Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 280
Filtrar
1.
Nat Commun ; 15(1): 5458, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38937429

RESUMO

Respirovirus 3 is a leading cause of severe acute respiratory infections in vulnerable human populations. Entry into host cells is facilitated by the attachment glycoprotein and the fusion glycoprotein (F). Because of its crucial role, F represents an attractive therapeutic target. Here, we identify 13 F-directed heavy-chain-only antibody fragments that neutralize recombinant respirovirus 3. High-resolution cryo-EM structures of antibody fragments bound to the prefusion conformation of F reveal three distinct, previously uncharacterized epitopes. All three antibody fragments bind quaternary epitopes on F, suggesting mechanisms for neutralization that may include stabilization of the prefusion conformation. Studies in cotton rats demonstrate the prophylactic efficacy of these antibody fragments in reducing viral load in the lungs and nasal passages. These data highlight the potential of heavy-chain-only antibody fragments as effective interventions against respirovirus 3 infection and identify neutralizing epitopes that can be targeted for therapeutic development.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Microscopia Crioeletrônica , Epitopos , Animais , Anticorpos Neutralizantes/imunologia , Humanos , Anticorpos Antivirais/imunologia , Epitopos/imunologia , Sigmodontinae , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/química , Proteínas Virais de Fusão/imunologia , Proteínas Virais de Fusão/química , Feminino , Camelus/imunologia , Camelus/virologia
2.
J Virol Methods ; 327: 114923, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38561124

RESUMO

This study describes the development and preliminary validation of a new serological assay using MERS-CoV S1 protein in an indirect enzyme-linked immunosorbent assay (ELISA) format. This assay has the advantage of being able to test MERS-CoV serum samples in a PC2 laboratory without the need for a high-level biocontainment laboratory (PC3 or PC4), which requires highly trained and skilled staff and a high level of resources and equipment. Furthermore, this MERS-CoV S1 ELISA enables a larger number of samples to be tested quickly, with results obtained in approximately five hours. The MERS-CoV S1 ELISA demonstrated high analytical specificity, with no cross-reactivity observed in serum of animals infected with other viruses, including different coronaviruses. We tested 166 positive and 40 negative camel serum samples and have estimated the diagnostic sensitivity (DSe) to be 99.4% (95% CI: 96.7 - 100.0%) and diagnostic specificity (DSp) to be 100% (95% CI: 97.2%-100.0%) relative to the assigned serology results (ppNT and VNT) using a S/P ratio cut-off value of >0.58. The findings of this study showed that our MERS-CoV S1 ELISA was more sensitive than the commercial EUROIMMUN ELISA (Se 99.4% vs 84.9%) and comparable to the ppNT assay, and therefore could be used as a diagnostic aid in countries in the Middle East where MERS-CoV is endemic in dromedary camels. The assay reagents and protocol were easily adapted and transferred from an Australian laboratory to a laboratory in the University of Hong Kong. Thus, the results described here show that the MERS-CoV S1 ELISA represents a cheap, rapid, robust, and reliable assay to support surveillance of MERS-CoV in camels in endemic regions.


Assuntos
Anticorpos Antivirais , Camelídeos Americanos , Camelus , Infecções por Coronavirus , Ensaio de Imunoadsorção Enzimática , Coronavírus da Síndrome Respiratória do Oriente Médio , Sensibilidade e Especificidade , Animais , Camelus/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Ensaio de Imunoadsorção Enzimática/métodos , Ensaio de Imunoadsorção Enzimática/veterinária , Camelídeos Americanos/virologia , Anticorpos Antivirais/sangue , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Testes Sorológicos/métodos , Glicoproteína da Espícula de Coronavírus/imunologia
3.
J Infect Public Health ; 17(6): 986-993, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38631068

