalian cells stably expressing G, lysate of infected mammalian cell culture supernatant, VLP), in different formats (iELISA, bELISA, cELISA) against samples from different locations from different species, whereas we used baculovirus-infected insect cell lysate as antigen against dog sera. In addition, the development stages vary from a proof-of-concept to interlaboratory validation. Hence, it is difficult to infer on the performance of our assay, which is still a proof-of-concept, in comparison to others.

Our results suggest that an iELISA using partially purified extracts of cells infected with baculovirus expressing RABV G could be of immense benefit to LMICs as it can be used in various laboratories, not only for the estimation of antibodies of pets intended for international travel but also for monitoring the success of mass immunization programmes. However, the requirement for infection of insect cells with baculovirus, the variable expression levels and consistent outcomes of purification remain as obstacles. Optimization of clones for consistent expression as well as alternative purification strategies may be useful in further improving of the assay.

Supporting information

S1 Table. Details of the samples with RFFIT and ELISA values.

(DOCX)

pone.0314516.s001.docx (68.4KB, docx)
S1 Raw images

(PPTX)

pone.0314516.s002.pptx (2.7MB, pptx)

Acknowledgments

PJ and PG acknowledge fellowships provided respectively by the Council of Scientific & Industrial Research (CSIR) and the Department of Biotechnology (DBT), both under the Ministry of Science and Technology, Government of India.

Data Availability

All relevant data are within the manuscript and its Supporting information files.

