ArticlePDF Available

Abstract and Figures

A prospective study of a dromedary camel herd during the 2013-14 calving season showed Middle East respiratory syndrome coronavirus infection of calves and adults. Virus was isolated from the nose and feces but more frequently from the nose. Preexisting neutralizing antibody did not appear to protect against infection.
No caption available
… 
Content may be subject to copyright.
EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.20,No.7,July2014 1231
MERS Coronavirus
in Dromedary
Camel Herd,
Saudi Arabia
Maged G. Hemida,1 Daniel K.W. Chu,1
Leo L.M. Poon, Ranawaka A.P.M. Perera,
Mohammad A. Alhammadi, Hoi-yee Ng,
Lewis Y. Siu, Yi Guan, Abdelmohsen Alnaeem,
and Malik Peiris
A prospective study of a dromedary camel herd during
the2013–14calving seasonshowedMiddle East respira-
tory syndrome coronavirus infection of calves and adults.
Viruswasisolatedfrom the nose andfeces but more fre-
quentlyfromthenose.Preexistingneutralizingantibodydid
not appear to protect against infection.
Ongoing transmission of Middle East respiratory syn-
drome coronavirus (MERS-CoV) to humans under-
scores the need to understand the animal sources of zoonot-
ic infection (1,2). MERS-CoV RNA has been detected in
dromedary camels (3,4), and dromedary infection precedes
human infection (5). We conducted a prospective study in
dromedary herds in Al-Hasa, Saudi Arabia, through the
peak calving season (December 2013–February 2014) to
document virologic features of MERS-CoV infection in
these animals.
The Study
We studied dromedaries at 2 farms in Al-Hasa, 4–5
km apart. Farm A had 70 animals; 4 were 1 month of
age, 8 were ≈1 year of age, and the rest were adults (>2
years of age). The herd did not go to pasture in the des-
ert (“zero-grazing”; type of grazing may inuence types
of potential exposures). The animals were sampled on 5
occasions during November 2013–February 2014. Farm
B (“semi–zero-grazing”) had 17 adults and 3 calves; its
herd was sampled in February 2014. Nasal, oral, or rectal
swab samples and blood samples were collected (Table 1;
online Technical Appendix Table, http://wwwnc.cdc.gov/
EID/article/20/7/14-0571-Techapp1.pdf). Swab and serum
samples were stored frozen at 80°C until testing.
Hydrolysis probe–based real-time reverse transcrip-
tion PCR (RT-PCR) targeting MERS-CoV upstream of
E (UpE) and open reading frame (ORF) 1a genes and a
broad-range RT-PCR reacting across the CoV family to
detect other CoVs were used as described (4). Specimens
initially positive for MERS-CoV were re-extracted and re-
tested to conrm the positive results.
The full genome of MERS-CoV was obtained directly
from the clinical specimens with 3–4 times coverage by se-
quencing PCR amplicons with overlapping sequence reads
and sequence assembly (4). Dromedary MERS-CoV full
genomes obtained in this study (GenBank accession nos.
KJ650295–KJ650297) were aligned with human MERS-
CoV genomes retrieved from GenBank. We constructed
full-genome phylogenies using MEGA5 with neighbor-
joining and bootstrap resampling of 500 replicates (6).
Virus isolation was attempted in Vero E6 cells. We tested
serum samples for neutralizing antibody titers using a vali-
dated MERS-CoV spike pseudoparticle neutralization test
(7) (online Technical Appendix).
At farm A, we detected MERS-CoV in 1 of 4 drom-
edaries sampled on November 30, none of 11 sampled on
December 4, nine of 11 sampled on December 30, and none
of 9 sampled on February 14 (Table 1). Of the 10 drom-
edaries that tested positive for MERS-CoV, 9 had parallel
nasal and fecal specimens tested, with virus detected in the
nasal swab specimens from 8 and the fecal specimen from
1. At the December 30 sampling, 7 of 8 calves and 2 of 3
adults tested positive for MERS-CoV, indicating that when
MERS-CoV circulates on a farm, both calves and adults
can be infected (online Technical Appendix Table). Be-
cause all 12 adults with serum collected before December
30 were seropositive (titers >320), it is likely, though not
certain, that the MERS-CoV infections in the 2 adults (nos.
21, 19Dam) sampled on December 30 were reinfections,
as has been reported for other CoVs (8). The seronegative
1-year-old calves, nos. 13 and 14, had the highest nasal vi-
ral loads (UpE assay 1.3 × 108 to 1.78 × 108/mL specimen),
and a 2-week-old calf, no. 22, with (presumably passively
acquired) titers of 1,280 became infected but had a much
lower viral load. Overall, these data suggest that prior in-
fection or passively acquired maternal antibody might not
provide complete protection from infection (online Techni-
cal Appendix Table).