RESUMO

Alkhumra hemorrhagic fever virus (AHFV) has spread beyond the Middle East. However, the actual global prevalence of the virus is yet unknown. This systematic review and meta-analysis, thus, followed the standard reporting guidelines to provide comprehensive details on the prevalence of Alkhumra virus infection globally. The pooled prevalence of AHFV globally was estimated at 1.3% (95% CI: 0.3-6.3), with higher prevalence in humans (3.4%, 95% CI: 0.4-25.0) compared to animals (0.7%, 95% CI: 0.3-1.8). The prevalence in ticks and camels were 0.7% and 0.2%, respectively. Overall, there was a high prevalence rate in Asia (2.6%) compared to Africa (0.5%), and a distinctly higher prevalence in Saudi Arabia (4.6%) compared to other parts of the world (<1%). Lower surveillance rate in humans was observed in recent years. These findings will aid public health preparedness, surveillance, and development of preventive measures due to AHFV's potential for outbreaks and severe health consequences.


Assuntos
Saúde Global , Animais , Humanos , África/epidemiologia , Ásia/epidemiologia , Camelus/virologia , Vírus da Encefalite Transmitidos por Carrapatos , Prevalência , Arábia Saudita/epidemiologia , Carrapatos/virologia
4.
BMC Vet Res ; 18(1): 322, 2022 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-35996137

RESUMO

Bluetongue (BT) is an insect-borne, non-contagious viral disease which affects domestic ruminants including camels and is transmitted by Culicoides spp. Clinical symptoms of BT are typically seen in sheep, although subclinical BT infections are mostly seen in cattle, goats, and camelids. The goal of the present study was to evaluate the sero-prevalence of Bluetongue virus (BTV) in camels from some governorates in Egypt's southern and northern regions, as well as the infection's potential risk factors. During 2020-2021, a cross sectional study was conducted to screen presence of anti-BTV antibodies in 400 serum samples, which were collected randomly from camels, examined using competitive enzyme-linked immunosorbent assay (cELISA). The sera of 102 out of 400 camels tested positive for BTV, representing a frequency of 25.5%. Moreover, the odds of sero-positivity were higher among camels living in Aswan (OR = 5.33, 95%CI: 2.35-12.11), especially in females (OR = 2.63, 95%CI = 1.44-4.09) during summer season (OR = 2.40, 95%CI = 1.20-4.81). Furthermore, the probability of getting BTV infection increased when camels were exposed to the insect vectors (OR = 1.63, 95%CI = 0.87-3.09). The high prevalence of BTV in camels in several Egyptian regions highlights the need for more epidemiological investigations of BTV infection in other ruminant species in order to better control BT disease in these regions.


Assuntos
Vírus Bluetongue , Bluetongue , Camelus , Animais , Anticorpos Antivirais/sangue , Bluetongue/epidemiologia , Vírus Bluetongue/imunologia , Vírus Bluetongue/isolamento & purificação , Camelus/virologia , Estudos Transversais , Egito/epidemiologia , Feminino , Masculino , Fatores de Risco , Estudos Soroepidemiológicos
5.
Front Immunol ; 13: 823949, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35173733

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) is an emergent coronavirus that has caused frequent zoonotic events through camel-to-human spillover. An effective camelid vaccination strategy is probably the best way to reduce human exposure risk. Here, we constructed and evaluated an inactivated rabies virus-vectored MERS-CoV vaccine in mice, camels, and alpacas. Potent antigen-specific antibody and CD8+ T-cell responses were generated in mice; moreover, the vaccination reduced viral replication and accelerated virus clearance in MERS-CoV-infected mice. Besides, protective antibody responses against both MERS-CoV and rabies virus were induced in camels and alpacas. Satisfyingly, the immune sera showed broad cross-neutralizing activity against the three main MERS-CoV clades. For further characterization of the antibody response induced in camelids, MERS-CoV-specific variable domains of heavy-chain-only antibody (VHHs) were isolated from immunized alpacas and showed potent prophylactic and therapeutic efficacies in the Ad5-hDPP4-transduced mouse model. These results highlight the inactivated rabies virus-vectored MERS-CoV vaccine as a promising camelid candidate vaccine.


