key: cord-0696186-lgrk3zs6 authors: Naeem, Asif; Hamed, Maaweya E.; Alghoribi, Majed F.; Aljabr, Waleed; Alsaran, Hadel; Enani, Mushira A.; Alosaimi, Bandar title: Molecular Evolution and Structural Mapping of N-Terminal Domain in Spike Gene of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) date: 2020-05-02 journal: Viruses DOI: 10.3390/v12050502 sha: 7baf292c44a79a7159bbacdc500d29a929f7e4a6 doc_id: 696186 cord_uid: lgrk3zs6 The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a lethal zoonotic pathogen circulating in the Arabian Peninsula since 2012. There is no vaccine for MERS and anti-viral treatment is generally not applicable. We investigated the evolution of the MERS-CoV spike gene sequences and changes in viral loads over time from patients in Saudi Arabia from 2015–2017. All the MERS-CoV strains belonged to lineage 5, and showed high sequence homology (99.9%) to 2017 strains. Recombination analysis showed a potential recombination event in study strains from patients in Saudi Arabia. The spike gene showed eight amino acid substitutions, especially between the A1 and B5 lineage, and contained positively selected codon 1020. We also determined that the viral loads were significantly (p < 0.001) higher in fatal cases, and virus shedding was prolonged in some fatal cases beyond 21 days. The viral concentration peaked during the first week of illness, and the lower respiratory specimens had higher levels of MERS-CoV RNA. The presence of the diversifying selection and the topologies with the structural mapping of residues under purifying selection suggested that codon 1020 might have a role in the evolution of spike gene during the divergence of different lineages. This study will improve our understanding of the evolution of MERS-CoV, and also highlights the need for enhanced surveillance in humans and dromedaries. The presence of amino acid changes at the N-terminal domain and structural mapping of residues under positive selection at heptad repeat 1 provides better insight into the adaptive evolution of the spike gene and might have a potential role in virus-host tropism and pathogenesis. Middle East Respiratory Syndrome (MERS) was discovered in a patient who died from acute respiratory illness in Jeddah, Saudi Arabia in 2012. A novel coronavirus (MERS-CoV) was isolated from this patient [1] . The virus was initially designated as HCoV (human coronavirus)-Erasmus University Ethics approval and consent to participate: All procedures performed in this study involving clinical specimen were in accordance with the ethical standards of the institutional and national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. The study was approved by the Institutional Review Board at King Fahad Medical City (IRB Log No. 16 -346 approved on 27 September 2016). Informed consent to participate was waived or not required since only remaining left-over specimens were used for this study. Availability of data and materials: The clinical datasets used and analyzed during the current study are available from the corresponding author on request. The handling of respiratory samples, as well as preparation of aliquots and viral RNA extraction, was performed using appropriate personal protective equipment in the biosafety level 3 laboratory (Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia). The study specimens were obtained from a population (n = 13) admitted to various hospitals in Riyadh, Saudi Arabia with six associated deaths. All samples (n = 40) testing positive for MERS-CoV locally by real-time reverse transcription PCR (rRT-PCR) assay and admitted to the hospital from 1 September 2015-16 November 2017, were used for this study (Table 1 ). Written informed consent was obtained from the patients at the time of admission. Nasopharyngeal swab specimens, tracheal aspirates, sputum, or bronchoalveolar lavages were collected for laboratory-based investigations. A case of MERS, according to the Saudi Arabian Ministry of Health definition, is fever and acute respiratory illness in a patient who has a positive test result for MERS-CoV infection. Demographic information, medical history, and outcome information were collected. Respiratory specimens were used for upE [30] and ORF1a rRT-PCR assays [31] for MERS-CoV initial screening and further molecular testing. RNA was extracted from the samples as described earlier [32] using a viral RNA mini kit (Qiagen). A 2× sputum lysis buffer (10 g of N-acetylcysteine/liter, 0.9% w/v sodium chloride) was used for the treatment of sputum samples for 30 min in a shaking incubator. Swabs were dissolved in the lysis buffer. A 25-µL reaction