Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Extensive recoding of dengue virus type 2 specifically reduces replication in primate cells without gain-of-function in Aedes aegypti mosquitoes

  • Charles B. Stauft,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Stony Brook University, Department of Molecular Genetics and Microbiology, Stony Brook University School of Medicine, Stony Brook, New York, United States of America, Codagenix, Incorporated, Farmingdale, New York, United States of America

  • Sam H. Shen,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Stony Brook University, Department of Molecular Genetics and Microbiology, Stony Brook University School of Medicine, Stony Brook, New York, United States of America

  • Yutong Song,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Stony Brook University, Department of Molecular Genetics and Microbiology, Stony Brook University School of Medicine, Stony Brook, New York, United States of America

  • Oleksandr Gorbatsevych,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Stony Brook University, Department of Molecular Genetics and Microbiology, Stony Brook University School of Medicine, Stony Brook, New York, United States of America

  • Emmanuel Asare,

    Roles Visualization, Writing – original draft, Writing – review & editing

    Affiliation Stony Brook University, Department of Molecular Genetics and Microbiology, Stony Brook University School of Medicine, Stony Brook, New York, United States of America

  • Bruce Futcher,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Stony Brook University, Department of Molecular Genetics and Microbiology, Stony Brook University School of Medicine, Stony Brook, New York, United States of America

  • Steffen Mueller,

    Roles Writing – original draft, Writing – review & editing

    Affiliations Stony Brook University, Department of Molecular Genetics and Microbiology, Stony Brook University School of Medicine, Stony Brook, New York, United States of America, Codagenix, Incorporated, Farmingdale, New York, United States of America

  • Anne Payne,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Wadsworth Center, New York State Department of Health, Slingerlands, New York, United States of America

  • Matthew Brecher,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Wadsworth Center, New York State Department of Health, Slingerlands, New York, United States of America

  • Laura Kramer,

    Roles Conceptualization, Investigation, Project administration, Supervision, Writing – original draft, Writing – review & editing

    Affiliations Wadsworth Center, New York State Department of Health, Slingerlands, New York, United States of America, School of Public Health, State University of New York at Albany, Rensselaer, New York, United States of America

  • Eckard Wimmer

    Roles Conceptualization, Funding acquisition, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing

    eckard.wimmer@stonybrook.edu

    Affiliations Stony Brook University, Department of Molecular Genetics and Microbiology, Stony Brook University School of Medicine, Stony Brook, New York, United States of America, Codagenix, Incorporated, Farmingdale, New York, United States of America

Abstract

Dengue virus (DENV), an arthropod-borne (“arbovirus”) virus, causes a range of human maladies ranging from self-limiting dengue fever to the life-threatening dengue shock syndrome and proliferates well in two different taxa of the Animal Kingdom, mosquitoes and primates. Mosquitoes and primates show taxonomic group-specific intolerance to certain codon pairs when expressing their genes by translation. This is called “codon pair bias”. By necessity, dengue viruses evolved to delicately balance this fundamental difference in their open reading frames (ORFs). We have undone the evolutionarily conserved genomic balance in the DENV2 ORF sequence and specifically shifted the encoding preference away from primates. However, this recoding of DENV2 raised concerns of ‘gain-of-function,’ namely whether recoding could inadvertently increase fitness for replication in the arthropod vector. Using mosquito cell lines and two strains of Aedes aegypti we did not observe any increase in fitness in DENV2 variants codon pair deoptimized for humans. This ability to disrupt and control DENV2’s host preference has great promise towards developing the next generation of synthetic vaccines not only for DENV but for other emerging arboviral pathogens such as chikungunya virus and Zika virus.

Introduction

Synthetic biology has the potential to revolutionize the rapid development of vaccines to prevent infectious diseases as the research paradigm shifts from empirical to rational design [1,2]. Since the first demonstration of an infectious fully synthetic virus in 2002 (7.5 kb) [3] and the ensuing initial societal concerns[4], rapid advances in DNA synthesis including decreased cost [5], has led to the general acceptance of synthetic organisms as a research tool[1,6,7]. This has also resulted in the rapid development of a new generation of synthetic vaccine candidates beneficial for humans and domesticated animals[1]. The use of novel beneficial variants of existing organisms for the treatment of cancer[8,9] or as vaccines[10,11] has become a new and exciting branch in molecular medical research. However, there have been apprehensions over the chemical synthesis of dangerous pathogens such as poliovirus [4] or 1918 Influenza virus [12]. Alarm has been expressed about the ethics of using this technology: while there is great promise for the production of novel vaccines to improve human health there are also risks if the technology is misused, a dilemma referred to as "Dual Use Research"[13].

DENV is an enveloped, plus stranded RNA arbovirus (genome ~11 kb) of the genus Flavivirus, family Flaviviridae. DENV is separated into 4 serotypes and is primarily transmitted by the urban-adapted Aedes aegypti mosquito, a vector that has become widely distributed in tropical and subtropical regions. Efforts leading to an effective DENV vaccine have been complicated by the requirement that it must be tetravalent. Subsequent infections with different serotypes of DENV may lead to severe or lethal disease mediated by antibody-dependent enhancement [14]. Complications with tetravalent DENV vaccines that have been recently reported call for new approaches to avoid unwanted outcomes [15,16]. Here we report further characterization of the first synthetic wild-type DENV2 based on the well-studied strain 16681[17], and several in silico designed attenuated DENV2 variants carrying large-scale, but selective, genomic recoding of the ORF.