Assuntos
Camelídeos Americanos/virologia , Camelus/virologia , Infecções por Coronavirus/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos T CD8-Positivos/imunologia , Camelídeos Americanos/imunologia , Camelus/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Cricetinae , Feminino , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Vacinação , Vacinas Sintéticas/imunologia , Células Vero , Vacinas Virais/genética
6.
Semin Respir Crit Care Med ; 42(6): 828-838, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34918324

RESUMO

The past two decades have witnessed the emergence of three zoonotic coronaviruses which have jumped species to cause lethal disease in humans: severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), Middle East respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-2. MERS-CoV emerged in Saudi Arabia in 2012 and the origins of MERS-CoV are not fully understood. Genomic analysis indicates it originated in bats and transmitted to camels. Human-to-human transmission occurs in varying frequency, being highest in healthcare environment and to a lesser degree in the community and among family members. Several nosocomial outbreaks of human-to-human transmission have occurred, the largest in Riyadh and Jeddah in 2014 and South Korea in 2015. MERS-CoV remains a high-threat pathogen identified by World Health Organization as a priority pathogen because it causes severe disease that has a high mortality rate, epidemic potential, and no medical countermeasures. MERS-CoV has been identified in dromedaries in several countries in the Middle East, Africa, and South Asia. MERS-CoV-2 causes a wide range of clinical presentations, although the respiratory system is predominantly affected. There are no specific antiviral treatments, although recent trials indicate that combination antivirals may be useful in severely ill patients. Diagnosing MERS-CoV early and implementation infection control measures are critical to preventing hospital-associated outbreaks. Preventing MERS relies on avoiding unpasteurized or uncooked animal products, practicing safe hygiene habits in health care settings and around dromedaries, community education and awareness training for health workers, as well as implementing effective control measures. Effective vaccines for MERS-COV are urgently needed but still under development.


Assuntos
Coronavírus da Síndrome Respiratória do Oriente Médio , Animais , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Camelus/virologia , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Surtos de Doenças/prevenção & controle , Humanos , Controle de Infecções/métodos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade
7.
Viruses ; 13(10)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34696370

RESUMO

Camelpox virus (CMLV) is the causative agent of camelpox, which frequently occurs in the Old World camelids-rearing countries except for Australia. It has also been described in experimentally inoculated New World camelids. Camelpox outbreaks are often experienced shortly after the rainy season, which occurs twice a year on the Arabian Peninsula because of the increased density of the insect population, particularly mosquitos. A systemic form of camelpox outbreak in seven dromedary camels was diagnosed by histology, virus isolation, and PCR. A phylogenetic analysis using full length CMLV genomes of the isolated CMLV strains showed a single phylogenetic unit without any distinctive differences between them. The United Arab Emirates (UAE) isolate sequences showed phylogenetical relatedness with CMLV isolates from Israel with only minor sequence differences. Although the sequences of viruses from both countries were closely related, the disease manifestation was vastly different. Our study shows that the virulence is not only determined by genetic features of CMLV alone but may also depend on other factors such as unknown aspects of the host (e.g., age, overall fitness), management, and the environment.


Assuntos
Camelus/virologia , Surtos de Doenças/estatística & dados numéricos , Surtos de Doenças/veterinária , Orthopoxvirus/genética , Infecções por Poxviridae/epidemiologia , Infecções por Poxviridae/veterinária , Animais , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Orthopoxvirus/classificação , Filogenia , Infecções por Poxviridae/mortalidade , Análise de Sequência de DNA , Emirados Árabes Unidos
8.
Emerg Microbes Infect ; 10(1): 1975-1987, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34570681