Four MERS-CoV–positive calves had mild respiratory
signs (cough, sneezing, respiratory discharge), abnormally
elevated body temperature, and loss of appetite at the De-
cember 30 sampling, which resolved over a few days. Three
calves from which paired serum samples were available
(Table 2; nos. 13, 15, 17) demonstrated >4-fold rising anti-
body titers to MERS-CoV. Calf no. 13 (1 year of age) had
Author afliations: King Faisal University,Al Hofuf, Saudi Arabia
(M.G. Hemida, M.A. Alhammadi, A. Alnaeem); Kafrelsheikh
University,KafrElsheikh,Egypt(M.G.Hemida);andTheUniversity
of Hong Kong, Hong Kong, China (D.K.W. Chu, L.L.M. Poon,
R.A.P.M.Perera,H.-y.Ng,L.Y.Siu,Y.Guan,M.Peiris)
DOI:http://dx.doi.org/10.3201/eid2007.140571 1These authors contributed equally to this article.
DISPATCHES
1232 EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.20,No.7,July2014
a high viral load and was seronegative at the rst MERS-
CoV–positive result (indicating that it had been recently
infected) but was MERS-CoV RNA negative 6 weeks
later, suggesting that virus shedding is not prolonged. We
did not detect virus RNA by RT-PCR in the 3 acute-phase
serum samples from infected dromedaries (nos. 1, 16, 17),
suggesting that acute infection is not associated with pro-
longed viremia. Dromedaries from farm B were sampled
once on February 11; all results were negative.
The full genomes of MERS-CoV sequenced directly
from a nasal swab specimen collected on November 30
were identical to those from a nasal swab specimen and a
fecal specimen collected on December 30. In addition, the
complete spike gene was sequenced from 4 other MERS-
CoV–positive nasal swab specimens, and these spike genes
were genetically identical.
Virus isolation in Vero E6 cells was attempted from
7 positive nasal swab and fecal specimens that had >106
copies/mL in the original sample in the UpE RT-PCR. Vi-
ruses were isolated from 2 nasal swab (nos. 13, 14) and
1 fecal swab (no. 19Dam) specimens collected on De-
cember 30; these were the specimens with high numbers
of MERS-CoV copies (9.27 × 107 to 1.78 × 108 copies/
mL). The full-genome sequence of 1 virus culture isolate
was obtained in parallel with that of the original virus in
the original clinical specimen. We observed 3 nucleotide
changes in ORF1b, spike, and membrane protein genes
in the isolates after 2 passages in Vero E6 cells, of which
2 were nonsynonymous, leading to changes in spike
(S1251F) and membrane proteins (T8I). This nding
highlights the importance of sequencing the viral genome
directly from clinical specimens.
MERS-CoVs circulating in dromedaries on farm A
during a 1-month period were genetically identical over the
full 30,100-nt genome in 3 viruses and the spike protein of
4 more viruses, giving a mutation rate of 0 nt substitutions
per site per day (95% credible interval 0 to 2.7 × 106). The
estimated mutation rate for epidemiologically unlinked hu-
man MERS-CoV was 3.1 × 106 (95% CI 2.4 × 106 to 3.8
× 106) (9).
Conclusions
The unusual genetic stability of MERS-CoV in
dromedaries, taken together with its high seroprevalence
(7,10–13), raises the hypothesis that dromedaries might be
the natural host for this virus. Further longitudinal stud-
ies of MERS-CoVs in dromedaries are needed to conrm
this hypothesis.
Genome organization of the dromedary MERS-CoV
detected in this study was identical to that of the virus in
humans. The virus strains clustered phylogenetically with-
in clade B (9) and were most closely related to the strain
MERS-CoV_FRA/UAE and to MERS-CoV detected in
Buraidah (Saudi Arabia) and Al-Hasa (Figure). The farm
is ≈300 km from United Arab Emirates and 600 km from
Buraidah. Dromedaries move between Al-Hasa and Burai-
dah and, more limitedly, between Al-Hasa and United
Arab Emirates.
The full-genome sequence of MERS-CoV from drom-
edaries in this study is 99.9% similar to genomes of hu-
man clade B MERS-CoV. The spike gene is the major
determinant for virus host specicity. In comparison with
other publically available human MERS-CoV sequences,
we found 6-nt mutations in the spike gene unique to these
dromedary viruses. Of these, 3 (S457G, L773F, and V810I)
were nonsynonymous. These amino acid changes are lo-
cated outside the binding interface between MERS-CoV
spike protein and human DPP4 receptor, suggesting these
amino acid differences are unlikely to affect receptor bind-
ing. Thus, these dromedary viruses may retain capacity
to infect humans, as Chu et al. suggested for dromedary
MERS-CoV in Egypt (4).
MERS-CoV may be isolated from nasal swab speci-
mans and feces, indicating that both could be possible sourc-
es of virus transmission to humans and other animals, but
virus detection rates were higher in nasal swab specimens.