There are multiple methods of recoding a viral genome to achieve attenuation including the introduction of random point mutations [18], scrambling of codons while maintaining natural biases [19], reduction of codon bias for the host organism [20], and, as described here, changing of codon pair bias (CPB) to negative values[2,21]. Previously, our laboratory has exploited the universal phenomenon of CPB [22,23], whereby codons are prone to pair more or less frequently than expected with one another, independently of individual codon bias. Adjacent codons can form up to 36 different pairs that can encode the same pair of amino acids. The relative frequency of these pairs of codons can be represented by the natural logarithm of the ratio of the observed codon pair frequency to the expected codon pair frequency. This ratio is referred to as a codon pair score (CPS), and codon pairs that pair more frequently will have a positive “favorable” CPS while those unlikely to form a pair will have a more negative “disfavored” score. The nonrandom distribution of preferences for codon pairs is referred to as CPB [21]. Codon pair deoptimization (encoding an ORF largely with codon pairs with negative scores, see below), e.g. lowering the CPS, of a pathogen’s genome always results in attenuation across viral orders [17,2429].

Available evidence suggests that CPB exists in all known taxa, including bacteria and yeast [22]. CPB for mammals is distinct from CPB in insects [17]. Arboviruses such as DENV, Zika virus, and chikungunya virus must balance their CPB if they wish to replicate well in these different taxa. These organisms are optimal for studying the effects of altered CPB as they must contend with the translational machinery of primates and mosquitoes. It is possible to adjust the CPS of a virus to be negative (disfavored) with respect to mammals but be neutral with respect to Aedes mosquitoes which we described previously [17].

Codon pair deoptimization (CPD) describes our method to introduce into a reading frame of the DENV coding sequence a large number of synonymous, disfavored (bad) codon pairs with negative codon pair scores without introducing mutations in the polyprotein or changing the use of existing codons. We note that the biological difference between “good” and “bad” codon pairs is small. However, if a large number of “bad” codon pairs are introduced into the ORF, the effect is disastrous for viral gene expression [21]. Our recoding strategy has raised a concern that dramatic recoding of the DENV genome may have unforeseen and dangerous consequences vis a vis the invertebrate vector. Whether through interaction between the RNAi based immune system of the mosquito [30] or some other means yet unidentified, there is a possibility that a sequence unknown to nature could have advantages that would enhance infection. We have, therefore, studied DENV variants for increased replication in mosquito cells. No apparent “gain-of-function” of these engineered strains was observed in vitro or in vivo using two different strains of Ae. aegypti.

Materials and methods

Construction of CPD and wild-type DENV2

Wild-type DENV2 virus was designed based on strain 16681 genome (Accession # U87411) and divided into four fragments incorporating 26 silent mutations as described previously [17]. CP-deoptimized viruses were recovered from transfection of C6/36 cells with capped RNA transcribed using HiScribe T7 in vitro transcription kit (New England Biolabs). Virus stocks were grown in C6/36 cells by harvesting cell culture supernatant 6 days post-infection at an MOI of 0.1. Viruses were titrated in C6/36 cells using a focus forming assay [17].

Cell cultures and virus production

Vero E6 (CCL-81), A549 (CCL-185), LLC-MK2 (CCL-7), and BHK-21 (CCL-10) cells were acquired from the American Type Tissue Culture collection (ATCC, Manassas VA) and grown in Modified Eagle’s Medium (MEM) supplemented with 10% fetal bovine serum (FBS; Gemclone) and Penicillin/Streptomycin (CellGro). U-937 (CRL-1593.2) and THP-1 (TIB-202) monocyte cells were grown in RPMI-1640 with 10% FBS and no antibiotics. U-937 and THP-1 cells were induced to become macrophages prior to experimentation as described previously [31]. All mammalian cell lines were maintained at 5% CO2 and 37°C. C6/36 cells (ATCC) were grown in Modified Eagle’s Medium (MEM) supplemented with 10% FBS, 1% non-essential amino acids (NEAA; Gibco), and 1x Penicillin/Streptomycin. Toxorhynchites amboinensis TRA-171 (CRL-1591) cells and Ae. aegypti CCL-125 cell lines were acquired from the ATCC. TRA-171 cells were grown in a 1:1 mix of L-15 and Mitsuhashi/Maramorosch medium (ATCC) supplemented with 2mM Glutamine, 0.05% BSA, 1% NEAA, and 10% heat-inactivated FBS. CCL-125 cells were grown in MEM supplemented with 1% NEAA, Penicillin/Streptomycin, and 20% FBS. C6/36 and CCL-125 cells were incubated at 28°C and 5% CO2 while TRA-171 cells were incubated at 28°C without CO2. Growth kinetics were examined in each cell line by infecting the cells with each virus at a MOI of 1.0, collecting supernatant daily for 5–10 days, and titrating the virus in C6/36 cells using a focus forming assay (FFA).