RESUMO

ABSTRACTTick-borne viruses (TBVs) capable of transmitting between ticks and hosts have been increasingly recognized as a global public health concern. In this study, Hyalomma ticks and serum samples from camels were collected using recorded sampling correlations in eastern Kenya. Viromes of pooled ticks were profiled by metagenomic sequencing, revealing a diverse community of viruses related to at least 11 families. Five highly abundant viruses, including three novel viruses (Iftin tick virus, Mbalambala tick virus [MATV], and Bangali torovirus [BanToV]) and new strains of previously identified viruses (Bole tick virus 4 [BLTV4] and Liman tick virus [LMTV]), were characterized in terms of genome sequences, organizations, and phylogeny, and their molecular prevalence was investigated in individual ticks. Moreover, viremia and antibody responses to these viruses have been investigated in camels. MATV, BLTV4, LMTV, and BanToV were identified as viral pathogens that can potentially cause zoonotic diseases. The transmission patterns of these viruses were summarized, suggesting three different types according to the sampling relationships between viral RNA-positive ticks and camels positive for viral RNA and/or antibodies. They also revealed the frequent transmission of BanToV and limited but effective transmission of other viruses between ticks and camels. Furthermore, follow-up surveys on TBVs from tick, animal, and human samples with definite sampling relationships are suggested. The findings revealed substantial threats from the emerging TBVs and may guide the prevention and control of TBV-related zoonotic diseases in Kenya and in other African countries.


Assuntos
Camelus/virologia , Infecções por Vírus de RNA/transmissão , Infecções por Vírus de RNA/veterinária , Vírus de RNA/genética , Doenças Transmitidas por Carrapatos/virologia , Carrapatos/virologia , Animais , Genoma Viral/genética , Humanos , Quênia/epidemiologia , RNA Viral/genética , Infestações por Carrapato/epidemiologia , Doenças Transmitidas por Carrapatos/epidemiologia , Carrapatos/classificação , Viroma/genética
9.
Virol J ; 18(1): 160, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34348751

RESUMO

BACKGROUND: Hepatitis E is an enteric and zoonotic disease caused by hepatitis E virus (HEV) that is mainly transmitted via the faecal-oral route through contaminated food or the environment. The virus is an emerging infectious agent causing acute human infection worldwide. A high seroprevalence of the disease was reported in pregnant women in Addis Ababa, Ethiopia, raising significant public health concern. The presence of HEV specific antibodies were also reported in dromedary camels in the country; however, the infectious virus and/or the viral genome have not been demonstrated to date in animal samples. METHODS: To address this gap, a total of 95 faecal samples collected from both apparently healthy pigs of uncharacterised types (50 samples) in Burayu and Addis Ababa areas and camels (Camelus dromedarius, 45 samples) in west Hararghe were screened for the presence of HEV genome using universal primers in a fully nested reverse transcription polymerase chain reaction (nRT-PCR). The protocol is capable of detecting HEV in faecal samples from both pigs and camels. RESULTS: The nRT-PCR detected HEV genes in six (12%) pig faecal samples and one camel sample (2.2%). Therefore, the results indicate that HEV is circulating in both pigs and camels in Ethiopia and these animals and their products could serve as a potential source of infection for humans. CONCLUSION: The detection of HEV in both animals could raise another concern regarding its public health importance as both animals' meat and camel milk are consumed in the country. Further studies to determine the prevalence and distribution of the virus in different animals and their products, water bodies, food chain, and vegetables are warranted, along with viral gene sequencing for detailed genetic characterisation of the isolates circulating in the country. This information is critically important to design and institute appropriate control and/or preventive measures.