Table 1. RT-PCRofdromedarycamelsamplesforMERS-CoV,
Al-Hasa, Saudi Arabia*
Farm,samplingdate
Age†/no.
sampled
No.specimens
positive/no.tested
Nasal Oral Fecal
FarmA
2013Nov30
Calf, 0
ND
ND
ND
Adult, 4 1/1 0/2 0/4
2013Dec4
Calf, 9
ND
0/9
0/7
Adult,2
ND
0/2
0/2
2013Dec30
Calf,8
7/8
0/1
0/6
Adult, 3
1/3
0
1/3
2014Feb14 Calf,7 0/7 ND 0/7
Adult,2
0/2
ND
0/2
FarmB:2014Feb11 Calf, 3 0/3 ND 0/3
Adult, 3
0/3
ND
0/3
*Data on individual d romedaries are providedinonlineTechnicalAppendix
Table, http://wwwnc.cdc.gov/EID/article/20/7/14-0571-Techapp1.pdf. RT-
PCR,reversetranscriptionPCR;MERS-CoV,MiddleEastrespiratory
syndromecoronavirus;ND,notdone.
Adults are 614yofage;calvesare40dto2yofage.
Two different dromedaries were positive in nasal and fecal swabs.
Table2.LongitudinalsamplingofMERS-CoVpositive dromedary
camel calves on farm A, Al-Hasa, Saudi Arabia*
Sample collection
date
Sex/age
RT-PCR
result
Titer
13 2013Dec30 F/1y Positive <20
2014Feb14
F/1y
Negative
640
2013Dec30
F/1y
Positive
20
2014Feb14 F/1y Negative 160
2013Dec30
F/40d
Positive
80
2014Feb14 F/3mo Negative 1,280
2013Dec30
F/1y
Positive
NA
2014Feb14
F/1y
Negative
320
*MERS-CoV,MiddleEastrespiratorysyndromecoronavirus;RT-PCR,
reverse transcription PCR.
EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.20,No.7,July2014 1233
MERS-CoVinCamelHerd
Our preliminary data suggest that preexisting MERS-CoV
antibody might not completely protect against re-infection;
however, this question needs more investigation.
We thank the King Faisal University Deanship of Scientif-
ic Research for their support (grant no. 143011). This research
was funded by a research contract from the National Insti-
tute of Allergy and Infectious Diseases, National Institutes of
Health (contract no. HHSN266200700005C), and a grant
from the European Community Seventh Framework Program
(FP7/2007-2013) under project European Management Platform
for Emerging and Re-emerging Disease entities (grant agreement
no. 223498) (EMPERIE).
Dr Hemida is an assistant professor of molecular virology at
King Faisal University, Saudi Arabia. His primary research inter-
ests are virus–host interactions and the molecular biology of CoVs.
References
1. World Health Organization. Middle East respiratory syndrome
coronavirus (MERSCoV). Summary and literature update—as of
27 March 2014 [cited 2014 Apr 11]. http://www.who.int/csr/disease/
coronavirus_infections/MERS_CoV_Update_27_March_2014.
pdf?ua=1
2. The WHO MERS-CoV Research Group. State of knowledge and
data gaps of Middle East respiratory syndrome coronavirus (MERS-
CoV) in humans. PLoS Curr. 2013;5:pii:ecurrents.outbreaks.0bf719
e352e7478f8ad85fa30127ddb8.
3. Alagaili AN, Briese T, Mishra N, Kapoor V, Sameroff SC, de Wit E,
et al. Middle East respiratory syndrome coronavirus infection in
dromedary camels in Saudi Arabia. MBio. 2014;5:e00884-14.
http://dx.doi.org/10.1128/mBio.01002-14
4. Chu DKW, Poon LLM, Gomaa MM, Shehata MM, Perera RA,
Zeid DA, et al. MERS coronaviruses in dromedary camels, Egypt.
[Internet]. Emerg Infect Dis. 2014 Jun [cited 2014 Mar 31].
5. Memish ZA, Cotton M, Meyer B, Watson SJ, Alsaha AJ,
Al Rabeeah AA, et al. Human infection with MERS corona-
virus after exposure to infected camels, Saudi Arabia, 2013.
[Internet]. Emerg Infect Dis. 2014. http://dx.doi.org/10.3201/
eid2006.140402
6. Tamura K, Peterson D, Peterson N, Stecher G, Nei M, Kumar S.
MEGA5: molecular eevolutionary genetics analysis using maxi-
mum likelihood, evolutionary distance, and maximum parsimony
methods. Mol Biol Evol. 2011;28:2731–9. http://dx.doi.org/10.1093/
molbev/msr121
7. Perera RA, Wang P, Gomaa MR, El-Shesheny R, Kandeil A,
Bagato O, et al. Seroepidemiology for MERS coronavirus using
microneutralisation and pseudoparticle virus neutralisation assays
reveal a high prevalence of antibody in dromedary camels in Egypt,
June 2013. Euro Surveill. 2013;18:20574.