was prepared containing 5 µL of RNA, as described previously [30] . Briefly, 12.5 µL of 2× reaction buffer provided with the Superscript III one-step RT-PCR system with Platinum Taq Polymerase (Invitrogen, Carlsbad, CA, USA), 1 µL of reverse transcriptase/Taq mixture from the kit, 0.4 µL of a 50 mM magnesium sulfate solution (Invitrogen, Carlsbad, CA, USA), 1 µL each of 10 µM forward and reverse upE primers with 400 nM concentration in the final solution, as well as 0.5 µL of 10 µM upE probe with 200 nM concentration in final solution was used. Molecular grade deionized water was used to make the final volume to 25 µL. Thermal cycling involved reverse transcription at 55 • C for 20 min, followed by denaturation at 95 • C for 3 min, and then 45 cycles of denaturation and extension at 95 • C for 15 s and 58 • C for 30 s. A 25 µL reaction was prepared containing 5 µL of RNA, as described previously [31] . Briefly, 12.5 µL of 2× reaction buffer from the Superscript III one-step RT-PCR system with Platinum Taq Polymerase (Invitrogen, Carlsbad, CA, USA), 1 µL of reverse transcriptase/Taq mixture from the kit, 0.4 µL of a 50 mM MgCl 2 solution (Invitrogen, Carlsbad, CA, USA), 1 µL each of 10 µM forward and reverse of EMC-Orf1a primers with 400 nM concentration in the final solution, as well as 0.5 µL of 10 µM EMC-Orf1a probe with 200 nM concentration in the final solution was used. Molecular grade deionized water was used to make the final volume to 25 µL. Thermal cycling involved reverse transcription at 55 • C for 20 min, followed by denaturation at 95 • C for 3 min, and then 45 cycles of denaturation and extension at 95 • C for 15 s, and 58 • C for 30 s. Extracted viral RNA was reverse transcribed using random hexamers (Invitrogen, Carlsbad, CA, USA) at 52 • C for 60 min with Superscript III reverse transcriptase (Invitrogen, Carlsbad, CA, USA) by following the manufacturer's instructions. Five µL of the cDNA was amplified by PCR using MERS-CoV specific primer sets with the Phusion Flash High Fidelity PCR master mix kit (Thermo Scientific, Carlsbad, CA, USA). PCR cycling conditions were as follows: an initial activation step at 95 • C for 15 min, followed by 35 cycles of 95 • C for 1 min, 55 • C for 1 min, and 68 • C for 2 min, with a final extension of 68 • C for 10 min on a Veriti PCR system (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA, USA). Amplified products were purified with the ExoSAP-IT Express PCR Cleanup Reagent (Thermo Fisher Scientific, Waltham, MA, USA), the nucleotide sequences of the amplified fragments from study isolates, with high copy numbers, were determined using M13-tailed PCR primers and BigDye Terminator v3.1 Cycle Sequence kit according to the manufacturer's instructions (Applied Biosystems, San Diego, CA, USA). Removal of unincorporated ddNTP's, salts, and dye blobs were performed by BigDye XTerminator Purification Kit (Applied Biosystems, San Diego, CA, USA). Capillary electrophoresis was performed in SeqStudio Sanger sequencer (Applied Biosystems, San Diego, CA, USA), and analysis of Sanger sequencing data, by assembly and editing, was conducted by Sequencher 5.4.6 (Gene Codes Corporation, Ann Arbor, MI, USA). Nucleotide sequences, except primer regions, were aligned with those retrieved from the Genbank Nucleotide Sequence Database. Inspection and manual modification and evolutionary analyses were conducted using Molecular Evolutionary Genetics Analysis Version X (MEGA-X), phylogenies were estimated using neighbor-joining and maximum likelihood methods implemented in MEGA, version X [33] . The neighbor-joining method used maximum composite likelihood distance estimation, and maximum likelihood used the Tamura-Nei (TN93) model of nucleotide substitution with γ-distributed rate variation (TN93 + G) and bootstrapping (1000 replicates). The genomic data was analyzed and submitted to a public database (GenBank) under accession numbers MK858156-MK858164 and MN735679-MN735682 (Supplementary Table S1 ). Potential recombination events among MERS-CoV S gene complete reference sequences retrieved from GenBank sequence database (n = 147) and the study strains (n = 13) (Supplementary Table S1 ) were identified using automated RDP, GENECONV, BOOTSCAN, MaxChi, CHIMAERA, and SISCAN methods provided in RDP4 software [34] . Moreover, the evidence of recombinant breakpoint was confirmed using GARD in Datamonkey web server [35] . Since recombination can affect phylogenetic tree inferences [36] , we followed the procedure presented in [37] . Thus, a phylogenetic tree based on each recombinant fragment was reconstructed after selecting the TN93 + G model