Funding Statement

This research was funded in part by the Department of Biotechnology, Ministry of Science and Technology, Government of India (BT/ADV/Canine Health/GADVASU/2017-18). The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  • 1.Tarantola A. Four Thousand Years of Concepts Relating to Rabies in Animals and Humans, Its Prevention and Its Cure. Trop Med Infect Dis. 2017;2(2). Epub 20170324. doi: 10.3390/tropicalmed2020005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Yousaf MZ, Qasim M, Zia S, Khan M, Ashfaq UA, Khan S. Rabies molecular virology, diagnosis, prevention and treatment. Virol J. 2012;9:50. Epub 20120221. doi: 10.1186/1743-422X-9-50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rupprecht CE, Briggs D, Brown CM, Franka R, Katz SL, Kerr HD, et al. Use of a reduced (4-dose) vaccine schedule for postexposure prophylaxis to prevent human rabies: recommendations of the advisory committee on immunization practices. MMWR Recomm Rep. 2010;59(RR-2):1–9. . [PubMed] [Google Scholar]
  • 4.WHO. WHO expert consultation on rabies, Third Report. Geneva, Switzerland: World Health Organization, 2018.
  • 5.Zhu S, Guo C. Rabies Control and Treatment: From Prophylaxis to Strategies with Curative Potential. Viruses. 2016;8(11). Epub 20161028. doi: 10.3390/v8110279 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Krebs JW, Mandel EJ, Swerdlow DL, Rupprecht CE. Rabies surveillance in the United States during 2004. J Am Vet Med Assoc. 2005;227(12):1912–25. doi: 10.2460/javma.2005.227.1912 . [DOI] [PubMed] [Google Scholar]
  • 7.Meslin FX, Fishbein DB, Matter HC. Rationale and prospects for rabies elimination in developing countries. Curr Top Microbiol Immunol. 1994;187:1–26. doi: 10.1007/978-3-642-78490-3_1 . [DOI] [PubMed] [Google Scholar]
  • 8.Fishbein DB, Robinson LE. Rabies. N Engl J Med. 1993;329(22):1632–8. doi: 10.1056/NEJM199311253292208 . [DOI] [PubMed] [Google Scholar]
  • 9.Ives A, Dieuzy-Labaye I, Abela-Ridder B. Global characteristics of the rabies biologics market in 2017. Vaccine. 2019;37 Suppl 1(Suppl 1):A73–A6. Epub 20181109. doi: 10.1016/j.vaccine.2018.10.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Coleman PG, Dye C. Immunization coverage required to prevent outbreaks of dog rabies. Vaccine. 1996;14(3):185–6. doi: 10.1016/0264-410x(95)00197-9 . [DOI] [PubMed] [Google Scholar]
  • 11.Mindekem R, Lechenne MS, Naissengar KS, Oussiguere A, Kebkiba B, Moto DD, et al. Cost Description and Comparative Cost Efficiency of Post-Exposure Prophylaxis and Canine Mass Vaccination against Rabies in N’Djamena, Chad. Front Vet Sci. 2017;4:38. Epub 20170403. doi: 10.3389/fvets.2017.00038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Fitzgerald EA. Potency test of antirabies serum and immunoglobulins. In: Meslin FX, Kaplan M.M., Koprowsky H., editor. Laboratory techniques in rabies. 4th ed. Geneva: World Health Organization; 1996. p. 417–22. [Google Scholar]
  • 13.Webster LT, Dawson J.R. Early diagnosis of rabies by mouse inoculation. Measurement of humoral immunity to rabies by mouse protection test. Proc Sco Exp Biol Med. 1935;32:570–3. [Google Scholar]
  • 14.Cliquet F, Aubert M, Sagne L. Development of a fluorescent antibody virus neutralisation test (FAVN test) for the quantitation of rabies-neutralising antibody. J Immunol Methods. 1998;212(1):79–87. doi: 10.1016/s0022-1759(97)00212-3 . [DOI] [PubMed] [Google Scholar]
  • 15.Smith JS, Yager P.A., Baer G.M. A rapid fuorescent focus inhibition test (RFFIT) for determining rabies neutralizing antibody. In: Meslin XF, Kaplan M.M., Koprowski H., editor. Laboratory techniques in rabies. 4th ed. Geneva: World Health Organization; 1996. p. 200–8. [Google Scholar]
  • 16.Aubert MF. Practical significance of rabies antibodies in cats and dogs. Rev Sci Tech. 1992;11(3):735–60. doi: 10.20506/rst.11.3.622 . [DOI] [PubMed] [Google Scholar]
  • 17.Briggs DJ, Schweitzer K. Importation of dogs and cats to rabies-free areas of the world. Vet Clin North Am Small Anim Pract. 2001;31(3):573–83, viii. doi: 10.1016/s0195-5616(01)50610-5 . [DOI] [PubMed] [Google Scholar]
  • 18.Moore SM. Challenges of Rabies Serology: Defining Context of Interpretation. Viruses. 2021;13(8). Epub 20210731. doi: 10.3390/v13081516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Moore SM, Hanlon CA. Rabies-specific antibodies: measuring surrogates of protection against a fatal disease. PLoS Negl Trop Dis. 2010;4(3):e595. Epub 20100309. doi: 10.1371/journal.pntd.0000595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jakel V, Konig M, Cussler K, Hanschmann K, Thiel HJ. Factors influencing the antibody response to vaccination against rabies. Dev Biol (Basel). 2008;131:431–7. . [PubMed] [Google Scholar]
  • 21.Bedekovic T, Simic I, Kresic N, Lojkic I, Mihaljevic Z, Sucec I, et al. Evaluation of ELISA for the detection of rabies virus antibodies from the thoracic liquid and muscle extract samples in the monitoring of fox oral vaccination campaigns. BMC Vet Res. 2016;12:76. Epub 20160510. doi: 10.1186/s12917-016-0701-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wasniewski M, Cliquet F. Evaluation of ELISA for detection of rabies antibodies in domestic carnivores. J Virol Methods. 2012;179(1):166–75. Epub 20111103. doi: 10.1016/j.jviromet.2011.10.019 . [DOI] [PubMed] [Google Scholar]
  • 23.Nie J, Wu X, Ma J, Cao S, Huang W, Liu Q, et al. Development of in vitro and in vivo rabies virus neutralization assays based on a high-titer pseudovirus system. Sci Rep. 2017;7:42769. Epub 20170220. doi: 10.1038/srep42769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Arai YT, Kimura M, Sakaue Y, Hamada A, Yamada K, Nakayama M, et al. Antibody responses induced by immunization with a Japanese rabies vaccine determined by neutralization test and enzyme-linked immunosorbent assay. Vaccine. 2002;20(19–20):2448–53. doi: 10.1016/s0264-410x(02)00188-3 . [DOI] [PubMed] [Google Scholar]
  • 25.Atanasiu P, Perrin P, Delagneau JF. Use of an enzyme immunoassay with protein A for rabies antigen and antibody determination. Dev Biol Stand. 1980;46:207–15. . [PubMed] [Google Scholar]
  • 26.Barton LD, Campbell JB. Measurement of rabies-specific antibodies in carnivores by an enzyme-linked immunosorbent assay. J Wildl Dis. 1988;24(2):246–58. doi: 10.7589/0090-3558-24.2.246 . [DOI] [PubMed] [Google Scholar]
  • 27.Cardoso TC, Silva LH, Albas A, Ferreira HL, Perri SH. Rabies neutralizing antibody detection by indirect immunperoxidase serum neutralization assay performed on chicken embryo related cell line. Mem Inst Oswaldo Cruz. 2004;99(5):531–4. Epub 20041103. doi: 10.1590/s0074-02762004000500013 . [DOI] [PubMed] [Google Scholar]