8. Addie DD, Dennis JM, Toth S, Callanan JJ, Reid S, Jarrett O.
Long-term impact on a closed household of pet cats of natural
infection with feline coronavirus, feline leukaemia virus and feline
immunodeciency virus. Vet Rec. 2000;146:419–24. http://dx.doi.
org/10.1136/vr.146.15.419
9. Cotten M, Watson SJ, Zumla AI, Makhdoom HQ, Palser AL,
Ong SH, et al. Spread, circulation, and evolution of the Middle
East respiratory syndrome coronavirus. MBio. 2014;5:e01062-13.
http://dx.doi.org/10.1128/mBio.01062-13
10. Reusken CB, Haagmans BL, Müller MA, Gutierrez C, Godeke GJ,
Meyer B, et al. Middle East respiratory syndrome coronavirus
Figure. Phylogenetic tree of
Middle East respiratory syndrome
coronavirus (MERS-CoV) full
genomes (29,901 nt after trimming
the ends) or near–full genomes
from humans and dromedary
camels. The tree was constructed
by using neighbor-joining methods
with bootstrap resampling of 500
replicates. The most divergent
MERS-CoV,EgyptNRCE-HKU205,
was used as outgroup. Bold
type indicates camel MERS-CoV
genomes from this study. GenBank
accession numbers of genome
sequences included in this study are
KJ477102, KF600652, KF600630,
KF600651, KF186567, KF600627,
KF186564, KF600634, KF600632,
KF600644, KF600647, KF600645,
KF186565, KF186566, KF745068,
KF600620, KF600612, KC667074,
KC164505, KF192507, KF600613,
KF600628, KF961222, KF961221,
KC776174, and JX869059.
Scale bar indicates nucleotide
substitutions per site.
DISPATCHES
1234 EmergingInfectiousDiseases•www.cdc.gov/eid•Vol.20,No.7,July2014
neutralising serum antibodies in dromedary camels: a comparative
serological study. Lancet Infect Dis. 2013;13:859–66. http://dx.doi.
org/10.1016/S1473-3099(13)70164-6
11. Reusken CB, Ababneh M, Raj VS, Meyer B, Eljarah A,
Abutarbush S, et al. Middle East respiratory syndrome coronavirus
(MERS-CoV) serology in major livestock species in an
affected region in Jordan, June to September 2013. Euro Surveill.
2013;18:20662.
12. Hemida MG, Perera RA, Wang P, Alhammadi MA, Siu LY, Li M,
et al. Middle East respiratory syndrome (MERS) coronavirus
seroprevalence in domestic livestock in Saudi Arabia, 2010 to
2013. Euro Surveill. 2013;18:20659.
13. Meyer B, Müller MA, Corman VM, Reusken CB, Ritz D,
Godeke GJ, et al. Antibodies against MERS coronavirus in drom-
edary camels, United Arab Emirates, 2003 and 2013. Emerg Infect
Dis. 2014;20:552–9. http://dx.doi.org/10.3201/eid2004.131746
Address for correspondence; Malik Peiris, School of Public Health,
The University of Hong Kong, 21 Sassoon Rd, Pokfulam, Hong
Kong Special Administrative Region; email: malik@hku.hk; or
Abdelmohsen Alnaeem, Department of Clinical Studies, College of
Veterinary Medicine, King Faisal University, Saudi Arabia; email:
aaalnaeem@kfu.edu.sa
... [1,2,3]. Camel diseases are not only of veterinary interest but also of public health concern [4,5]. Tapeworms of the genus Echinococcus are the cause of the zoonotic disease known as cystic echinococcosis (CE). ...
Article
Full-text available
TAPEWORMS of the genus Echinococcus are the cause of the zoonotic disease known as cystic echinococcosis (CE). This work aimed to detect the prevalence, the fertility of cysts, the viability of protoscolices, as well as the molecular and phylogenetic analysis of CE infecting one-humped camels (Camelus dromedarius) in Beheria, Egypt. Between August 2022 and July 2023, 156 camels at the Kom Hamada abattoir were checked for the presence of hydatid cysts during routine meat inspection. Using phylogenetic analysis of the Cox1 and Nad1 sequences, the cysts were molecularly identified. The overall prevalence in slaughtered animals was 16.67%. The prevalence in the liver and lungs was 3.85% and 96.15%, respectively. Spring had the highest prevalence (28.11%). The fertility rate was 72.22%. The viability rate was 87.5% from fertile lung cysts. Phylogenetic analysis confirmed the existence of Echinococcus canadensis (E. granulosus G6). The study area had a high prevalence of CE in one-humped camels in the study area, which requires the implementation of an active control program. Moreover, our data confirms that the genotype involved is E. canadensis. Further studies on CE in camels and other ruminants in Egypt should be carried out to illustrate factors associated with this disease.
... MERS-CoV infection has resulted in approximately 2700 fatalities and sustained low-level local transmission in Saudi Arabia (WHO, 2012). Relatives of MERS-CoV have been found in several animal species, including bats, camels, hedgehogs and pangolins (Chen et al., 2023;Chu et al., 2014;Hemida et al., 2014;Lau et al., 2019;Luo et al., 2018;Mols et al., 2023;Shishido & Letizia, 2015;Speranskaya et al., 2023). Dromedary camels have been well-documented as intermediate hosts of MERS-CoV, and subsequent research has uncovered many relatives of MERS-CoV in bats, supporting the hypothesis that the origins and evolution of MERS-CoV trace back to bats (Anthony et al., 2017;Chu et al., 2014;Cui et al., 2019;Lau et al., 2013;Memish et al., 2013). ...