with the lowest BIC score using the MEGA software version X [33] . The selective pressure on encoding S gene of MERS-CoV was examined by calculating the ratio of synonymous and non-synonymous substitutions (dN/dS, defined as ω) across lineage on a codon-by-codon basis. The individual site-specific selection pressure and ω were estimated using the single likelihood ancestor counting (SLAC), fixed effects likelihood (FEL), and mixed-effects model of evolution (MEME) methods contained in the HYPHY package [35] . All analyses utilized the Datamonkey online tool (http://www.datamonkey.org). The value of ω was estimated based on the neighbor-joining trees under the GTR substitution model. The significance level for a positively selected site by either SLAC/FEL/MEME or all three methods was accepted at 0.1. The NetNGlyc 1.0 server was used to predict the potential N-linked glycosylation sites (amino acids Asn-X-Ser/Thr, whereby X is any amino acid except for Asp or Pro) [38] . A threshold value of >0.5 for the average potential score suggests glycosylation. To measure the viral load, a one-step multiplex quantitative rRT-PCR was performed with RealStar ® MERS-CoV (upE & ORF1a) RT-PCR Kit (Altona Diagnostics, Hamburg, Germany). All assays were performed in triplicate with the 7500 Fast Real-time PCR system (Applied Biosystems ® , Grand Island, NY, USA). Positive controls of known concentrations of 1.0 × 10 5 copies per µL were kindly provided by the European Virus Archives (EVA). Each sample was tested in triplicate, and the mean of the three values was shown as the copy number of the sample. Samples were defined as negative if the copy number fell below 10. The copy number was calculated based on the standard curve generated by real-time RT-PCR using the control RNA and expressed as copies/mL. The independent t-test was used to assess the statistical significance of differences between groups. In all analyses, p < 0.05 was used to indicate statistical significance. Viral RNA copy number was compared between groups (dead and recovered) by Mann-Whitney U test, and the proportion of cases with detectable RNA was compared using Fisher's exact test. Demographic and clinical characteristics were analyzed, and differences assessed for significance by using Chi-square test, Wilcoxon rank-sum, and Kruskal-Wallis tests where appropriate. Statistical analyses were performed using the IBM SPSS Statistics software (version 22.0, IBM Corporation, Armonk, NY, USA). The 160 complete S gene sequences of MERS-CoV clustered into two major clades, A and B (Figure 1 ). Clade A had only five strains, while most of the strains clustered into clade B. Clade A has historically been divided into two lineages; lineage A1, containing the first reported MERS-CoV strain from Saudi Arabia in 2012 and two Jordan strains. While lineage A2 contained two camel strains from the United Arab Emirates (UAE) and Egypt. Clade B had a further six lineages containing human strains. Lineage B2 to B6 mostly contained the Saudi strains. B3 lineage was mostly populated by the South Korean strains, and the same strain carried to various countries like Bahrain, Qatar, China, Saudi Arabia, and the UAE during 2015. A comparative genetic analysis of 160 strains isolated in 27 countries, including Saudi Arabia, demonstrated that the circulating 13 MERS-CoV strains from our study showed the highest identity (99.9%) with the strains recently isolated in Saudi Arabia [39] [40] [41] . Based on the respiratory specimen available, S gene sequences from 13 patients appeared to have a close relationship with each other and belonged to lineage 5 [42] . There were three amino acid substitutions in the N terminal domain (NTD) of the S gene in the same comparison ( Table 2 ). The 2017 strains from our study, i.e., MK858160-Saudi Arabia-37-2017, MK858161-Saudi Arabia-40-2017, and MK858162-Saudi Arabia-41-2017 had three amino acid substitutions at S126A, F228L, and L745F, respectively. The only 2015 strain (MN735679-Saudi Arabia-03-2015) from our study also had a single mutation at S126A. In the phylogenetic analysis, the virus was located in a lineage of locally endemic 2016 and 2017 Saudi Arabia strains (lineage 5 in Figure 1 ) but was distinguished from 2015 strains. The recombination analysis showed that there was one significant (p-value ≤ 0.05) event related to the study strain 19 (major parent) and the reference strain from clade A (minor parent) that was detected by all recombinant analytical methods (RGBMCST) with all automated runs available in RDP4. The recombination detection program (RDP) plot and genetic algorithm for recombination