  • 28.Cliquet F, McElhinney LM, Servat A, Boucher JM, Lowings JP, Goddard T, et al. Development of a qualitative indirect ELISA for the measurement of rabies virus-specific antibodies from vaccinated dogs and cats. J Virol Methods. 2004;117(1):1–8. doi: 10.1016/j.jviromet.2003.12.001 . [DOI] [PubMed] [Google Scholar]
  • 29.Cliquet F, Muller T, Mutinelli F, Geronutti S, Brochier B, Selhorst T, et al. Standardisation and establishment of a rabies ELISA test in European laboratories for assessing the efficacy of oral fox vaccination campaigns. Vaccine. 2003;21(21–22):2986–93. doi: 10.1016/s0264-410x(03)00102-6 . [DOI] [PubMed] [Google Scholar]
  • 30.Cliquet F, Sagne L, Schereffer JL, Aubert MF. ELISA test for rabies antibody titration in orally vaccinated foxes sampled in the fields. Vaccine. 2000;18(28):3272–9. doi: 10.1016/s0264-410x(00)00127-4 . [DOI] [PubMed] [Google Scholar]
  • 31.Cliquet F, Verdier Y, Sagne L, Aubert M, Schereffer JL, Selve M, et al. Neutralising antibody titration in 25,000 sera of dogs and cats vaccinated against rabies in France, in the framework of the new regulations that offer an alternative to quarantine. Rev Sci Tech. 2003;22(3):857–66. doi: 10.20506/rst.22.3.1437 . [DOI] [PubMed] [Google Scholar]
  • 32.Esterhuysen JJ, Prehaud C, Thomson GR. A liquid-phase blocking ELISA for the detection of antibodies to rabies virus. J Virol Methods. 1995;51(1):31–42. doi: 10.1016/0166-0934(94)00098-2 . [DOI] [PubMed] [Google Scholar]
  • 33.Feyssaguet M, Dacheux L, Audry L, Compoint A, Morize JL, Blanchard I, et al. Multicenter comparative study of a new ELISA, PLATELIA RABIES II, for the detection and titration of anti-rabies glycoprotein antibodies and comparison with the rapid fluorescent focus inhibition test (RFFIT) on human samples from vaccinated and non-vaccinated people. Vaccine. 2007;25(12):2244–51. Epub 20061227. doi: 10.1016/j.vaccine.2006.12.012 . [DOI] [PubMed] [Google Scholar]
  • 34.Fitria Y, Febrianto N, Putri RE, Rahmadani I, Subekti DT. Evaluation of In-House ELISA for Antirabies Antibodies Detection in Domestic Canine. Vet Med Int. 2023;2023:4096258. Epub 20230125. doi: 10.1155/2023/4096258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Grassi M, Wandeler AI, Peterhans E. Enzyme-linked immunosorbent assay for determination of antibodies to the envelope glycoprotein of rabies virus. J Clin Microbiol. 1989;27(5):899–902. doi: 10.1128/jcm.27.5.899-902.1989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Katayama S, Yamanaka M, Ota S, Shimizu Y. A new quantitative method for rabies virus by detection of nucleoprotein in virion using ELISA. J Vet Med Sci. 1999;61(4):411–6. doi: 10.1292/jvms.61.411 . [DOI] [PubMed] [Google Scholar]
  • 37.Kavaklova L, Eskenazy M, Gancheva T, Vacheva V. Enzyme-linked immunosorbent assay for quantification of rabies antibodies in human sera. Acta Virol. 1984;28(5):422–7. . [PubMed] [Google Scholar]
  • 38.Korimbocus J, Dehay N, Tordo N, Cano F, Morgeaux S. Development and validation of a quantitative competitive ELISA for potency testing of equine anti rabies sera with other potential use. Vaccine. 2016;34(28):3310–6. Epub 20160503. doi: 10.1016/j.vaccine.2016.04.086 . [DOI] [PubMed] [Google Scholar]
  • 39.Ma X, Niezgoda M, Blanton JD, Recuenco S, Rupprecht CE. Evaluation of a new serological technique for detecting rabies virus antibodies following vaccination. Vaccine. 2012;30(36):5358–62. Epub 20120627. doi: 10.1016/j.vaccine.2012.06.037 . [DOI] [PubMed] [Google Scholar]
  • 40.Nicholson KG, Prestage H. Enzyme-linked immunosorbent assay: a rapid reproducible test for the measurement of rabies antibody. J Med Virol. 1982;9(1):43–9. doi: 10.1002/jmv.1890090107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Piza AS, Santos JL, Chaves LB, Zanetti CR. An ELISA suitable for the detection of rabies virus antibodies in serum samples from human vaccinated with either cell-culture vaccine or suckling-mouse-brain vaccine. Rev Inst Med Trop Sao Paulo. 1999;41(1):39–43. doi: 10.1590/s0036-46651999000100008 . [DOI] [PubMed] [Google Scholar]
  • 42.Rathnadiwakara H, Gunatilake M, Cliquet F, Wasniewski M, Thammitiyagodage M, Karunakaran R, et al. Detection of immunity in sheep following anti-rabies vaccination. Clin Exp Vaccine Res. 2023;12(2):97–106. Epub 20230430. doi: 10.7774/cevr.2023.12.2.97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shiota S, Mannen K, Matsumoto T, Yamada K, Yasui T, Takayama K, et al. Development and evaluation of a rapid neutralizing antibody test for rabies. J Virol Methods. 2009;161(1):58–62. Epub 20090527. doi: 10.1016/j.jviromet.2009.05.018 . [DOI] [PubMed] [Google Scholar]
  • 44.Sugiyama M, Yoshiki R, Tatsuno Y, Hiraga S, Itoh O, Gamoh K, et al. A new competitive enzyme-linked immunosorbent assay demonstrates adequate immune levels to rabies virus in compulsorily vaccinated Japanese domestic dogs. Clin Diagn Lab Immunol. 1997;4(6):727–30. doi: 10.1128/cdli.4.6.727-730.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wasniewski M, Almeida I, Baur A, Bedekovic T, Boncea D, Chaves LB, et al. First international collaborative study to evaluate rabies antibody detection method for use in monitoring the effectiveness of oral vaccination programmes in fox and raccoon dog in Europe. J Virol Methods. 2016;238:77–85. Epub 20161014. doi: 10.1016/j.jviromet.2016.10.006 . [DOI] [PubMed] [Google Scholar]
  • 46.Wasniewski M, Guiot AL, Schereffer JL, Tribout L, Mahar K, Cliquet F. Evaluation of an ELISA to detect rabies antibodies in orally vaccinated foxes and raccoon dogs sampled in the field. J Virol Methods. 2013;187(2):264–70. Epub 20121129. doi: 10.1016/j.jviromet.2012.11.022 . [DOI] [PubMed] [Google Scholar]
  • 47.Wasniewski M, Labbe A, Tribout L, Rieder J, Labadie A, Schereffer JL, et al. Evaluation of a rabies ELISA as an alternative method to seroneutralisation tests in the context of international trade of domestic carnivores. J Virol Methods. 2014;195:211–20. Epub 20131023. doi: 10.1016/j.jviromet.2013.10.021 . [DOI] [PubMed] [Google Scholar]
  • 48.Yang LM, Zhao LZ, Hu RL, Shi ZS, Liu WJ. A novel double-antigen sandwich enzyme-linked immunosorbent assay for Measurement of Antibodies against rabies virus. Clin Vaccine Immunol. 2006;13(8):966–8. doi: 10.1128/CVI.00102-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Servat A, Feyssaguet M, Blanchard I, Morize JL, Schereffer JL, Boue F, et al. A quantitative indirect ELISA to monitor the effectiveness of rabies vaccination in domestic and wild carnivores. J Immunol Methods. 2007;318(1–2):1–10. Epub 20061102. doi: 10.1016/j.jim.2006.07.026 . [DOI] [PubMed] [Google Scholar]