Article
Full-text available
Angiotensin-converting enzyme 2 (ACE2) was recognized as an entry receptor shared by coronaviruses from Sarbecovirus and Setracovirus subgenera, including three human coronaviruses: SARS-CoV, SARS-CoV-2, and NL63. We recently disclosed that NeoCoV and three other merbecoviruses (PDF-2180, MOW15-22, PnNL 2018B), which are MERS-CoV relatives found in African and European bats, also utilize ACE2 as their functional receptors through unique receptor binding mechanisms. This unexpected receptor usage assumes significance, particularly in light of the prior recognition of Dipeptidyl peptidase-4 (DPP4) as the only known protein receptor for merbecoviruses. In contrast to other ACE2-using coronaviruses, NeoCoV and PDF-2180 engage a distinct and relatively compact binding surface on ACE2, facilitated by protein-glycan interactions, which is demonstrated by the Cryo-EM structures of the receptor binding domains (RBDs) of these viruses in complex with a bat ACE2 orthologue. These findings further support the hypothesis that phylogenetically distant coronaviruses, characterized by distinct RBD structures, can independently evolve to acquire ACE2 affinity during inter-species transmission and adaptive evolution. To date, these viruses have exhibited limited efficiency in entering human cells, although single mutations like T510F in NeoCoV can overcome the incompatibility with human ACE2. In this review, we present a comprehensive overview of ACE2-using merbecoviruses, summarize our current knowledge regarding receptor usage and host tropism determination, and deliberate on potential strategies for prevention and intervention, with the goal of mitigating potential future outbreaks caused by spillover of these viruses.
... Further testing for the presence of IDV involved archival camel sera, which were originally tested for various pathogens [86][87][88][89][90]. In these studies, very high IDV seroprevalence was observed: 98% in Mongolia (n = 100), 100% in Saudi Arabia (n = 76), 100% in Australia (n = 23), and 100% in Nigeria (n = 100). ...
Article
Full-text available
Influenza D virus (IDV) infections have been observed in animals worldwide, confirmed through both serological and molecular tests, as well as virus isolation. IDV possesses unique properties that distinguish it from other influenza viruses, primarily attributed to the hemagglutinin-esterase fusion (HEF) surface glycoprotein, which determines the virus’ tropism and wide host range. Cattle are postulated to be the reservoir of IDV, and the virus is identified as one of the causative agents of bovine respiratory disease (BRD) syndrome. Animals associated with humans and susceptible to IDV infection include camels, pigs, small ruminants, and horses. Notably, high seroprevalence towards IDV, apart from cattle, is also observed in camels, potentially constituting a reservoir of the virus. Among wild and captive animals, IDV infections have been confirmed in feral pigs, wild boars, deer, hedgehogs, giraffes, wildebeests, kangaroos, wallabies, and llamas. The transmission potential and host range of IDV may contribute to future viral differentiation. It has been confirmed that influenza D may pose a threat to humans as a zoonosis, with seroprevalence noted in people with professional contact with cattle.
... Notably, this study detected CoVNJ99 and CoVNJ142 in E. cachinus; CoVNJ99 and CoVNJ142 are associated with CoVs carried by dromedary camel and bovines, which are intermediate hosts for the Middle East respiratory syndrome coronavirus. MERS-CoV is repeatedly detected in dromedary camels, and MERS-CoV isolated from dromedary camels is genetically and phenotypically similar to the CoVs that infect humans, including the virus spike protein, suggesting that CoVs in dromedary camels may be transmitted to humans [25]. True primary zoonotic infection is difficult to identify, and it is possible that the host or other intermediate host will be altered during the transmission of infection to humans [26]. ...
Article
Full-text available
The genetic diversity of coronaviruses (CoVs) is high, and their infection in animals has not yet been fully revealed. By RT-PCR detection of the partial RNA-dependent RNA polymerase (RdRp) gene of CoVs, we screened a total of 502 small mammals in the Dali and Nujiang prefectures of Western Yunnan Province, China. The number of overall CoV positives was 20, including β-CoV (n = 13) and α-CoV (n = 7), with a 3.98% prevalence in rectal tissue samples. The identity of the partial RdRp genes obtained for 13 strains of β-CoV was 83.42–99.23% at the nucleotide level, and it is worth noting that the two strains from Kachin red-backed voles showed high identity to BOV-36/IND/2015 from Indian bovines and DcCoV-HKU23 from dromedary camels (Camelus dromedarius) in Morocco; the nucleotide identity was between 97.86 and 98.33%. Similarly, the identity of the seven strains of α-CoV among the partial RdRp sequences was 94.00–99.18% at nucleotide levels. The viral load in different tissues was measured by quantitative RT-PCR (qRT-PCR). The average CoV viral load in small mammalian rectal tissue was 1.35 × 106 copies/g; differently, the mean CoV viral load in liver, heart, lung, spleen, and kidney tissue was from 0.97 × 103 to 3.95 × 103 copies/g, which revealed that CoV has extensive tropism in rectal tissue in small mammals (p < 0.0001). These results revealed the genetic diversity, epidemiology, and infective tropism of α-CoV and β-CoV in small mammals from Dali and Nujiang, which deepens the comprehension of the retention and infection of coronavirus in natural hosts.