Assuntos
Vírus da Hepatite E , Hepatite E , Doenças dos Suínos , Animais , Camelus/virologia , Etiópia/epidemiologia , Feminino , Anticorpos Anti-Hepatite , Hepatite E/epidemiologia , Hepatite E/veterinária , Vírus da Hepatite E/genética , Humanos , Filogenia , Gravidez , RNA Viral , Estudos Soroepidemiológicos , Suínos/virologia , Doenças dos Suínos/epidemiologia
10.
AAPS PharmSciTech ; 22(5): 173, 2021 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-34105037

RESUMO

Middle East respiratory syndrome (MERS) is a lethal respiratory disease with its first case reported back in 2012 (Jeddah, Saudi Arabia). It is a novel, single-stranded, positive-sense RNA beta coronavirus (MERS-CoV) that was isolated from a patient who died from a severe respiratory illness. Later, it was found that this patient was infected with MERS. MERS is endemic to countries in the Middle East regions, such as Saudi Arabia, Jordan, Qatar, Oman, Kuwait and the United Arab Emirates. It has been reported that the MERS virus originated from bats and dromedary camels, the natural hosts of MERS-CoV. The transmission of the virus to humans has been thought to be either direct or indirect. Few camel-to-human transmissions were reported earlier. However, the mode of transmission of how the virus affects humans remains unanswered. Moreover, outbreaks in either family-based or hospital-based settings were observed with high mortality rates, especially in individuals who did not receive proper management or those with underlying comorbidities, such as diabetes and renal failure. Since then, there have been numerous reports hypothesising complications in fatal cases of MERS. Over the years, various diagnostic methods, treatment strategies and preventive measures have been strategised in containing the MERS infection. Evidence from multiple sources implicated that no treatment options and vaccines have been developed in specific, for the direct management of MERS-CoV infection. Nevertheless, there are supportive measures outlined in response to symptom-related management. Health authorities should stress more on infection and prevention control measures, to ensure that MERS remains as a low-level threat to public health.


Assuntos
Infecções por Coronavirus/imunologia , Infecções por Coronavirus/fisiopatologia , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Animais , Antivirais/administração & dosagem , Antivirais/imunologia , Camelus/virologia , Quirópteros/virologia , Infecções por Coronavirus/terapia , Infecções por Coronavirus/transmissão , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Arábia Saudita/epidemiologia , Zoonoses Virais/epidemiologia , Zoonoses Virais/imunologia , Zoonoses Virais/transmissão
11.
Cells ; 10(6)2021 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-34070971

RESUMO

The recent SARS-CoV-2 pandemic has refocused attention to the betacoronaviruses, only eight years after the emergence of another zoonotic betacoronavirus, the Middle East respiratory syndrome coronavirus (MERS-CoV). While the wild source of SARS-CoV-2 may be disputed, for MERS-CoV, dromedaries are considered as source of zoonotic human infections. Testing 100 immune-response genes in 121 dromedaries from United Arab Emirates (UAE) for potential association with present MERS-CoV infection, we identified candidate genes with important functions in the adaptive, MHC-class I (HLA-A-24-like) and II (HLA-DPB1-like), and innate immune response (PTPN4, MAGOHB), and in cilia coating the respiratory tract (DNAH7). Some of these genes previously have been associated with viral replication in SARS-CoV-1/-2 in humans, others have an important role in the movement of bronchial cilia. These results suggest similar host genetic pathways associated with these betacoronaviruses, although further work is required to better understand the MERS-CoV disease dynamics in both dromedaries and humans.


Assuntos
Imunidade Adaptativa/genética , Camelus/virologia , Doenças Transmissíveis Emergentes/imunologia , Infecções por Coronavirus/imunologia , Imunidade Inata/genética , Zoonoses/imunologia , Animais , Anticorpos Antivirais , Brônquios/citologia , Brônquios/fisiologia , COVID-19/genética , COVID-19/imunologia , COVID-19/virologia , Camelus/genética , Camelus/imunologia , Cílios/fisiologia , Doenças Transmissíveis Emergentes/genética , Doenças Transmissíveis Emergentes/transmissão , Doenças Transmissíveis Emergentes/virologia , Infecções por Coronavirus/genética , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/virologia , Reservatórios de Doenças/virologia , Feminino , Predisposição Genética para Doença , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Masculino , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , Mucosa Respiratória/citologia , Mucosa Respiratória/fisiologia , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Emirados Árabes Unidos , Replicação Viral/genética , Replicação Viral/imunologia , Zoonoses/genética , Zoonoses/transmissão , Zoonoses/virologia
12.
Parasit Vectors ; 14(1): 342, 2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34187526