  • 50.Astray RM, Jorge SA, Pereira CA. Rabies vaccine development by expression of recombinant viral glycoprotein. Arch Virol. 2017;162(2):323–32. Epub 20161031. doi: 10.1007/s00705-016-3128-9 . [DOI] [PubMed] [Google Scholar]
  • 51.Yokomizo AY, Jorge SA, Astray RM, Fernandes I, Ribeiro OG, Horton DS, et al. Rabies virus glycoprotein expression in Drosophila S2 cells. I. Functional recombinant protein in stable co-transfected cell line. Biotechnol J. 2007;2(1):102–9. doi: 10.1002/biot.200600211 . [DOI] [PubMed] [Google Scholar]
  • 52.Hu YC. Baculovirus as a highly efficient expression vector in insect and mammalian cells. Acta Pharmacol Sin. 2005;26(4):405–16. doi: 10.1111/j.1745-7254.2005.00078.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kidd IM, Emery VC. The use of baculoviruses as expression vectors. Appl Biochem Biotechnol. 1993;42(2–3):137–59. doi: 10.1007/BF02788049 . [DOI] [PubMed] [Google Scholar]
  • 54.Prehaud C, Takehara K, Flamand A, Bishop DH. Immunogenic and protective properties of rabies virus glycoprotein expressed by baculovirus vectors. Virology. 1989;173(2):390–9. doi: 10.1016/0042-6822(89)90551-5 . [DOI] [PubMed] [Google Scholar]
  • 55.Ramya R, Mohana Subramanian B, Sivakumar V, Senthilkumar RL, Sambasiva Rao KR, Srinivasan VA. Expression and solubilization of insect cell-based rabies virus glycoprotein and assessment of its immunogenicity and protective efficacy in mice. Clin Vaccine Immunol. 2011;18(10):1673–9. Epub 20110803. doi: 10.1128/CVI.05258-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Tuchiya K, Matsuura Y, Kawai A, Ishihama A, Ueda S. Characterization of rabies virus glycoprotein expressed by recombinant baculovirus. Virus Res. 1992;25(1–2):1–13. doi: 10.1016/0168-1702(92)90095-q . [DOI] [PubMed] [Google Scholar]
  • 57.Tuffereau C, Benejean J, Alfonso AM, Flamand A, Fishman MC. Neuronal cell surface molecules mediate specific binding to rabies virus glycoprotein expressed by a recombinant baculovirus on the surfaces of lepidopteran cells. J Virol. 1998;72(2):1085–91. doi: 10.1128/JVI.72.2.1085-1091.1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dietzschold B. Techniques for the purification of rabies virus, its subunits and recombinant products. In: Meslin FX, Kaplan M.M., Koprowski, editor. Laboratory techniques in rabies. Geneva: World Health Organization; 1996. p. 175–80. [Google Scholar]
  • 59.Reed LJ, Muench H. A simple method of estimating fifty per cent endpoints. Am J Hyg. 1938;27:493–7. [Google Scholar]
  • 60.WHO. Laboratory techniques in rabies. 4th ed. Geneva: World Health Organization; 1996. [Google Scholar]
  • 61.Sharada R. Expression of glycoprotein gene of rabies virus and evaluation of recombinant protein for seromonitoring of vaccinal antibodies in dogs. Bangalore, India: Karnataka Veterinary Animal and Fisheries Sciences University; 2015. [Google Scholar]
  • 62.Anonymous. Bac-to-Bac baculovirus expression system user guide. Invitrogen Life Technologies Corporation; 2015.
  • 63.Thrusfield M. Veterinary Epidemiology. Oxford, U.K.: Blackwell Science; 2007. [Google Scholar]
  • 64.Altman DG. Practical statistics for medical research. 1st ed. London: Chapman & Hall; 1991. [Google Scholar]
  • 65.Gaudin Y, Ruigrok RW, Tuffereau C, Knossow M, Flamand A. Rabies virus glycoprotein is a trimer. Virology. 1992;187(2):627–32. doi: 10.1016/0042-6822(92)90465-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Whitt MA, Buonocore L, Prehaud C, Rose JK. Membrane fusion activity, oligomerization, and assembly of the rabies virus glycoprotein. Virology. 1991;185(2):681–8. doi: 10.1016/0042-6822(91)90539-n . [DOI] [PubMed] [Google Scholar]
  • 67.Benmaamar R, Astray RM, Wagner R, Pereira CA. High-level expression of rabies virus glycoprotein with the RNA-based Semliki Forest Virus expression vector. J Biotechnol. 2009;139(4):283–90. Epub 20081224. doi: 10.1016/j.jbiotec.2008.12.009 . [DOI] [PubMed] [Google Scholar]
  • 68.Burger SR, Remaley AT, Danley JM, Moore J, Muschel RJ, Wunner WH, et al. Stable expression of rabies virus glycoprotein in Chinese hamster ovary cells. J Gen Virol. 1991;72 (Pt 2):359–67. doi: 10.1099/0022-1317-72-2-359 . [DOI] [PubMed] [Google Scholar]
  • 69.Lafon M, Wiktor TJ, Macfarlan RI. Antigenic sites on the CVS rabies virus glycoprotein: analysis with monoclonal antibodies. J Gen Virol. 1983;64 (Pt 4):843–51. doi: 10.1099/0022-1317-64-4-843 . [DOI] [PubMed] [Google Scholar]
  • 70.Shakin-Eshleman SH, Remaley AT, Eshleman JR, Wunner WH, Spitalnik SL. N-linked glycosylation of rabies virus glycoprotein. Individual sequons differ in their glycosylation efficiencies and influence on cell surface expression. J Biol Chem. 1992;267(15):10690–8. . [PubMed] [Google Scholar]
  • 71.Wojczyk B, Shakin-Eshleman SH, Doms RW, Xiang ZQ, Ertl HC, Wunner WH, et al. Stable secretion of a soluble, oligomeric form of rabies virus glycoprotein: influence of N-glycan processing on secretion. Biochemistry. 1995;34(8):2599–609. doi: 10.1021/bi00008a026 . [DOI] [PubMed] [Google Scholar]
  • 72.Wojczyk BS, Stwora-Wojczyk M, Shakin-Eshleman S, Wunner WH, Spitalnik SL. The role of site-specific N-glycosylation in secretion of soluble forms of rabies virus glycoprotein. Glycobiology. 1998;8(2):121–30. doi: 10.1093/glycob/8.2.121 . [DOI] [PubMed] [Google Scholar]
  • 73.Gaudin Y, Moreira S, J BNJ, Blondel D, Flamand A, Tuffereau C. Soluble ectodomain of rabies virus glycoprotein expressed in eukaryotic cells folds in a monomeric conformation that is antigenically distinct from the native state of the complete, membrane-anchored glycoprotein. J Gen Virol. 1999;80 (Pt 7):1647–56. doi: 10.1099/0022-1317-80-7-1647 . [DOI] [PubMed] [Google Scholar]
  • 74.Gupta PK, Sharma S, Walunj SS, Chaturvedi VK, Raut AA, Patial S, et al. Immunogenic and antigenic properties of recombinant soluble glycoprotein of rabies virus. Vet Microbiol. 2005;108(3–4):207–14. doi: 10.1016/j.vetmic.2005.04.007 . [DOI] [PubMed] [Google Scholar]
  • 75.Jallet C, Jacob Y, Bahloul C, Drings A, Desmezieres E, Tordo N, et al. Chimeric lyssavirus glycoproteins with increased immunological potential. J Virol. 1999;73(1):225–33. doi: 10.1128/JVI.73.1.225-233.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Koraka P, Bosch BJ, Cox M, Chubet R, Amerongen G, Lovgren-Bengtsson K, et al. A recombinant rabies vaccine expressing the trimeric form of the glycoprotein confers enhanced immunogenicity and protection in outbred mice. Vaccine. 2014;32(36):4644–50. Epub 20140621. doi: 10.1016/j.vaccine.2014.06.058 . [DOI] [PubMed] [Google Scholar]
  • 77.Piza AT, Pieri KM, Lusa GM, Caporale GM, Terreran MT, Machado LA, et al. Effect of the contents and form of rabies glycoprotein on the potency of rabies vaccination in cattle. Mem Inst Oswaldo Cruz. 2002;97(2):265–8. doi: 10.1590/s0074-02762002000200022 . [DOI] [PubMed] [Google Scholar]