Infection of Aedes aegypti mosquitoes

Ae. aegypti mosquitoes were kindly provided by G.D. Ebel, Colorado State University, Fort Collins, CO, USA, originally collected in Poza Rica, Mexico. 3–5 day old females were fed infectious blood meals containing 4x106 FFU/mL of DENV2syn, Ehmin, NS3hmin, or NS5hmin. Blood meals were held at 37°C during feeding using a Hemotek Apparatus (Discovery Workshops). Engorged mosquitoes were sorted and maintained at 27°C with 16:8 hours light:dark cycle. Thirty individuals per group were collected at 5 days post infection for RNA isolation and detection using Taqman qRT-PCR. For the ID50 experiment, an established laboratory colony of Ae. aegypti was infected by intrathoracic injection with DENV2syn, Ehmin, NS3hmin, or NS5hmin. Viral stocks were diluted 10-fold from 200 FFU/ml to 0.2 FFU/ml in mosquito diluent solution and 4 ul injected into each mosquito. Twenty surviving mosquitoes from each group were collected on day 5 p.i. and analyzed by Taqman PCR. A non-linear sigmoidal dose response curve was generated to calculate the ID50 for each virus which were compared using Student’s t-test (GraphPad Prism v 7.04, La Jolla, CA). Additionally, the Mexican strain of Ae. aegypti was infected by intrathoracic injection with ~2 x 104 FFU of DENV2syn, Ehmin, NS3hmin, or NS5hmin as previously described [32]. Ten mosquitoes per group were collected on days 3, 6, 9, and 12 and analyzed by Taqman and FFA to see if there is any difference in rate of infection or total titer in the mosquitoes.

Results

Synthesis and analysis of synthetic dengue type 2 infectious cDNA (DENV2syn)

Following in silico design, we synthesized three live-attenuated DENV2 viruses, each individually with human codon pair deoptimized coding sequences in one specific coding region of the polyprotein, yet leaving the mosquito-specific codon pair bias nearly wild type (Fig 1). We refer to these DENV variants as Ehmin, NS3hmin, and NS5hmin. All three proteins targeted here play multiple roles in the replicative cycle of DENV [33]. The number of nucleotide changes in, and the codon pair scores of, the recoded sequences Ehmin, NS3hmin, and NS5hmin are summarized in Table 1. We note that because only synonymous codons were moved codon bias (“codon usage”) remained unchanged. Similarly, the amino acid sequences of Ehmin, NS3hmin, and NS5hmin remained wild type. By design, the codon pair scores of the hmin constructs for all three proteins are significantly more negative than that of the wt sequences whereas the scores for insect ORFs remained almost wild type[17].

thumbnail
Fig 1. Diagram of the DENV2 genome marking the polyprotein coding region and the principle coding regions with the color-coded regions indicating the recoded regions.

https://doi.org/10.1371/journal.pone.0198303.g001

thumbnail
Table 1. Relative CPS of each synthetic virus and mutations compared to wild-type.

CPS is the natural logarithm of the ratio of the observed codon pair frequency to the expected codon pair frequency for the targeted region. Silent nucleotide changes for DENV2syn were introduced to aid cloning.

https://doi.org/10.1371/journal.pone.0198303.t001

Replication of DENV2 Ehmin, NS3hmin, and NS5hmin variants in human macrophages and other primate cells

Macrophages are the predominant target cells in human dengue virus infections [34,35] and, as such, represent an in vitro model for attenuation of our synthetic DENV2 variants in the human host. Therefore, we focused on the growth properties of codon-pair deoptimized variants Ehmin, NS3hmin, and NS5hmin of DENV2syn in cultured human macrophage cells as well as other cells of primate/human origin (Fig 2). As our viruses were selectively deoptimized for attenuation in humans, we tested human THP-1 (Fig 2A) and U937 (Fig 2B) cells (induced monocyte-derived macrophages), the human lung epithelial cells A549 (Fig 2C)[36], and two primate cells lines, Vero (Fig 2D) and LLC-MK2 (Fig 2E), for virus replication. Significant 1–2 log10 reduction in Vero cells was observed with Ehmin. Attenuation was far more pronounced for NS3hmin and NS5hmin (-5 log10 reduction in titer). With NS3hmin infection of THP-1 cells, viral titers were static in cell culture supernatant, however, infection was confirmed using immune staining and visualization as shown by microscopy (S1 Fig). Together, these data confirm our hypothesis that CPD specific for human ORFs reduced the ability of the variants to proliferate in human and primate cells.

thumbnail
Fig 2. Replication of DENV2syn, Ehmin, and NS3hmin in human and primate cell lines.

Human induced macrophage cell lines THP-1 (A) and U937 (B) were infected with each virus at a MOI of 1.0 and supernatant collected on 0–3 dpi (Days Post Infection) and titrated by FFA in C6/36 cells. Primate Vero (C) and LLC-MK2(D) as well as human lung epithelial A549 (E) cell lines were infected with each virus at a MOI of 1.0 and supernatant from 2dpi titrated by FFA in C6/36 cells.

https://doi.org/10.1371/journal.pone.0198303.g002

Replication of Ehmin, NS3hmin, and NS5hmin variants in mosquito cell cultures was not affected by our recoding

CPD leads to DENV2 variants with genetic sequences never before seen in nature [17]. Using a variety of human pathogens we have shown that human codon pair deoptimization for these agents leads to deficiencies in virus proliferation[37]. These DENV ORFs were deoptimized for human CPS, but, by design the CPS for Ae aegypti was left essentially wild-type. Thus, we did not expect this recoding to affect growth in mosquito-derived cells (15).