Article
Full-text available
Bat coronaviruses cause a wide range of illnesses in humans and animals. Bats are known to harbor a wide diversity of Alphacoronaviruses and Betacoronaviruses. Betacoronaviruses have been linked to Severe Acute Respiratory Syndrome (SARS), Middle East Respiratory Syndrome (MERS), and other diseases such as gastroenteritis, bronchiolitis, and pneumonia. In the last 20 years, three betacoronaviruses emerged and caused widespread outbreaks in humans, including two deadly betacoronavirus epidemics, SARS-CoV, with mortality rate of 10%, and MERS-CoV, with mortality rate of 34.7%, and SARS-CoV-2, which caused the COVID-19 pandemic, with mortality rate of 3.4%. Studies have shown that bats are the main natural reservoirs for these viruses or their ancestral viruses. Observed variations in bat coronavirus genomes indicate that these viruses may have a potential to transmit to other hosts in close contact with humans and subsequently transmit to humans. As of today, there are no reported cases of direct coronavirus transmission from bats to humans. One reason for this might be that intermediate hosts are required for the transmission of bat coronaviruses to humans. Further studies are needed to map the amino acids and genomic regions responsible for the interactions between the spike of coronavirus and its receptors.
Article
Full-text available
Nidovirales is one order of RNA virus, with the largest single-stranded positive sense RNA genome enwrapped with membrane envelope. It comprises four families (Arterividae, Mesoniviridae, Roniviridae, and Coronaviridae) and has been circulating in humans and animals for almost one century, posing great threat to livestock and poultry,as well as to public health. Nidovirales shares similar life cycle: attachment to cell surface, entry, primary translation of replicases, viral RNA replication in cytoplasm, translation of viral proteins, virion assembly, budding, and release. The viral RNA synthesis is the critical step during infection, including genomic RNA (gRNA) replication and subgenomic mRNAs (sg mRNAs) transcription. gRNA replication requires the synthesis of a negative sense full-length RNA intermediate, while the sg mRNAs transcription involves the synthesis of a nested set of negative sense subgenomic intermediates by a discontinuous strategy. This RNA synthesis process is mediated by the viral replication/transcription complex (RTC), which consists of several enzymatic replicases derived from the polyprotein 1a and polyprotein 1ab and several cellular proteins. These replicases and host factors represent the optimal potential therapeutic targets. Hereby, we summarize the Nidovirales classification, associated diseases, “replication organelle,” replication and transcription mechanisms, as well as related regulatory factors.
Preprint
Full-text available
Since the emergence of SARS-CoV-2 in Wuhan in 2019 its host reservoir has not been established. Phylogenetic analysis was performed on whole genome sequences (WGS) of 71 coronaviruses and a Breda virus. A subset comprising two SARS-CoV-2 Wuhan viruses and 8 of the most closely related coronavirus sequences were used for host reservoir analysis using Bayesian Evolutionary Analysis Sampling Trees (BEAST). Within these genomes, 20 core genome fragments were combined into 2 groups each with similar clock rates (5.9x10 -3 and 1.1x10 -3 subs/site/year). Pooling the results from these fragment groups yielded a most recent common ancestor (MRCA) shared between SARS-COV-2 and the bat isolate RaTG13 around 2007 (95% HPD: 2003, 2011). Further, the host of the MRCA was most likely a bat (probability 0.64 - 0.87). Hence, the spillover into humans must have occurred at some point between 2007 and 2019 and bats may have been the most likely host reservoir.
Article
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) that was first identified in Saudi Arabia in 2012 is caused by a virus of 2C beta Corona virus lineage that expresses the dipeptidyl peptidase 4 (DPP4) receptor and is densely endemic in dromedary camels of East Africa and the Arabian Peninsula. The first case of MERS-CoV was detected from a 60-year-old man from Saudi Arabia, who died of severe respiratory distress and renal failure. The virus spread over 27 countries with high fatality rate of 35.4% making an ongoing public health threat. Extensive works performed on origin of MERS-CoV indicated that, bats are ancestral host due to their huge species variations, long time fly, and unique behavior of multispecies coexistence. Strong evidence of human-to-human transmission was obtained from epidemiological and genomic studies investigating clustering of cases in hospitals and among household contacts. Zoonotic transmission in human results clinical symptoms ranging from mild to severe respiratory symptoms. Similarly, humans can be infected through direct contact with infected camels or their milk, urine or other body fluids. For identification and characterization of the virus, diagnostic methods such as serological tests, molecular methods and neutralizing assays are used. Studies indicated that, MERS-CoV entrenched in the Arabian Peninsula has acquired increased pathogenic potential for humans due to mutation in their spike protein. Although human cases are yet not detected in Africa, if pathogenic clade B viruses from the Arabian Peninsula are introduced into Africa, they are likely to become dominant and increases public health risks. Therefore, the purpose of this review is to highlight MERS-CoV in dromedary camel and its public health importance.