  • 78.Rath A, Choudhury S, Batra D, Kapre SV, Rupprecht CE, Gupta SK. DNA vaccine for rabies: relevance of the trans-membrane domain of the glycoprotein in generating an antibody response. Virus Res. 2005;113(2):143–52. doi: 10.1016/j.virusres.2005.05.002 . [DOI] [PubMed] [Google Scholar]
  • 79.Ashraf S, Singh PK, Yadav DK, Shahnawaz M, Mishra S, Sawant SV, et al. High level expression of surface glycoprotein of rabies virus in tobacco leaves and its immunoprotective activity in mice. J Biotechnol. 2005;119(1):1–14. doi: 10.1016/j.jbiotec.2005.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dietzschold B, Gore M, Marchadier D, Niu HS, Bunschoten HM, Otvos L Jr., et al. Structural and immunological characterization of a linear virus-neutralizing epitope of the rabies virus glycoprotein and its possible use in a synthetic vaccine. J Virol. 1990;64(8):3804–9. doi: 10.1128/JVI.64.8.3804-3809.1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wojczyk BS, Czerwinski M, Stwora-Wojczyk MM, Siegel DL, Abrams WR, Wunner WH, et al. Purification of a secreted form of recombinant rabies virus glycoprotein: comparison of two affinity tags. Protein Expr Purif. 1996;7(2):183–93. doi: 10.1006/prep.1996.0026 . [DOI] [PubMed] [Google Scholar]
  • 82.Yadav DK, Ashraf S, Singh PK, Tuli R. Localization of rabies virus glycoprotein into the endoplasmic reticulum produces immunoprotective antigen. Protein J. 2012;31(6):447–56. doi: 10.1007/s10930-012-9420-y . [DOI] [PubMed] [Google Scholar]
  • 83.Mebatsion T, Frost JW, Krauss H. Enzyme-linked immunosorbent assay (ELISA) using staphylococcal protein A for the measurement of rabies antibody in various species. Zentralbl Veterinarmed B. 1989;36(7):532–6. doi: 10.1111/j.1439-0450.1989.tb00640.x . [DOI] [PubMed] [Google Scholar]
  • 84.Muhamuda K, Madhusudana SN, Ravi V. Development and evaluation of a competitive ELISA for estimation of rabies neutralizing antibodies after post-exposure rabies vaccination in humans. Int J Infect Dis. 2007;11(5):441–5. Epub 20070223. doi: 10.1016/j.ijid.2006.09.013 . [DOI] [PubMed] [Google Scholar]
  • 85.Thraenhart O, Kuwert EK. Enzyme immunoassay for demonstration of rabies-virus antibodies after immunisation. Lancet. 1977;2(8034):399–400. doi: 10.1016/s0140-6736(77)90321-x . [DOI] [PubMed] [Google Scholar]
  • 86.Welch RJ, Anderson BL, Litwin CM. An evaluation of two commercially available ELISAs and one in-house reference laboratory ELISA for the determination of human anti-rabies virus antibodies. J Med Microbiol. 2009;58(Pt 6):806–10. doi: 10.1099/jmm.0.006064-0 . [DOI] [PubMed] [Google Scholar]
  • 87.Bahloul C, Taieb D, Kaabi B, Diouani MF, Ben Hadjahmed S, Chtourou Y, et al. Comparative evaluation of specific ELISA and RFFIT antibody assays in the assessment of dog immunity against rabies. Epidemiol Infect. 2005;133(4):749–57. doi: 10.1017/s095026880500381x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.De Benedictis P, Mancin M, Cattoli G, Capua I, Terrregino C. Serological methods used for rabies post vaccination surveys: an analysis. Vaccine. 2012;30(38):5611–5. Epub 20120710. doi: 10.1016/j.vaccine.2012.06.089 . [DOI] [PubMed] [Google Scholar]
  • 89.Karamany RM, Kazar J, Malik SA, al-Mufti S, Badriya M. Rapid quantitative assay of rabies post-vaccination antibody by ELISA. APMIS Suppl. 1988;3:40–3. . [PubMed] [Google Scholar]
  • 90.Perrin P, Rollin PE, Sureau P. A rapid rabies enzyme immuno-diagnosis (RREID): a useful and simple technique for the routine diagnosis of rabies. J Biol Stand. 1986;14(3):217–22. doi: 10.1016/0092-1157(86)90006-5 . [DOI] [PubMed] [Google Scholar]
  • 91.Servat A, Cliquet F, Oie Reference Laboratory for Rabies WHO. Collaborative study to evaluate a new ELISA test to monitor the effectiveness of rabies vaccination in domestic carnivores. Virus Res. 2006;120(1–2):17–27. Epub 20060405. doi: 10.1016/j.virusres.2006.02.011 . [DOI] [PubMed] [Google Scholar]
  • 92.Moore SM, Pralle S, Engelman L, Hartschuh H, Smith M. Rabies vaccine response measurement is assay dependent. Biologicals. 2016;44(6):481–6. Epub 20161007. doi: 10.1016/j.biologicals.2016.09.007 . [DOI] [PubMed] [Google Scholar]
  • 93.Nale JM, Pharande RR, Majee SB, Gandge RS, Sawane MP, Ingle SA. Serosurveillance of rabies antibodies in dogs in Mumbai region by using indirect ELISA. Comp Immunol Microbiol Infect Dis. 2021;76:101655. Epub 20210424. doi: 10.1016/j.cimid.2021.101655 . [DOI] [PubMed] [Google Scholar]
  • 94.Rodriguez MC, Fontana D, Garay E, Prieto C. Detection and quantification of anti-rabies glycoprotein antibodies: current state and perspectives. Appl Microbiol Biotechnol. 2021;105(18):6547–57. Epub 20210827. doi: 10.1007/s00253-021-11515-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Moore SM, Gilbert A, Vos A, Freuling CM, Ellis C, Kliemt J, et al. Rabies Virus Antibodies from Oral Vaccination as a Correlate of Protection against Lethal Infection in Wildlife. Trop Med Infect Dis. 2017;2(3). Epub 20170721. doi: 10.3390/tropicalmed2030031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhao R, Yu P, Shan Y, Thirumeni N, Li M, Lv Y, et al. Rabies virus glycoprotein serology ELISA for measurement of neutralizing antibodies in sera of vaccinated human subjects. Vaccine. 2019;37(41):6060–7. Epub 20190827. doi: 10.1016/j.vaccine.2019.08.043 . [DOI] [PubMed] [Google Scholar]
  • 97.Scholer L, Le-Trilling VTK, Dittmer U, Fiedler M, Trilling M. Establishment and clinical validation of an in-cell-ELISA-based assay for the rapid quantification of Rabies lyssavirus neutralizing antibodies. PLoS Negl Trop Dis. 2022;16(5):e0010425. Epub 20220510. doi: 10.1371/journal.pntd.0010425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Fooks AR. Infection with RABV and other Lyssaviruses. Manual of diagnostic tests and vaccines for terrestrial animals. Paris: World Organization for Animal Health (Office Internationale des Epizooties); 2018. p. 578–612. [Google Scholar]
  • 99.Zhang S, Liu Y, Zhang F, Hu R. Competitive ELISA using a rabies glycoprotein-transformed cell line to semi-quantify rabies neutralizing-related antibodies in dogs. Vaccine. 2009;27(15):2108–13. Epub 20090206. doi: 10.1016/j.vaccine.2009.01.126 . [DOI] [PubMed] [Google Scholar]
  • 100.Fontana D, Rodriguez MC, Garay E, Russo S, Prieto C. Optimization and validation of a blocking ELISA for quantitation of anti-rabies immunoglobulins in multispecies sera. Appl Microbiol Biotechnol. 2020;104(9):4127–39. Epub 20200313. doi: 10.1007/s00253-020-10490-6 . [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Jian Xu