To investigate selective attenuation by codon pair deoptimization, we originally used the mosquito cell line C6/36. This was derived from Aedes albopictus larvae and is now known to have mutations in the RNAi response. It has been proposed that these mutations (i.e., a lack of effective RNAi) facilitate infection by DENV [38]. Therefore, we carried out and report here studies using other mosquito cell cultures, specifically TRA-171 and CCL-125 cells. TRA-171 cells have been derived from the larvae of the non-blood feeding species of mosquito Toxorhynchites amboinensis [39]; they have no identified deficiencies in insect innate immunity. CCL-125 cells have been derived from larvae of Aedes aegypti [40] and were initially reported to be refractory to DENV infection [41]. Recently, however, CCL125 have been shown to support DENV2 16681 replication after high MOI infection [42]. Our synthetic DENV2syn, which is based on DENV2 16681[17], also replicated to high (≤107 FFU) titers in CCL-125 cells (Fig 3B). Growth kinetics and virus production of DENVsyn and its human-deoptimized variants (Ehmin, NS3hmin, and NS5hmin) were identical in TRA-171 (Fig 3C), CCL-125 (Fig 3D), and Ae. albopictus C6/36 cells (Fig 3E) cells. This result strongly supports our hypothesis that human specific deoptimization indeed results in selective attenuation. We conclude that while growth of CPD DENV2 was poor in primate cells, wild-type growth is retained in mosquito cells.

thumbnail
Fig 3. Growth kinetics of DENV2syn, Ehmin, NS3hmin, and NS5hmin in mosquito cell lines.

Mosquito cell lines TRA-171 (A) and CCL-125 (B) were infected with each virus at a MOI of 0.1 or 1.0, respectively, and supernatant samples titrated by FFA daily for one-week post-infection. Mosquito cell lines TRA-171 (C), CCL-125 (D), and C6/36 (E) were infected with each virus at a MOI of 1.0, 5.0, or 1.0, respectively, and samples collected after incubation at 28°C for 2 days to compare titer by FFA in C6/36 cells.

https://doi.org/10.1371/journal.pone.0198303.g003

Infection and replication in Aedes aegypti mosquitoes were not affected by our recoding

To confirm the results obtained in mosquito cell cultures that indicated selective deoptimization, we also tested DENV2syn and the human CPD variants in live Ae. aegypti mosquitoes. We tested the variants for changes in proliferation in two strains of Ae. aegypti mosquitoes using both intrathoracic inoculation and infection per os. Viral stocks were diluted ten-fold from 200 to 0.2 FFU/ml in mosquito diluent solution and 4 μL inoculated into female Ae. aegypti). Mosquitoes were collected on day 5 and analyzed by Taqman RT-PCR to calculate the infectious-dose 50% (ID50). No significant difference in ID50 was observed between the DENV2hmin variants and wild-type DENV2 16681 by t-test (P>0.09; Table 2).

thumbnail
Table 2. ID50 of intrathoracically inoculated Aedes aegypti.

https://doi.org/10.1371/journal.pone.0198303.t002

We further tested viral replication in mosquitoes by using both intrathoracic injection (to bypass the midgut infection and replication barriers) as well as infectious bloodmeal (to measure viral replication under more natural conditions). Adult Ae. aegypti were intracranially injected with 2 x 104 FFU virus. Mosquitoes were collected on days 3, 6, 9, and 12 p.i. and analyzed by Taqman RT-PCR to see if there were any differences in total viral RNA titer. Among infected mosquitoes, the mean viral RNA titers were statistically similar at 3, 6, 9, and 12 days post infection with significantly but slightly reduced titers on days 3 (Ehmin) and 12 (Ehmin, NS3hmin) post-inoculation (Fig 4). Ae. aegypti were then fed infectious bloodmeals containing ~4.0 x 106 FFU/mL and tested for infection by Taqman RT-PCR. The rates of infection were low, but similar for each deoptimized virus compared to wild-type (Table 3).

thumbnail
Fig 4. Infection kinetics in intrathoracically inoculated Aedes aegypti.

Mosquitoes were inoculated with ~2 x 104 FFU of each DENV strain and carcasses tested for viral RNA by Taqman qRT-PCR at 3, 6, 9, and 12 days post infection.

https://doi.org/10.1371/journal.pone.0198303.g004

thumbnail
Table 3. Aedes aegypti infected with synthetic wild-type and deoptimized viruses.

https://doi.org/10.1371/journal.pone.0198303.t003

Attenuation of CPD DENV by increased temperature sensitivity

The phenotype of temperature-sensitivity plays a significant role in some live virus vaccines as, for example, in live attenuated influenza vaccines [43] or the oral poliovirus vaccine [44]. Moreover, temperature sensitivity has been previously reported as a phenotype for codon-pair-deoptimized human respiratory syncytial viruses (RSV)[26,45]. Therefore, we examined deoptimized DENV2 variants Ehmin and NS3hmin for temperature sensitivity in infected Vero cells (Fig 5). As expected of selective human deoptimization, Ehmin and NS3hmin replication was significantly reduced in Vero cells of primate origin (Fig 5B). Because each variant replicates identically and forms equivalent FFU in C6/36 cells [17], all samples were assayed by FFA in C6/36 cells.

thumbnail
Fig 5. Growth curves at 32, 37, 39°C for DENV2syn, Ehmin, NS3hmin in Vero cells.

Vero cells were infected at a MOI of 0.01 and infected cell supernatant titrated in C6/36 cells for 6 days post infection (A). Differences in titer between infected cells grown at 32°C and 37°C or 39°C were compared for each DENV variant (B).

https://doi.org/10.1371/journal.pone.0198303.g005

Briefly, DENV2syn, Ehmin, or NS3hmin were used to infect Vero cells at a multiplicity-of-infection (MOI) of 0.01 at 32°C, 37°C, or 39°C (Fig 5). Infected cells were incubated at the respective temperature and samples were taken daily for 6 days-post-infection (DPI). DENV2syn replication was not affected by incubation at 32°C or 37°C (Fig 5A), however, at 39°C a 2 log10 FFU/mL drop (Fig 5B) in maximum titer was observed. In Ehmin infected cells, infectious virus was recovered at all three temperatures (Fig 3A) with a 2–3 log10 FFU/mL drop between 32°C and 37°C and an even greater drop of nearly 4 log10 maximum titer (6 dpi) was observed between 32°C and 39°C (Fig 3B). Infection of Vero cells with NS3hmin revealed a similar trend in growth kinetics with reductions in maximum titer like those observed for Ehmin (Fig 5). Temperature sensitivity experiments were attempted in C6/36 cells; however, the cultures were nonviable at 33°C and 37°C.