Article
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) that was first identified in Saudi Arabia in 2012 is caused by a virus of 2C beta Corona virus lineage that expresses the dipeptidyl peptidase 4 (DPP4) receptor and is densely endemic in dromedary camels of East Africa and the Arabian Peninsula. The first case of MERS-CoV was detected from a 60-year-old man from Saudi Arabia, who died of severe respiratory distress and renal failure. The virus spread over 27 countries with high fatality rate of 35.4% making an ongoing public health threat. Extensive works performed on origin of MERS-CoV indicated that, bats are ancestral host due to their huge species variations, long time fly, and unique behavior of multispecies coexistence. Strong evidence of human-to-human transmission was obtained from epidemiological and genomic studies investigating clustering of cases in hospitals and among household contacts. Zoonotic transmission in human results clinical symptoms ranging from mild to severe respiratory symptoms. Similarly, humans can be infected through direct contact with infected camels or their milk, urine or other body fluids. For identification and characterization of the virus, diagnostic methods such as serological tests, molecular methods and neutralizing assays are used. Studies indicated that, MERS-CoV entrenched in the Arabian Peninsula has acquired increased pathogenic potential for humans due to mutation in their spike protein. Although human cases are yet not detected in Africa, if pathogenic clade B viruses from the Arabian Peninsula are introduced into Africa, they are likely to become dominant and increases public health risks. Therefore, the purpose of this review is to highlight MERS-CoV in dromedary camel and its public health importance.
Article
Full-text available
The Middle East respiratory syndrome (MERS) is proposed to be a zoonotic disease; however, the reservoir and mechanism for transmission of the causative agent, the MERS coronavirus, are unknown. Dromedary camels have been implicated through reports that some victims have been exposed to camels, camels in areas where the disease has emerged have antibodies to the virus, and viral sequences have been recovered from camels in association with outbreaks of the disease among humans. Nonetheless, whether camels mediate transmission to humans is unresolved. Here we provide evidence from a geographic and temporal survey of camels in the Kingdom of Saudi Arabia that MERS coronaviruses have been circulating in camels since at least 1992, are distributed countrywide, and can be phylogenetically classified into clades that correlate with outbreaks of the disease among humans. We found no evidence of infection in domestic sheep or domestic goats.
Article
Full-text available
We identified the near-full-genome sequence (29,908 nt, >99%) of Middle East respiratory syndrome coronavirus (MERS-CoV) from a nasal swab specimen from a dromedary camel in Egypt. We found that viruses genetically very similar to human MERS-CoV are infecting dromedaries beyond the Arabian Peninsula, where human MERS-CoV infections have not yet been detected.
Article
Full-text available
We investigated a case of human infection with Middle East respiratory syndrome coronavirus (MERS-CoV) after exposure to infected camels. Analysis of the whole human-derived virus and 15% of the camel-derived virus sequence yielded nucleotide polymorphism signatures suggestive of cross-species transmission. Camels may act as a direct source of human MERS-CoV infection.
Article
Full-text available
Middle East respiratory syndrome coronavirus (MERS-CoV) has caused an ongoing outbreak of severe acute respiratory tract infection in humans in the Arabian Peninsula since 2012. Dromedary camels have been implicated as possible viral reservoirs. We used serologic assays to analyze 651 dromedary camel serum samples from the United Arab Emirates; 151 of 651 samples were obtained in 2003, well before onset of the current epidemic, and 500 serum samples were obtained in 2013. Recombinant spike protein-specific immunofluorescence and virus neutralization tests enabled clear discrimination between MERS-CoV and bovine CoV infections. Most (632/651, 97.1%) camels had antibodies against MERS-CoV. This result included all 151 serum samples obtained in 2003. Most (389/651, 59.8%) serum samples had MERS-CoV-neutralizing antibody titers >1,280. Dromedary camels from the United Arab Emirates were infected at high rates with MERS-CoV or a closely related, probably conspecific, virus long before the first human MERS cases.
Article
Full-text available
The Middle East respiratory syndrome (MERS) is proposed to be a zoonotic disease; however, the reservoir and mechanism for transmission of the causative agent, the MERS coronavirus, are unknown. Dromedary camels have been implicated through reports that some victims have been exposed to camels, camels in areas where the disease has emerged have antibodies to the virus, and viral sequences have been recovered from camels in association with outbreaks of the disease among humans. Nonetheless, whether camels mediate transmission to humans is unresolved. Here we provide evidence from a geographic and temporal survey of camels in the Kingdom of Saudi Arabia that MERS coronaviruses have been circulating in camels since at least 1992, are distributed countrywide, and can be phylogenetically classified into clades that correlate with outbreaks of the disease among humans. We found no evidence of infection in domestic sheep or domestic goats. IMPORTANCE This study was undertaken to determine the historical and current prevalence of Middle East respiratory syndrome (MERS) coronavirus infection in dromedary camels and other livestock in the Kingdom of Saudi Arabia, where the index case and the majority of cases of MERS have been reported.