7 Jun 2024

PONE-D-24-15752Evaluation of the immune status of dogs vaccinated against rabies by an enzyme-linked immunosorbent assay using crude preparations of insect cells infected with a recombinant baculovirus encoding the rabies virus glycoprotein genePLOS ONE

Dear Dr. Isloor,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process. As you can see the comments from our reviewers, they pointed out several major and minor issues for your manuscript. The editor also suggests that the figures (e.g., Fig.1 and Fig.2) need significant improvement to meet the standards and quality requirements of this journal. Please provide also the raw gel images and data points used for all figures. 

Please submit your revised manuscript by Jul 22 2024 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Jian Xu, Ph.D.

Academic Editor

PLOS ONE

Journal requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. In your Methods section, please provide additional details regarding participant consent from the owners of the animals. In the ethics statement in the Methods and online submission information, please ensure that you have specified (1) whether consent was informed and (2) what type you obtained (for instance, written or verbal). If the need for consent was waived by the ethics committee, please include this information.

3. PLOS requires an ORCID iD for the corresponding author in Editorial Manager on papers submitted after December 6th, 2016. Please ensure that you have an ORCID iD and that it is validated in Editorial Manager. To do this, go to ‘Update my Information’ (in the upper left-hand corner of the main menu), and click on the Fetch/Validate link next to the ORCID field. This will take you to the ORCID site and allow you to create a new iD or authenticate a pre-existing iD in Editorial Manager. Please see the following video for instructions on linking an ORCID iD to your Editorial Manager account: https://www.youtube.com/watch?v=_xcclfuvtxQ

4. We note that you have included the phrase “data not shown” in your manuscript. Unfortunately, this does not meet our data sharing requirements. PLOS does not permit references to inaccessible data. We require that authors provide all relevant data within the paper, Supporting Information files, or in an acceptable, public repository. Please add a citation to support this phrase or upload the data that corresponds with these findings to a stable repository (such as Figshare or Dryad) and provide and URLs, DOIs, or accession numbers that may be used to access these data. Or, if the data are not a core part of the research being presented in your study, we ask that you remove the phrase that refers to these data.

5. PLOS ONE now requires that authors provide the original uncropped and unadjusted images underlying all blot or gel results reported in a submission’s figures or Supporting Information files. This policy and the journal’s other requirements for blot/gel reporting and figure preparation are described in detail at https://journals.plos.org/plosone/s/figures#loc-blot-and-gel-reporting-requirements and https://journals.plos.org/plosone/s/figures#loc-preparing-figures-from-image-files. When you submit your revised manuscript, please ensure that your figures adhere fully to these guidelines and provide the original underlying images for all blot or gel data reported in your submission. See the following link for instructions on providing the original image data: https://journals.plos.org/plosone/s/figures#loc-original-images-for-blots-and-gels.  

In your cover letter, please note whether your blot/gel image data are in Supporting Information or posted at a public data repository, provide the repository URL if relevant, and provide specific details as to which raw blot/gel images, if any, are not available. Email us at plosone@plos.org if you have any questions.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The article by Santosh et al., entitled "Evaluation of the immune status of dogs vaccinated against rabies by an enzyme-linked immunosorbent assay using crude preparations of insect cells infected with a recombinant baculovirus encoding the rabies virus glycoprotein gene," addresses the challenge of evaluating rabies vaccination effectiveness in dogs by proposing an ELISA as a practical alternative to the currently recommended tests, RFFIT and FAVN, which involve the handling of live rabies virus. The authors developed an ELISA using partially purified extracts of insect cells infected with baculovirus expressing the rabies virus glycoprotein (RV G). This method was evaluated against sera from vaccinated dogs, showing good concordance with RFFIT, with sensitivity and specificity of 90.48% and 80.00%, respectively. The proposed system offers a quick screening tool to determine the presence and approximate level of antibodies, and it holds potential for monitoring mass vaccination programs and certifying dogs for international travel. The study is well-conceived and contributes valuable insights into rabies vaccination monitoring. Thus, the manuscript can be accepted after minor revisions.

Minor Revisions

1. Introduction: Include the new species name Lyssavirus rabies according to ICTV (https://ictv.global/report/chapter/rhabdoviridae/rhabdoviridae/lyssavirus).

2. Sentence Improvement: Improve the sentence “However, there is no means to verify the success of the vaccination programmes other than keeping a record of the vaccination.” to provide a clearer context.

3. Cell Line Description: On page 10, line 206, specify which cell line was used and provide a detailed description.

4. Figure 1: Ensure the order of the description of the wells matches the order shown in the image to avoid confusion for the readers.

5. Figure 2C: Clarify which fraction was submitted by specifying “One of the fractions (indicate which) was submitted...”

6. Table 3: Include a footnote indicating that the kappa (κ) value of 0.61–0.80 is considered “substantial agreement” and not “almost perfect agreement.”

7. ELISA Antigen Explanation: Provide a better explanation as to why the purified G protein was not used to perform the ELISA, and describe the difficulties encountered in the purification step.

8. Comparison with Other Studies: Include a comparison with values found in other studies:

- Cliquet F, McElhinney LM, Servat A, Boucher JM, Lowings JP, Goddard T, Mansfield KL, Fooks AR (2004) Development of a qualitative indirect ELISA for the measurement of rabies virus-specific antibodies from vaccinated dogs and cats. J Virol Methods 117:1–8. https://doi.org/10.1016/j.jviromet.2003.12.001.

- Cliquet F, Müller T, Mutinelli F, Geronutti S, Brochier B, Selhorst T, Schereffer JL, Krafft N, Burow J, Schameitat A, Schlüter H, Aubert M (2003) Standardisation and establishment of a rabies ELISA test in European laboratories for assessing the efficacy of oral fox vaccination campaigns. Vaccine 21:2986–2993. https://doi.org/10.1016/S0264-410X(03)00102-6.

- Cliquet F, Sagné L, Schereffer JL, Aubert MF (2000) ELISA test for rabies antibody titration in orally vaccinated foxes sampled in the fields. Vaccine 18:3272–3279. https://doi.org/10.1016/S0264-410X(00)00127-4.

- Wasniewski M, Cliquet F. Evaluation of ELISA for detection of rabies antibodies in domestic carnivores. J Virol Methods. 2012 Jan;179(1):166-75. doi: 10.1016/j.jviromet.2011.10.019.