The spread of DENV2 variants in cell culture by focus forming assay (FFA) was also used to measure viral attenuation. FFA was conducted with each variant after simultaneous incubation at 28°C, 32°C, 37°C, or 39°C. Focus forming unit (FFU) size was not visibly different for DENV2syn between 28°C and 37°C in Vero cells, however, a slight decrease in FFU size was observed at 39°C. This was in stark contrast for Ehmin FFU: at 32°C, 37°C, and 39°C the FFU size decreased dramatically with increasing temperature (not shown). FFU size at 28°C was like that at 32°C, and at no temperature was parity observed between DENV2syn and Ehmin FFU size. The NS5hmin and NS3hmin variants did not produce FFU at any temperature after up to 8 days of incubation. Therefore, while DENV2 is somewhat temperature sensitive, the human deoptimized viruses appear to be more sensitive to increased temperature.

Discussion

We have recoded individually three coding regions of the DENV2 polyprotein according to a master plan: increasing the number of “bad” codon pairs (negative codon pair scores) to reduce expression in primate cells while retaining a wild type average of codon pairs for wildtype expression in mosquito cells (Fig 1A). Since wildtype DENV can grow in Ae. species mosquitoes and humans, we speculated that the codon pairs used by DENV have a fine balance between invertebrate vector and vertebrate host. We have destroyed this calibrated, natural equilibrium through large-scale recoding (15) thereby generating DENV2 Ehmin, NS3hmin, and NS5hmin variants.

The calculated disruption of human codon pair score in CPD DENV2 variants resulted in attenuation using clinically relevant cultured human macrophage cells that we had not previously used. Moreover, CPD DENV2 variants were attenuated in a panel of different primate and human cell lines and are being tested for attenuation, immunogenicity and protection from challenge in Rhesus macaques at the Caribbean Primate Research Center in Puerto Rico (manuscript in preparation).

To address concerns about ‘gain-of-function’ and demonstrate the specificity of attenuation in CP-deoptimized DENV strains, we initially conducted limited experiments in C6/36 cells. We have confirmed here in other mosquito cell lines as well as in two strains of live Ae aegypti that our recoded DENV strains do not exhibit any unforeseen effects on replication. Our previous studies with DENV2syn indicate that it is possible to leverage the codon pair bias difference between humans and mosquitoes to selectively attenuate the virus in one but not the other [17]. We chose to initiate our studies with DENV2 16681, a well-studied laboratory strain that has a passage history in mammalian cells and has formed the basis of vaccine candidate in clinical trials[46,47]. Since then, we have initiated research into CHIKV and ZIKV. Our CPD approach for attenuating flaviviruses is being independently pursued for Yellow Fever virus [48] and Zika virus[49]. Although this work is limited to DENV2 16681, we hope to extend our method of selective deoptimization to the other three DENV serotypes, and to other mosquito-borne viruses including emerging or re-emerging threats such as Zika virus [50], yellow fever virus, and chikungunya virus [51].

The mechanism of attenuation by codon-pair deoptimization is under investigation by our lab and others but remains to be solved. We suggest that it is likely the result of multiple factors; these include one or several parameters of the following:(i) the increased frequencies of CpG and UpA dinucleotides[5254], possibly linked to an activation of the innate immune response, (ii) temperature sensitivity of the expression of recoded ORFs [26,29], (iii) stability of the recoded mRNAs[24,29], and (iv) possible complications with protein folding [28]. Unlike our previous work with Influenza A virus, dengue virus expresses its genome as a polyprotein. This is intriguing as the ribosome travels through the entire ORF thereby encountering individual segments of codon pair deoptimized sequences. Unlike the loss of protein expression observed in CPD Influenza viruses [24,29] or reduced protein expression in a codon pair deoptimized poliovirus variant [49]we do not have evidence of specific loss of recoded protein expression in DENV2syn infected cells. Importantly, neither decreased temperature (Fig 5), nor addition of innate immune inhibitor TBK-1 (not shown), nor addition of Jak Inhibitor I [17] completely rescued replication of the deoptimized DENV variants. However, in BHK (Baby Hamster Kidney) cells, which are deficient in RIG-I signaling [55], our viruses replicated identically to wild-type [17] which may indicate the role of this pathway in CPD DENV attenuation.

An intriguing report has recently been published that CpG dinucleotides are involved in the recognition of RNA by the Zinc-finger antiviral protein “ZAP”, leading to degradation of mRNA [18]. Indeed, in Table 4 we have enlisted the increase of CpG dinucleotide frequency in the CPD variants. The function of ZAP would link the innate immune response to the recognition of codon pair deoptimized viral genomes against DENV [18] and resulting degradation of viral RNA[56].

Specifically relevant to DENV, CpG dinucleotides, while depleted in mammalian mRNAs, are observed with predicted frequency and show no downward bias in insects though UpA are depleted in the genomes of both insects and humans[57,58]. Although UpA frequencies were elevated in all three recoded DENV2 variants, there did not seem to be any adverse effects of this in either cell culture or live mosquitoes. These differences would impose contrasting selective pressures on DENV and other arboviruses which alternate replication in vertebrate hosts and arthropod vectors[59]. The CpG and UpA frequencies are more commonly found across codons in rare codon pairs, and so were increased in all of our CPD DENV2 variants. This increase was unavoidable and is very difficult to completely separate from changes due solely to codon pair bias, though there are current efforts in our lab to separate the two phenomena in poliovirus [60].