detection (GARD) showed that the recombination occurred between nt positions 1-2541 of the major parent, and 2542-4059 of the minor parent ( Figure 2 ). This breakpoint generated two recombinant fragments indicating statistically significant incongruence in lineage 5. The first fragment (aa 1-848) involves the S1 subunit with NTD, RBD, Sub-domain 1 and 2, followed by a linker region. The second fragment (aa 848-1353) involves almost the entire S2 subunit regions. Additional phylogenetic analyses were separately performed based on two recombinant fragments. The results of the phylogenetic analyses demonstrated that, based on the recombinant fragments, the Saudi strains were grouped together with the parent strain, separated from MERS-CoV from other countries (data not shown). The results of the phylogenetic analyses, based on each recombinant fragment, confirmed the findings of recombination events generated from GARD ( Figure 2 ). We assessed the adaptive evolution of MERS-CoV, dN/dS ratios (ω) in the S gene across the 13 strains with codon-by-codon basis calculations. The S gene was expected to be under selection pressure to escape the host immune responses; therefore, the rate of change was determined by ω values where ω < 1 indicated the presence of negative or purifying selection, neutral selection pressure was indicated by ω = 1, and positive or diversifying selection was shown by ω > 1 [43] . The analysis showed that the overall ω was 0.1741737, with most of the codons having ω < 1, indicating purifying selection. However, the codon 1020 had an ω value of more than 1 by the mixed-effects model of evolution (MEME) method with statistical significance, which indicated the presence of functional constraints during the evolutionary process. Strains belonging to clade B were predicted to have codon 1020 and 1267 under positive selection. Amino acid residues in the receptor-binding domain (RBD) remained conserved and were not found to be under selection pressure. The majority of MERS-CoV strains possessed three potential glycosylation sites in the S1 subunit at amino acid positions 135 and 206 in the NTD region, and S2 subunit at amino acid position 1243 which were present in all lineage B5 strains. We did not find any mutations resulting in a loss of potential glycosylation site especially in the RBD region. analyses were separately performed based on two recombinant fragments. The results of the phylogenetic analyses demonstrated that, based on the recombinant fragments, the Saudi strains were grouped together with the parent strain, separated from MERS-CoV from other countries (data not shown). The results of the phylogenetic analyses, based on each recombinant fragment, confirmed the findings of recombination events generated from GARD ( Figure 2 ). Results of recombination analysis using genetic algorithm for recombination detection (GARD) in the Datamonkey web server. The red arrow indicates a significant breakpoint that was detected (p-value < 0.05). The x-axis represents the nucleotide site. The two recombinant fragments are presented in the green and blue bars. We assessed the adaptive evolution of MERS-CoV, dN/dS ratios (ω) in the S gene across the 13 strains with codon-by-codon basis calculations. The S gene was expected to be under selection pressure to escape the host immune responses; therefore, the rate of change was determined by ω values where ω < 1 indicated the presence of negative or purifying selection, neutral selection pressure was indicated by ω = 1, and positive or diversifying selection was shown by ω > 1 [43] . The analysis showed that the overall ω was 0.1741737, with most of the codons having ω < 1, indicating purifying selection. However, the codon 1020 had an ω value of more than 1 by the mixed-effects Figure 2 . Results of recombination analysis using genetic algorithm for recombination detection (GARD) in the Datamonkey web server. The red arrow indicates a significant breakpoint that was detected (p-value < 0.05). The x-axis represents the nucleotide site. The two recombinant fragments are presented in the green and blue bars. The models of S gene from each clade were built. We found that the amino acid substitutions observed by multiple alignments appeared to be randomly distributed throughout the structure ( Figure 3A ). The MERS-CoV S gene has 25 potential N-linked glycosylation sites ( Figure 3B ). Most of the N-linked glycosylation sites are located on the S1 subunit and the C-terminal region (including the HR2 region and the region preceding HR2) of the S2 subunit ( Figure 3B ). For FR, the HR1 region, and the central helix, there