- Zajac MD (2019) Development and evaluation of a rabies enzyme-linked immunosorbent assay (ELISA) targeting IgM and IgG in human sera. https://doi.org/10.13140/RG.2.2.32365.54242.

- Zhao R, Yu P, Shan Y, Thirumeni N, Li M, Lv Y, Li J, Ren W, Huang L, Wei J, Sun Y, Zhu W, Sun L (2019) Rabies virus glycoprotein serology ELISA for measurement of neutralizing antibodies in sera of vaccinated human subjects. Vaccine 37:6060–6067. https://doi.org/10.1016/j.vaccine.2019.08.043.

9. Limitations of the Study: The authors should include a sentence in the discussion topic that reports the limitations of the study.

10. In the study by Kramps et al. (1994), the authors demonstrated that their blocking ELISA was superior to a commercially available indirect ELISA and to the 24-hour virus neutralization test in detecting low antibody levels in serum. Additionally, this blocking ELISA was capable of detecting specific antibodies in serum as early as 7 days post-infection. Thus, it would be beneficial to evaluate whether the newly developed ELISA here can detect antibodies as early as 7 days post-infection, similar to Kramps et al. studies.

Kramps JA, Magdalena J, Quak J, Weerdmeester K, Kaashoek MJ, Maris-Veldhuis MA, Rijsewijk FA, Keil G, van Oirschot JT. A simple, specific, and highly sensitive blocking enzyme-linked immunosorbent assay for detection of antibodies to bovine herpesvirus 1. J Clin Microbiol. 1994 Sep;32(9):2175-81. doi: 10.1128/jcm.32.9.2175-2181.1994.

Kramps JA, Banks M, Beer M, Kerkhofs P, Perrin M, Wellenberg GJ, Oirschot JT. Evaluation of tests for antibodies against bovine herpesvirus 1 performed in national reference laboratories in Europe. Vet Microbiol. 2004 Sep 8;102(3-4):169-81. doi: 10.1016/j.vetmic.2004.07.003.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2024 Dec 3;19(12):e0314516. doi: 10.1371/journal.pone.0314516.r002

Author response to Decision Letter 0


28 Oct 2024

Responses to reviewers

We thank the reviewer for a very detailed scrutiny of the manuscript. The suggestions have significantly improved the manuscript. Provided below are point by point responses to the suggestions.

Reviewer #1

1. Introduction: Include the new species name Lyssavirus rabies according to ICTV (https://ictv.global/report/chapter/rhabdoviridae/rhabdoviridae/lyssavirus). We have also changed the abbreviation of rabies virus from RV to RABV throughout the manuscript, in accordance with ICTV.

Response: This has been included right in the beginning of the Introduction (see line 47 in the clean copy of the revised manuscript).

2. Sentence Improvement: Improve the sentence “However, there is no means to verify the success of the vaccination programmes other than keeping a record of the vaccination.” to provide a clearer context.

Response: Context has been provided (see lines 55-58 in the clean copy of the revised manuscript)).

3. Cell Line Description: On page 10, line 206, specify which cell line was used and provide a detailed description.

Response: Cell line specification and description are now provided (see lines 247-250 in the clean copy of the revised manuscript)).

4. Figure 1: Ensure the order of the description of the wells matches the order shown in the image to avoid confusion for the readers.

Response: We thank the reviewer for pointing this out. The description has now been modified for all the three panels for clarity (see lines 298, 306 and 309 in the clean copy of the revised manuscript).

5. Figure 2C: Clarify which fraction was submitted by specifying “One of the fractions (indicate which) was submitted...”

Response: It was pooled fraction 5-8, and this has been specified now (see line 337 in the clean copy of the revised manuscript).

6. Table 3: Include a footnote indicating that the kappa (κ) value of 0.61–0.80 is considered “substantial agreement” and not “almost perfect agreement.”

Response: A foot note has been added to the Table (see lines 391-392 in the clean copy of the revised manuscript). It may be noted that we had stated it to be ‘good’ agreement in the description (see line 388 in the clean copy of the revised manuscript).

7. ELISA Antigen Explanation: Provide a better explanation as to why the purified G protein was not used to perform the ELISA, and describe the difficulties encountered in the purification step.

Response: A sentence is now included in the Discussion (see lines 419-423 in the clean copy of the revised manuscript).

8. Comparison with Other Studies: Include a comparison with values found in other studies:

- Cliquet F, McElhinney LM, Servat A, Boucher JM, Lowings JP, Goddard T, Mansfield KL, Fooks AR (2004) Development of a qualitative indirect ELISA for the measurement of rabies virus-specific antibodies from vaccinated dogs and cats. J Virol Methods 117:1–8. https://doi.org/10.1016/j.jviromet.2003.12.001.

- Cliquet F, Müller T, Mutinelli F, Geronutti S, Brochier B, Selhorst T, Schereffer JL, Krafft N, Burow J, Schameitat A, Schlüter H, Aubert M (2003) Standardisation and establishment of a rabies ELISA test in European laboratories for assessing the efficacy of oral fox vaccination campaigns. Vaccine 21:2986–2993. https://doi.org/10.1016/S0264-410X(03)00102-6.

- Cliquet F, Sagné L, Schereffer JL, Aubert MF (2000) ELISA test for rabies antibody titration in orally vaccinated foxes sampled in the fields. Vaccine 18:3272–3279. https://doi.org/10.1016/S0264-410X(00)00127-4.

- Wasniewski M, Cliquet F. Evaluation of ELISA for detection of rabies antibodies in domestic carnivores. J Virol Methods. 2012 Jan;179(1):166-75. doi: 10.1016/j.jviromet.2011.10.019.

- Zajac MD (2019) Development and evaluation of a rabies enzyme-linked immunosorbent assay (ELISA) targeting IgM and IgG in human sera. https://doi.org/10.13140/RG.2.2.32365.54242.

- Zhao R, Yu P, Shan Y, Thirumeni N, Li M, Lv Y, Li J, Ren W, Huang L, Wei J, Sun Y, Zhu W, Sun L (2019) Rabies virus glycoprotein serology ELISA for measurement of neutralizing antibodies in sera of vaccinated human subjects. Vaccine 37:6060–6067. https://doi.org/10.1016/j.vaccine.2019.08.043.

Response: We appreciate the reviewer for suggesting and drawing our attention to this. We have now added an entire paragraph in the Discussion on this (lines 471-503 in the clean copy of the revised manuscript), but have focussed on those studies which have tested for sera from animals, particularly carnivores. Although we agree with the importance of application of such tests in humans, in the context of our assay, studies using human sera only have been omitted.