Supporting information

S1 Fig. Deconvolution microscopy of THP-1 cells infected with DENV2syn, Ehmin, NS3hmin at an MOI of 5.0 or mock-infected.

At 2 days post infection, infected cells were fixed and stained using 4G2 anti-E or anti-NS3 primary antibodies as well as Alexa 488 (E) or fluorescein isothiocyanate (NS3) conjugated secondary antibodies.

https://doi.org/10.1371/journal.pone.0198303.s001

(PDF)

Acknowledgments

We thank our colleagues J. Cello and J. Mugavero for discussion of aspects of this work.

References

  1. 1. Stauft CB, Wimmer E. Synthetic Viruses. eLS. John Wiley & Sons, Ltd; 2017.
  2. 2. Shen SH, Futcher B, Yang C, Wimmer E. Synthetic Biology Approaches for Vaccine Development. Reviews in Cell Biology and Molecular Medicine. Wiley-VCH Verlag GmbH & Co. KGaA; 2006. Available: http://onlinelibrary.wiley.com/doi/10.1002/3527600906.mcb.20120059/abstract
  3. 3. Cello J, Paul AV, Wimmer E. Chemical synthesis of poliovirus cDNA: generation of infectious virus in the absence of natural template. Science. 2002;297: 1016–1018. pmid:12114528
  4. 4. Wimmer E. The test‐tube synthesis of a chemical called poliovirus: The simple synthesis of a virus has far‐reaching societal implications. EMBO reports. 2006;7: S3–S9. pmid:16819446
  5. 5. Mueller S, Coleman JR, Wimmer E. Putting synthesis into biology: a viral view of genetic engineering through de novo gene and genome synthesis. Chem Biol. 2009;16: 337–347. pmid:19318214
  6. 6. Wimmer E, Paul AV. Synthetic Poliovirus and Other Designer Viruses: What Have We Learned from Them? Annual Review of Microbiology. 2011;65: 583–609. pmid:21756105
  7. 7. Wimmer E, Mueller S, Tumpey TM, Taubenberger JK. Synthetic viruses: a new opportunity to understand and prevent viral disease. Nat Biotech. 2009;27: 1163–1172. pmid:20010599
  8. 8. Cello. Oncolytic poliovirus therapy and immunization with poliovirus-infected cell lysate induces potent antitumor immunity against neuroblastoma in vivo. International Journal of Oncology. 2010;38.
  9. 9. Toyoda H, Yin J, Mueller S, Wimmer E, Cello J. Oncolytic treatment and cure of neuroblastoma by a novel attenuated poliovirus in a novel poliovirus-susceptible animal model. Cancer research. 2007;67: 2857–2864. pmid:17363609
  10. 10. Coffin JM. Attenuation by a thousand cuts. N Engl J Med. 2008;359: 2283–2285. pmid:19020330
  11. 11. Mueller S, Coleman JR, Papamichail D, Ward CB, Nimnual A, Futcher B, et al. Live attenuated influenza virus vaccines by computer-aided rational design. Nat Biotechnol. 2010;28: 723–726. pmid:20543832
  12. 12. Tumpey TM, Basler CF, Aguilar PV, Zeng H, Solórzano A, Swayne DE, et al. Characterization of the Reconstructed 1918 Spanish Influenza Pandemic Virus. Science. 2005;310: 77–80. pmid:16210530
  13. 13. National Academies of Sciences E, Affairs P and G, Committee on Science T, Management C on DUR of CO for F. Managing Dual Use Research of Concern [Internet]. National Academies Press (US); 2017. Available: https://www.ncbi.nlm.nih.gov/books/NBK458500/
  14. 14. Alwis R de, Williams KL, Schmid MA, Lai C-Y, Patel B, Smith SA, et al. Dengue Viruses Are Enhanced by Distinct Populations of Serotype Cross-Reactive Antibodies in Human Immune Sera. PLOS Pathog. 2014;10: e1004386. pmid:25275316
  15. 15. Guy B, Barrere B, Malinowski C, Saville M, Teyssou R, Lang J. From research to phase III: Preclinical, industrial and clinical development of the Sanofi Pasteur tetravalent dengue vaccine. Vaccine. 2011;29: 7229–7241. pmid:21745521
  16. 16. Dyer O. Philippines halts dengue immunisation campaign owing to safety risk. BMJ. 2017;359: j5759. pmid:29233814
  17. 17. Shen SH, Stauft CB, Gorbatsevych O, Song Y, Ward CB, Yurovsky A, et al. Large-scale recoding of an arbovirus genome to rebalance its insect versus mammalian preference. Proc Natl Acad Sci USA. 2015;112: 4749–4754. pmid:25825721
  18. 18. Takata MA, Gonçalves-Carneiro D, Zang TM, Soll SJ, York A, Blanco-Melo D, et al. CG dinucleotide suppression enables antiviral defence targeting non-self RNA. Nature. 2017; pmid:28953888
  19. 19. Nougairede A, Fabritus LD, Aubry F, Gould EA, Holmes EC, Lamballerie X de. Random Codon Re-encoding Induces Stable Reduction of Replicative Fitness of Chikungunya Virus in Primate and Mosquito Cells. PLOS Pathogens. 2013;9: e1003172. pmid:23436995