are no N-linked glycosylation sites ( Figure 3C,D) . Based on the homology 3D crystal structure model, no specific evolutionary pattern was found in the MERS-CoV S gene. However, we found that the codon 1020 residue could differentiate between clade A and B ( Figure 3C,E) . Clade A strains have a glutamine at codon 1020 (Q1020), while clade B strains have an arginine residue (R1020). These results taken together, with the selection pressure analysis, suggest that codon 1020 might have a role during the evolutionary process of MERS-CoV S gene and the diversity of lineages in clade B. After translation, the 180-kDa oligomeric S gene of the coronaviruses is cleaved into an S1 receptor-binding unit and an S2 membrane fusion unit [44] . S2 subunit contains an internal fusion peptide and two heptad regions (HR1 and HR2) ( Figure 3D ). HR2 is present at the membrane anchor, while HR1 is 170 amino acids upstream. The structure of the HR region of the MERS-CoV has been solved through its side chain [39] ; we observed glutamine residue replacing arginine (observed in human-derived viruses) at 1020 position ( Figure 3E ). It might have led to the loss of side-chain interactions with M1266 due to another substitution at position 1267 in the HR2 region. That might also affect the hydrogen bond with the HR1 heptad and its stability. After translation, the 180-kDa oligomeric S gene of the coronaviruses is cleaved into an S1 receptor-binding unit and an S2 membrane fusion unit [44] . S2 subunit contains an internal fusion peptide and two heptad regions (HR1 and HR2) ( Figure 3D ). HR2 is present at the membrane anchor, while HR1 is 170 amino acids upstream. The structure of the HR region of the MERS-CoV has been solved through its side chain [39] ; we observed glutamine residue replacing arginine (observed in human-derived viruses) at 1020 position ( Figure 3E ). It might have led to the loss of side-chain , and the lower box shows the arginine residue (R1020). Hydrogens were removed for a better view. The figures were produced using PyMOL. A total of 13 patients' throat swab and nasal swab specimens were analyzed in this study. The median incubation period was 12 days (range 0 to 32). Six patients died, out of which three patients were admitted to the hospital after almost three weeks of disease onset in 2015. The remaining fatal cases were admitted to the hospital during the first week of disease onset with highly significant (p < 0.001) viral loads (nasal swabs and tracheal aspirate specimens) in 2017 (Figure 4) . The highest viral loads, in terms of MERS-CoV RNA copies detected by rRT-PCR in respiratory specimens, were observed during the first week of disease onset. The median value was 6.91 log 10 copies per milliliter among the patients that died, and 5.25 log 10 copies per milliliter among the patients that recovered (p = 0.05). The peak in viral load of tracheal aspirates was higher and occurred earlier in patients that died than recovered patients (Figure 4) . RNA concentration exceeded 10 7 copies per milliliter in samples from patients that died except the other three patients in the same group. On the other hand, RNA concentration did not exceed 10 4 copies per milliliter in samples from patients that recovered (p < 0.001). A total of 13 patients' throat swab and nasal swab specimens were analyzed in this study. The median incubation period was 12 days (range 0 to 32). Six patients died, out of which three patients were admitted to the hospital after almost three weeks of disease onset in 2015. The remaining fatal cases were admitted to the hospital during the first week of disease onset with highly significant (p < 0.001) viral loads (nasal swabs and tracheal aspirate specimens) in 2017 (Figure 4) . The highest viral loads, in terms of MERS-CoV RNA copies detected by rRT-PCR in respiratory specimens, were observed during the first week of disease onset. The median value was 6.91 log10 copies per milliliter among the patients that died, and 5.25 log10 copies per milliliter among the patients that recovered (p = 0.05). The peak in viral load of tracheal aspirates was higher and occurred earlier in patients that died than recovered patients (Figure 4) . RNA concentration exceeded 10 7 copies per milliliter in samples from patients that died except the other three patients in the same group. On the other hand, RNA concentration did not exceed 10 4 copies per milliliter in samples from patients that recovered (p < 0.001). We determined the S gene sequences from 13 patients and conducted the phylogenetic analysis to determine the genetic relationship and heterogeneity between 