Article
Full-text available
Unlabelled: The Middle East respiratory syndrome coronavirus (MERS-CoV) was first documented in the Kingdom of Saudi Arabia (KSA) in 2012 and, to date, has been identified in 180 cases with 43% mortality. In this study, we have determined the MERS-CoV evolutionary rate, documented genetic variants of the virus and their distribution throughout the Arabian peninsula, and identified the genome positions under positive selection, important features for monitoring adaptation of MERS-CoV to human transmission and for identifying the source of infections. Respiratory samples from confirmed KSA MERS cases from May to September 2013 were subjected to whole-genome deep sequencing, and 32 complete or partial sequences (20 were ≥ 99% complete, 7 were 50 to 94% complete, and 5 were 27 to 50% complete) were obtained, bringing the total available MERS-CoV genomic sequences to 65. An evolutionary rate of 1.12 × 10(-3) substitutions per site per year (95% credible interval [95% CI], 8.76 × 10(-4); 1.37 × 10(-3)) was estimated, bringing the time to most recent common ancestor to March 2012 (95% CI, December 2011; June 2012). Only one MERS-CoV codon, spike 1020, located in a domain required for cell entry, is under strong positive selection. Four KSA MERS-CoV phylogenetic clades were found, with 3 clades apparently no longer contributing to current cases. The size of the population infected with MERS-CoV showed a gradual increase to June 2013, followed by a decline, possibly due to increased surveillance and infection control measures combined with a basic reproduction number (R0) for the virus that is less than 1. Importance: MERS-CoV adaptation toward higher rates of sustained human-to-human transmission appears not to have occurred yet. While MERS-CoV transmission currently appears weak, careful monitoring of changes in MERS-CoV genomes and of the MERS epidemic should be maintained. The observation of phylogenetically related MERS-CoV in geographically diverse locations must be taken into account in efforts to identify the animal source and transmission of the virus.
Article
Full-text available
In Saudi Arabia, including regions of Riyadh and Al Ahsa, pseudoparticle neutralisation (ppNT) and microneutralisation (MNT) tests detected no antibodies to Middle East Respiratory Syndrome coronavirus (MERS-CoV) in sheep (n= 100), goats (n= 45), cattle (n= 50) and chickens (n= 240). Dromedary camels however, had a high prevalence of MERS-CoV antibodies. Bovine coronavirus (BCoV) infected sera from cattle had no cross-reactivity in MERS-CoV ppNT or MNT, while many dromedary camels’ sera reacted to both BCoV and MERS-CoV. Some nevertheless displayed specific serologic reaction profiles to MERS-CoV.
Article
Full-text available
Between June and September 2013, sera from 11 dromedary camels, 150 goats, 126 sheep and 91 cows were collected in Jordan, where the first human Middle-East respiratory syndrome (MERS) cluster appeared in 2012. All sera were tested for MERS-coronavirus (MERS-CoV) specific antibodies by protein microarray with confirmation by virus neutralisation. Neutralising antibodies were found in all camel sera while sera from goats and cattle tested negative. Although six sheep sera reacted with MERS-CoV antigen, neutralising antibodies were not detected.
Article
Full-text available
Full text available at: http://currents.plos.org/outbreaks/article/state-of-knowledge-and-data-gaps-of-middle-east-respiratory-syndrome-coronavirus-mers-cov-in-humans-2/ Between September 2012 and 22 October 2013, 144 laboratory-confirmed and 17 probable MERS-CoV cases from nine countries were notified to WHO. We summarize what is known about the epidemiology, virology, phylogeny and emergence of MERS-CoV to inform public health policies. The median age of patients (n=161) was 50 years (range 14 months to 94 years), 64.5% were male and 63.4% experienced severe respiratory disease. 76.0% of patients were reported to have ≥1 underlying medical condition and fatal cases, compared to recovered or asymptomatic cases were more likely to have an underlying condition (86.8% vs. 42.4%, p<0.001). Analysis of genetic sequence data suggests multiple independent introductions into human populations and modelled estimates using epidemiologic and genetic data suggest R<sub>0</sub> is <1, though the upper range of estimates may exceed 1. Index/sporadic cases (cases with no epidemiologic-link to other cases) were more likely to be older (median 59.0 years vs. 43.0 years, p<0.001) compared to secondary cases, although these proportions have declined over time. 80.9% vs. 67.2% of index/sporadic and secondary cases, respectively, reported ≥1 underlying condition. Clinical presentation ranges from asymptomatic to severe pneumonia with acute respiratory distress syndrome and multi-organ failure. Nearly all symptomatic patients presented with respiratory symptoms and 1/3 of patients also had gastrointestinal symptoms. Sustained human-to-human transmission of MERS-CoV has not been observed. Outbreaks have been extinguished without overly aggressive isolation and quarantine suggesting that transmission of virus may be stopped with implementation of appropriate infection control measures.