9. Limitations of the Study: The authors should include a sentence in the discussion topic that reports the limitations of the study.

Response: Two sentences have been added at the end of Discussion (lines 508-511 in the clean copy of the revised manuscript).

10. In the study by Kramps et al. (1994), the authors demonstrated that their blocking ELISA was superior to a commercially available indirect ELISA and to the 24-hour virus neutralization test in detecting low antibody levels in serum. Additionally, this blocking ELISA was capable of detecting specific antibodies in serum as early as 7 days post-infection. Thus, it would be beneficial to evaluate whether the newly developed ELISA here can detect antibodies as early as 7 days post-infection, similar to Kramps et al. studies.

Kramps JA, Magdalena J, Quak J, Weerdmeester K, Kaashoek MJ, Maris-Veldhuis MA, Rijsewijk FA, Keil G, van Oirschot JT. A simple, specific, and highly sensitive blocking enzyme-linked immunosorbent assay for detection of antibodies to bovine herpesvirus 1. J Clin Microbiol. 1994 Sep;32(9):2175-81. doi: 10.1128/jcm.32.9.2175-2181.1994.

Kramps JA, Banks M, Beer M, Kerkhofs P, Perrin M, Wellenberg GJ, Oirschot JT. Evaluation of tests for antibodies against bovine herpesvirus 1 performed in national reference laboratories in Europe. Vet Microbiol. 2004 Sep 8;102(3-4):169-81. doi: 10.1016/j.vetmic.2004.07.003.

Response: Although we understand the argument for detection of antibodies as early as possible, we are not clear about the context of the studies suggested by the reviewer. They refer to bovine herpesvirus 1, and not to rabies. In addition, the context of the ELISA that we have developed is for monitoring antibodies after vaccination, whereas the mentioned studies about BHV-1 are after infection, which is impractical or inconsequential for rabies. The fundamental difference in these two cases is that the one for BHV-1 is for diagnostic purposes and the one for rabies is for monitoring purposes. Furthermore, early detection in case of rabies may not have a bearing on the potential to protect since early responses are dominated by IgM. It is thought that IgM is not as efficient as IgG against rabies virus due to its inability to penetrate interstitial tissue that has been impregnated with high levels of virus through the saliva in the bite of a rabid animal (Kennedy et al. 2007; Fooks 2018). Indeed, driving sustained IgG production is one of the main targets of vaccination against rabies (Flamand et al. 1993; Dorfmeier et al. 2013). Although we agree that a time-lapse experiment on the appearance of potentially protective antibodies is required, we feel that this discussion would be deviant from the focus of the manuscript.

References:

1. Dorfmeier, C.L.; Shen, S.; Tzvetkov, E.P.; McGettigan, J.P. 2013. Reinvestigating the role of IgM in rabies virus postexposure vaccination. J. Virol. 87:9217-9222.

2. Flamand, A.; Raux, H.; Gaudin, Y.; Ruigrok, R.W. 1993. Mechanisms of rabies virus neutralization. Virology 194:302-313.

3. Fooks, A. 2018. Rabies (Infection with RABV and Other Lyssaviruses). In Manual of Diagnotics Tests and Vaccines for Terrestrial Animals; World Organisation for Animal Health (OIE), Paris, France, 2018; pp. 578–612.

4. Kennedy, L.J.; Lunt, M.; Barnes, A.; McElhinney, L.; Fooks, A.R.; Baxter, D.N.; Ollier, W.E. 2007. Factors influencing the antibody response of dogs vaccinated against rabies. Vaccine 25:8500-8507.

Relevant Editorial Comments:

1. We note that you have included the phrase “data not shown” in your manuscript. Unfortunately, this does not meet our data sharing requirements. PLOS does not permit references to inaccessible data. We require that authors provide all relevant data within the paper, Supporting Information files, or in an acceptable, public repository. Please add a citation to support this phrase or upload the data that corresponds with these findings to a stable repository (such as Figshare or Dryad) and provide and URLs, DOIs, or accession numbers that may be used to access these data. Or, if the data are not a core part of the research being presented in your study, we ask that you remove the phrase that refers to these data.

Response: The phrase has been removed (see line 292 in the revised manuscript).

2. PLOS ONE now requires that authors provide the original uncropped and unadjusted images underlying all blot or gel results reported in a submission’s figures or Supporting Information files. This policy and the journal’s other requirements for blot/gel reporting and figure preparation are described in detail at https://journals.plos.org/plosone/s/figures#loc-blot-and-gel-reporting-requirements and https://journals.plos.org/plosone/s/figures#loc-preparing-figures-from-image-files. When you submit your revised manuscript, please ensure that your figures adhere fully to these guidelines and provide the original underlying images for all blot or gel data reported in your submission. See the following link for instructions on providing the original image data: https://journals.plos.org/plosone/s/figures#loc-original-images-for-blots-and-gels.

Response: Images of raw gels/blots are provided as a Supplementary file. It may be noted that panels 1B and 2A are already full images. The Supplementary file hence contains the images for panels 1A, 1B, 2B and 2C.

Attachment

Submitted filename: Responses to reviewers.docx

pone.0314516.s003.docx (19.7KB, docx)

Decision Letter 1

Jian Xu

12 Nov 2024

Evaluation of the immune status of dogs vaccinated against rabies by an enzyme-linked immunosorbent assay using crude preparations of insect cells infected with a recombinant baculovirus encoding the rabies virus glycoprotein gene

PONE-D-24-15752R1

Dear Dr. Isloor,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager® and clicking the ‘Update My Information' link at the top of the page. If you have any questions relating to publication charges, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Jian Xu, Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The author has addressed all the questions and made the necessary revisions in the manuscript, which is now ready for acceptance in PLOS ONE.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

**********

Acceptance letter

Jian Xu

20 Nov 2024

PONE-D-24-15752R1

PLOS ONE

Dear Dr. Isloor,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

If revisions are needed, the production department will contact you directly to resolve them. If no revisions are needed, you will receive an email when the publication date has been set. At this time, we do not offer pre-publication proofs to authors during production of the accepted work. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few weeks to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Jian Xu

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Table. Details of the samples with RFFIT and ELISA values.

    (DOCX)

    pone.0314516.s001.docx (68.4KB, docx)
    S1 Raw images

    (PPTX)

    pone.0314516.s002.pptx (2.7MB, pptx)
    Attachment

    Submitted filename: Responses to reviewers.docx

    pone.0314516.s003.docx (19.7KB, docx)

    Data Availability Statement

    All relevant data are within the manuscript and its Supporting information files.


    Articles from PLOS ONE are provided here courtesy of PLOS

    RESOURCES