13 Saudi and 147 reference MERS-CoV strains from humans. The evolution of clade A and clade B MERS-CoV based on S gene sequences was considerably different from each other. Historically clade B MERS-CoV has outnumbered clade A MERS-CoV strains (Figure 1 ). This might be due to the transmission advantage of We determined the S gene sequences from 13 patients and conducted the phylogenetic analysis to determine the genetic relationship and heterogeneity between 13 Saudi and 147 reference MERS-CoV strains from humans. The evolution of clade A and clade B MERS-CoV based on S gene sequences was considerably different from each other. Historically clade B MERS-CoV has out-numbered clade A MERS-CoV strains (Figure 1 ). This might be due to the transmission advantage of clade B MERS-CoV or sampling bias. The presence of strains from a single country like Saudi Arabia or UAE in various lineages indicates the sporadic transmission from dromedaries to humans with fatal outcome in some cases. Lineage B5 divergence and separate evolution from other clade B lineages might have caused the number of mutations in the S gene compared to lineage A2. This was shown by the deep branching pattern of lineage B5 in the phylogenetic tree, as well as a change of amino acids at different positions of the S1 and S2 subunit of the S gene ( Table 2) . The residue at 1020 is known to distinguish between clade A and B strains [39] , and it was positively selected among all the strains from our study, suggesting a role in the divergence of different lineages during the evolution of the S gene. This positive selection of codon 1020 suggests that the evolution of the Saudi strains was a genetic selection, and the virus was not imported, rather it continuously evolved in the population. However, further studies are required to determine the role of this mutation in the survival of lineage B strains over lineage A. The recombination analysis suggested one potential recombination event involving lineage 5. Absence of additional recombination events meant that double infection and super-infection might not have been present during the transmission of MERS-CoV. We could not detect any potential large recombination fragments among lineage 2, 3, and 5, as previously known [45] . That could be resolved by continued identification and characterization of circulating MERS-CoV strains and developing a better-detailed classification system for various MERS-CoVs in the future. We detected the positive selection in the S gene of MERS-CoVs, which indicates that they probably experienced strong adaptive evolutionary pressure from the host's immune system. The phylogenetic reconstruction analyses have established previously [42] that lineage 5 MERS-CoVs evolved from a recombinant virus composed of lineage 3 and lineage 4 viruses. The recombinant lineage 5 viruses were shown to have acquired the 5 part of ORF1ab and the 3 part of the S gene from lineage 4 and the remaining genomic regions from lineage 3. In our S gene-based phylogenies, lineage 5 viruses were closely related to lineage 3 and 4 viruses from Saudi Arabia. One of the limitations of our study was the availability of complete S gene sequences only, instead of the complete genome sequences, which did not allow us to look for amino acid substitutions in other regions, especially the non-structural proteins of the ORF1ab polyprotein. The glycan-binding site on the top of MERS-CoV NTD is covered by a short helix along with the N-linked glycan. Unlike BCoV and HKU1, this conformational change in NTD might make it unable to attach the cell surface by recognizing certain sugar molecules [46, 47] . The S1 subunit of MERS-CoV also contains two subdomains (I and II) along with NTD and RBD domains. These subdomains appear to support the NTD and RBD domains ( Figure 3B ). The S2 subunit forms the stem region of the S gene and is mainly composed of α-helices ( Figure 3B ). The S2 cleavage site is connected to the long upstream helix by a long linker region that exposes it to the peripheral side, and thus, becomes a target for endo-lysosomal proteases ( Figure 3B ). The helical fusion peptide is also exposed beneath the S2 cleavage site and provides a connection between the HR1 region involving three consecutive α-helices. A long central helix follows along the three-fold axis towards the viral membrane from the HR1 region ( Figure 3C ). The 3-D structure of the MERS-CoV S gene is very similar to influenza virus hemagglutinin (HA) protein, which also has two subunits (HA1 and HA2) [48] . Similarly, the HA1 subunit separates from HA2 before membrane fusion starts under a