  20. 20. Mueller S, Papamichail D, Coleman JR, Skiena S, Wimmer E. Reduction of the Rate of Poliovirus Protein Synthesis through Large-Scale Codon Deoptimization Causes Attenuation of Viral Virulence by Lowering Specific Infectivity. J Virol. 2006;80: 9687–9696. pmid:16973573
  21. 21. Coleman JR, Papamichail D, Skiena S, Futcher B, Wimmer E, Mueller S. Virus attenuation by genome-scale changes in codon pair bias. Science. 2008;320: 1784–1787. pmid:18583614
  22. 22. Buchan R, Stansfield I. Codon pair bias in prokaryotic and eukaryotic genomes. BMC Bioinformatics. 2005;6: P4.
  23. 23. Gutman GA, Hatfield GW. Nonrandom utilization of codon pairs in Escherichia coli. Proceedings of the National Academy of Sciences. 1989;86: 3699–3703.
  24. 24. Broadbent AJ, Santos CP, Anafu A, Wimmer E, Mueller S, Subbarao K. Evaluation of the attenuation, immunogenicity, and efficacy of a live virus vaccine generated by codon-pair bias de-optimization of the 2009 pandemic H1N1 influenza virus, in ferrets. Vaccine. 2016;34: 563–570. pmid:26655630
  25. 25. Diaz-San Segundo F, Medina GN, Ramirez-Medina E, Velazquez-Salinas L, Koster M, Grubman MJ, et al. Synonymous Deoptimization of Foot-and-Mouth Disease Virus Causes Attenuation In Vivo while Inducing a Strong Neutralizing Antibody Response. J Virol. 2015;90: 1298–1310. pmid:26581977
  26. 26. Le Nouën C, Brock LG, Luongo C, McCarty T, Yang L, Mehedi M, et al. Attenuation of human respiratory syncytial virus by genome-scale codon-pair deoptimization. Proc Natl Acad Sci USA. 2014;111: 13169–13174. pmid:25157129
  27. 27. Ni Y-Y, Zhao Z, Opriessnig T, Subramaniam S, Zhou L, Cao D, et al. Computer-aided codon-pairs deoptimization of the major envelope GP5 gene attenuates porcine reproductive and respiratory syndrome virus. Virology. 2014;450–451: 132–139. pmid:24503075
  28. 28. Wang B, Yang C, Tekes G, Mueller S, Paul A, Whelan SPJ, et al. Recoding of the vesicular stomatitis virus L gene by computer-aided design provides a live, attenuated vaccine candidate. MBio. 2015;6. pmid:25827413
  29. 29. Yang C, Skiena S, Futcher B, Mueller S, Wimmer E. Deliberate reduction of hemagglutinin and neuraminidase expression of influenza virus leads to an ultraprotective live vaccine in mice. Proc Natl Acad Sci USA. 2013;110: 9481–9486. pmid:23690603
  30. 30. Khoo CC, Piper J, Sanchez-Vargas I, Olson KE, Franz AW. The RNA interference pathway affects midgut infection- and escape barriers for Sindbis virus in Aedes aegypti. BMC Microbiology. 2010;10: 130. pmid:20426860
  31. 31. Daigneault M, Preston JA, Marriott HM, Whyte MKB, Dockrell DH. The Identification of Markers of Macrophage Differentiation in PMA-Stimulated THP-1 Cells and Monocyte-Derived Macrophages. PLOS ONE. 2010;5: e8668. pmid:20084270
  32. 32. Payne AF, Binduga-Gajewska I, Kauffman EB, Kramer LD. Quantitation of flaviviruses by fluorescent focus assay. Journal of Virological Methods. 2006;134: 183–189. pmid:16510196
  33. 33. Fields B, Knipe D, Howley P. Fields Virology. 6th ed. 2013.
  34. 34. Pryor MJ, Carr JM, Hocking H, Davidson AD, Li P, Wright PJ. Replication of dengue virus type 2 in human monocyte-derived macrophages: comparisons of isolates and recombinant viruses with substitutions at amino acid 390 in the envelope glycoprotein. Am J Trop Med Hyg. 2001;65: 427–434. pmid:11716094
  35. 35. Schaeffer E, Flacher V, Papageorgiou V, Decossas M, Fauny J-D, Krämer M, et al. Dermal CD14+ Dendritic Cell and Macrophage Infection by Dengue Virus Is Stimulated by Interleukin-4. Journal of Investigative Dermatology. 2015;135: 1743–1751. pmid:25521455
  36. 36. Chiu H-C, Hannemann H, Heesom KJ, Matthews DA, Davidson AD. High-Throughput Quantitative Proteomic Analysis of Dengue Virus Type 2 Infected A549 Cells. PLOS ONE. 2014;9: e93305. pmid:24671231
  37. 37. Stauft CB, Wimmer E. Synthetic Viruses. eLS. John Wiley & Sons, Ltd; 2018. Available: http://onlinelibrary.wiley.com/doi/10.1002/9780470015902.a0027771/abstract
  38. 38. Brackney DE, Scott JC, Sagawa F, Woodward JE, Miller NA, Schilkey FD, et al. C6/36 Aedes albopictus Cells Have a Dysfunctional Antiviral RNA Interference Response. PLOS Negl Trop Dis. 2010;4: e856. pmid:21049065
  39. 39. Kuno G. Persistent Infection of a Nonvector Mosquito Cell Line (TRA-171) with Dengue Viruses. INT. 1982;18: 45–55. pmid:6126465
  40. 40. SINGH KRP. CELL CULTURES DERIVED FROM LARVAE OF AEDES ALBOPICTUS (SKUSE) AND AEDES AEGYPTI (L.). Current Science. 1967;36: 506–508.