low pH environment in the endosome [49] . We found amino acid substitutions in the NTD region, which is one of the major functional sites of the S gene [50] . In addition, the strains involved in the present study had nine amino acid substitutions in the S1 and S2 subunits of S gene. The outbreak in South Korea in 2015 had 12 nt substitutions (NTD, 1 site; RBD, 2 or 3 sites), implying that the strains involved diverged from a different virus [51] . MERS-CoV strains previously associated with outbreaks in Korea and other neighboring countries were most closely related (99.9% nucleotide identity) to a human MERS-CoV strain isolated in Riyadh, Saudi Arabia, in 2017. Furthermore, these strains had five nucleotide substitutions (C309T, T519C, C2928T, T3375C, and T3598C) compared with a MERS-CoV strain found in Riyadh in 2017 [50] . Coronaviruses are known to undergo recombination [52] , and it is considered to be related to an increase in pathogenicity in other animal RNA viruses [53] . There is, however, little evidence that recombination in MERS-CoV has caused substantial changes in human-to-human transmission. At the same time, recombinant MERS-CoVs have been circulating in Saudi Arabia in camels [42] and humans since 2015 [21, 54] . However, these circulating variants have not caused any major change in human CoV epidemiology [21] . There has also been a report about the circulation of deletion variants of MERS-CoV among humans in Jordan [55] , again without any major changes in epidemiology [56] . We observed that the viral loads were higher in the fatal cases (Pt.11, Pt.12, and Pt.13) and the patients that survived (Pt.6 and Pt.9) had more prolonged viral shedding in respiratory secretions (throat swabs). Previous studies involving blood viral RNA had low detection rates in the early phase of infection in patients with confirmed diagnosis [57, 58] . These findings, however, are similar to studies involving the MERS-CoV outbreak in Saudi Arabia [59] , Korea [60] , and severe acute respiratory syndrome (SARS) [61] . In summary, we obtained genetic information about the circulating MERS-CoV strains in Saudi Arabia from 2015 to 2017, belonging to lineage 5. Codon 1020 was positively selected suggesting that this residue might have played a role in the evolution of the S gene during the divergence of different lineages. The MERS-CoV RNA concentration were highest during the first week of illness and lower respiratory tract specimens had higher levels than upper respiratory tract specimens. We hope that our genetic and viral loads data would support further epidemiological investigations of MERS-CoV. It will also add to our current understanding of the evolutionary process of the S gene of MERS-CoV and a need for better surveillance in humans and dromedary camels. The presence of amino acid changes at the N-terminal domain and the structural mapping of residues under positive selection at heptad repeat 1 provides better insight and might have a potential role in virus-host tropism and pathogenesis. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia Middle East respiratory syndrome coronavirus (MERS-CoV): Announcement of the Coronavirus Study Group Hospital-associated outbreak of middle East respiratory syndrome coronavirus: A serologic, epidemiologic, and clinical description Update and Clarification on Recent MERS Cases Reported by the Kingdom of Saudi Arabia Middle East Respiratory Syndrome Coronavirus (MERS-CoV): Transmission Dynamics and Prospects for Vaccine Development Fields Virology Genomic characterization of a newly discovered coronavirus associated with acute respiratory distress syndrome in humans Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC Molecular basis of binding between novel human coronavirus MERS-CoV and its receptor CD26 Structure of MERS-CoV spike receptor-binding domain complexed with human receptor DPP4 Structure-based discovery of Middle East respiratory syndrome coronavirus fusion inhibitor An observational, laboratory-based study of outbreaks of middle East respiratory syndrome coronavirus in Jeddah and Riyadh Hospital outbreak of Middle East respiratory syndrome coronavirus Family cluster of Middle East respiratory syndrome coronavirus infections A family cluster of Middle East Respiratory Syndrome Coronavirus infections related to a likely unrecognized asymptomatic or mild case Molecular epidemiology of human coronavirus OC43 reveals evolution of different genotypes over time and recent emergence of a novel genotype due to natural recombination Isolation and characterization of a novel Betacoronavirus subgroup A coronavirus, rabbit coronavirus HKU14, from domestic rabbits Origin and Possible Genetic Recombination of the Middle East Respiratory Syndrome Coronavirus from the First Imported Case in China: Phylogenetics and Coalescence Analysis Transmission and evolution of the Middle East respiratory syndrome coronavirus in Saudi Arabia: A descriptive genomic study Infectious Middle East Respiratory Syndrome Coronavirus Excretion and Serotype Variability Based on Live Virus Isolates from Patients in Saudi Arabia Epidemiology of a Novel Recombinant Middle East Respiratory Syndrome Coronavirus in Humans in Saudi Arabia The first case of the 2015 Korean Middle East Respiratory Syndrome outbreak Clinical features and viral diagnosis of two cases of infection with Middle East Respiratory Syndrome coronavirus: A report of nosocomial transmission Isolation of MERS coronavirus from a dromedary camel Spread of MERS to South Korea and China Epidemiological investigation of MERS-CoV spread in a single hospital in South Korea Transmission of Middle East Respiratory Syndrome Coronavirus Infections in Healthcare Settings Transmission of MERS-coronavirus in household contacts Detection of a novel human coronavirus by real-time reverse-transcription polymerase chain reaction Assays for laboratory confirmation of novel human coronavirus (hCoV-EMC) infections Identification of a novel coronavirus in patients with severe acute respiratory syndrome Molecular Evolutionary Genetics Analysis across computing platforms Human Coronavirus HKU1 Spike Protein Uses O-Acetylated sialic acid as an attachment receptor determinant and employs hemagglutinin-esterase protein as a receptor-destroying enzyme 2010: A suite of phylogenetic analysis tools for evolutionary biology The effect of recombination on the reconstruction of ancestral sequences Genetic diversity of circulating Saffold viruses in Pakistan and Afghanistan Prediction of N-glycosylation Sites in Human Proteins Molecular Evolution of MERS Coronavirus: Dromedaries as a Recent Intermediate Host or Long-Time Animal Reservoir? Rooting the phylogenetic tree of Middle East respiratory syndrome coronavirus by characterization of a conspecific virus from an African bat Laboratory investigation and phylogenetic analysis of an imported Middle East respiratory syndrome coronavirus case in Greece Co-circulation of three camel coronavirus species and recombination of MERS-CoVs in Saudi Arabia The population genetics of dN/dS Evidence for a coiled-coil structure in the spike proteins of coronaviruses Evolutionary dynamics of MERS-CoV: Potential recombination, positive selection and transmission Antigenic Drift of Hemagglutinin and Neuraminidase in seasonal H1N1 influenza Viruses from Saudi Arabia in 2014-2015 Variations in spike glycoprotein gene of MERS-CoV Imported case of Middle East respiratory syndrome coronavirus (MERS-CoV) infection from Oman to Thailand Recombination, reservoirs, and the modular spike: Mechanisms of coronavirus cross-species transmission Importation and recombination are responsible for the latest emergence of highly pathogenic PRRSV in China Increase in Middle East respiratory syndrome-coronavirus cases in Saudi Arabia linked to hospital outbreak with continued circulation of recombinant virus Deletion variants of Middle East respiratory syndrome coronavirus from humans Multihospital outbreak of a Middle East respiratory syndrome coronavirus deletion variant, Jordan: A molecular, serologic, and epidemiologic investigation The S protein of bovine coronavirus is a hemagglutinin recognizing 9-O-acetylated sialic acid as a receptor determinant Viral RNA in Blood as Indicator of Severe Outcome in Middle East Respiratory Syndrome Coronavirus Infection Viral Shedding and Antibody Response in 37 Patients With Middle East Respiratory Syndrome Coronavirus Infection Viral membrane fusion RDP4: Detection and analysis of recombination patterns in virus genomes MERS-CoV infection is associated with downregulation of genes encoding Th1 and Th2 cytokines/chemokines and elevated inflammatory innate immune response in the lower respiratory tract Viral Load Kinetics of MERS Coronavirus Infection Detection of SARS coronavirus Supplementary Materials: Supplementary materials can be found at http://www.mdpi.com/1999-4915/12/5/502/s1. Table S1 Geographical and temporal distribution of 160 MERS-CoV isolates used for phylogenetic analysis. The authors thank the Research Center at King Fahad Medical City for funding this study (Grant No 017-019). We also thank the European Virus Archives (EVA) for providing the assay controls. The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Middle