  41. 41. Singh KRP. Growth of Arboviruses in Aedes albopictus and A. aegypti Cell Lines. Arthropod Cell Cultures and Their Application to the Study of Viruses. Springer, Berlin, Heidelberg; 1971. pp. 127–133. Available: https://link.springer.com/chapter/10.1007/978-3-642-65224-0_20
  42. 42. Wikan N, Kuadkitkan A, Smith DR. The Aedes aegypti cell line CCL-125 is dengue virus permissive. Journal of Virological Methods. 2009;157: 227–230. pmid:19159646
  43. 43. Jin H, Zhou H, Lu B, Kemble G. Imparting Temperature Sensitivity and Attenuation in Ferrets to A/Puerto Rico/8/34 Influenza Virus by Transferring the Genetic Signature for Temperature Sensitivity from Cold-Adapted A/Ann Arbor/6/60. J Virol. 2004;78: 995–998. pmid:14694130
  44. 44. Paul AV, Mugavero J, Yin J, Hobson S, Schultz S, van Boom JH, et al. Studies on the Attenuation Phenotype of Polio Vaccines: Poliovirus RNA Polymerase Derived from Sabin Type 1 Sequence Is Temperature Sensitive in the Uridylylation of VPg. Virology. 2000;272: 72–84. pmid:10873750
  45. 45. Nouën CL, McCarty T, Brown M, Smith ML, Lleras R, Dolan MA, et al. Genetic stability of genome-scale deoptimized RNA virus vaccine candidates under selective pressure. PNAS. 2017;114: E386–E395. pmid:28049853
  46. 46. Butrapet S, Huang CY-H, Pierro DJ, Bhamarapravati N, Gubler DJ, Kinney RM. Attenuation Markers of a Candidate Dengue Type 2 Vaccine Virus, Strain 16681 (PDK-53), Are Defined by Mutations in the 5′ Noncoding Region and Nonstructural Proteins 1 and 3. J Virol. 2000;74: 3011–3019. pmid:10708415
  47. 47. Kinney RM, Butrapet S, Chang G-JJ, Tsuchiya KR, Roehrig JT, Bhamarapravati N, et al. Construction of Infectious cDNA Clones for Dengue 2 Virus: Strain 16681 and Its Attenuated Vaccine Derivative, Strain PDK-53. Virology. 1997;230: 300–308. pmid:9143286
  48. 48. Klitting R, Riziki T, Moureau G, Piorkowski G, Lamballerie X de. Exploratory re-encoding of Yellow Fever Virus genome: new insights for the design of live-attenuated viruses. bioRxiv. 2018; 256610.
  49. 49. Li P, Ke X, Wang T, Tan Z, Luo D, Miao Y, et al. Zika Virus Attenuation by Codon Pair Deoptimization Induces Sterilizing Immunity in Mouse Models. J Virol. 2018; JVI.00701-18. pmid:29925661
  50. 50. Fauci AS, Morens DM. Zika Virus in the Americas—Yet Another Arbovirus Threat. New England Journal of Medicine. 2016;0: null. pmid:26761185
  51. 51. Nhan TX, Musso D. The burden of chikungunya in the Pacific. Clinical Microbiology and Infection. 2015;21: e47–e48. pmid:25749562
  52. 52. Atkinson NJ, Witteveldt J, Evans DJ, Simmonds P. The influence of CpG and UpA dinucleotide frequencies on RNA virus replication and characterization of the innate cellular pathways underlying virus attenuation and enhanced replication. Nucleic Acids Research. 2014;42: 4527–4545. pmid:24470146
  53. 53. Futcher B, Gorbatsevych O, Shen SH, Stauft CB, Song Y, Wang B, et al. Reply to Simmonds et al.: Codon pair and dinucleotide bias have not been functionally distinguished. Proc Natl Acad Sci USA. 2015;112: E3635–3636. pmid:26071446
  54. 54. Burns CC, Campagnoli R, Shaw J, Vincent A, Jorba J, Kew O. Genetic Inactivation of Poliovirus Infectivity by Increasing the Frequencies of CpG and UpA Dinucleotides within and across Synonymous Capsid Region Codons. Journal of Virology. 2009;83: 9957–9969. pmid:19605476
  55. 55. Habjan M, Penski N, Spiegel M, Weber F. T7 RNA polymerase-dependent and -independent systems for cDNA-based rescue of Rift Valley fever virus. Journal of General Virology. 2008;89: 2157–2166. pmid:18753225
  56. 56. Zhu Y, Gao G. ZAP-mediated mRNA degradation. RNA Biology. 2008;5: 65–67. pmid:18418085
  57. 57. Simmen MW. Genome-scale relationships between cytosine methylation and dinucleotide abundances in animals. Genomics. 2008;92: 33–40. pmid:18485662
  58. 58. Campbell A, Mrazek J, Karlin S. Genome signature comparisons among prokaryote, plasmid, and mitochondrial DNA. Proceedings of the National Academy of Sciences. 1999;96: 9184–9189.
  59. 59. Lobo FP, Mota BEF, Pena SDJ, Azevedo V, Macedo AM, Tauch A, et al. Virus-Host Coevolution: Common Patterns of Nucleotide Motif Usage in Flaviviridae and Their Hosts. PLOS ONE. 2009;4: e6282. pmid:19617912
  60. 60. Song Y, Gorbatsevych O, Liu Y, Mugavero J, Shen SH, Ward CB, et al. Limits of variation, specific infectivity, and genome packaging of massively recoded poliovirus genomes. PNAS. 2017;114: E8731–E8740. pmid:28973853