RESUMO

BACKGROUND: Crimean-Congo hemorrhagic fever virus (CCHFV) belongs to the genus Orthonairovirus (Nairovididae) and is a (re)emerging tick-borne pathogen. It is endemic in most parts of Africa, Asia and southern Europe, and can cause severe hemorrhagic symptoms in humans, with high fatality rates (5-30%). METHODS: Hyalomma ticks were collected from four different livestock herds (cattle and camels) in Mauritania in 2018. The tick species were determined morphologically and confirmed molecularly by using the cytochrome oxidase 1 gene marker. For the detection of CCHFV, ticks were tested individually by one-step multiplex real-time reverse-transcriptase quantitative polymerase chain reaction. The small segment of all positive samples was sequenced to determine the CCHFV genotype. RESULTS: In total, 39 of the 1523 ticks (2.56%) collected from 63 cattles and 28 camels tested positive for CCHFV. Three Hyalomma species were identified. Hyalomma rufipes had the largest proportion of positivity (5.67%; 16/282), followed by Hyalomma dromedarii (1.89%; 23/1214). No Hyalomma impeltatum tested positive (0%; 0/21). Positive ticks were found in only six out of 91 host animals. Viral sequence analysis revealed the presence of two different CCHFV lineages (Africa I and Africa III). CONCLUSIONS: In this study, 2.56% of Hyalomma ticks collected from camels and cattle in Mauritania tested positive for CCHFV. However, the true prevalence of CCHFV in unfed ticks may be lower, as a considerable number of ticks may have been passively infected during blood-feeding by co-feeding ticks or due to viremia of the host. The results indicate the need to track the actual area of circulation of this virus.


Assuntos
Sangue , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Gado/parasitologia , Carrapatos/virologia , Animais , Camelus/parasitologia , Camelus/virologia , Bovinos/parasitologia , Bovinos/virologia , Comportamento Alimentar , Feminino , Genótipo , Vírus da Febre Hemorrágica da Crimeia-Congo/isolamento & purificação , Febre Hemorrágica da Crimeia/virologia , Gado/virologia , Masculino , Mauritânia , Filogenia , RNA Viral/genética , Carrapatos/genética , Carrapatos/fisiologia
13.
Zoonoses Public Health ; 68(5): 527-532, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33966359

RESUMO

Middle East respiratory syndrome (MERS) is caused by MERS-CoV that infects both human and camel. Camel is supposed to be the natural reservoir for human infection while the sources for most of the primary human infection cases are still not known. We identified two conserved pyrimidine nucleotides that flank UAAU element in MERS-CoV 5'-UTR. These conserved pyrimidine nucleotides distinguish MERS-CoVs into 3 types, that is, UUAAUU, CUAAUU and CUAAUC (referred to as U----U, C----U, and C----C types, respectively). Human MERS-CoV displays a genetic drift from U----U, C----U, to C----C from 2012 to 2019. Camel virus displays a genetic drift from U----U to C----U with a time lag when compared with human virus. The discrepancy in genetic drift seems not to support the notion that camel serves as the only natural reservoir for human infection.


Assuntos
Camelus/virologia , Infecções por Coronavirus/virologia , Deriva Genética , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Regiões 5' não Traduzidas/genética , Animais , Sequência de Bases , Infecções por Coronavirus/epidemiologia , Variação Genética , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/classificação , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Filogenia , Prevalência , Arábia Saudita/epidemiologia
14.
Viruses ; 13(4)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33807288

RESUMO

Middle East respiratory syndrome-related coronavirus (MERS-CoV) is a persistent zoonotic pathogen with frequent spillover from dromedary camels to humans in the Arabian Peninsula, resulting in limited outbreaks of MERS with a high case-fatality rate. Full genome sequence data from camel-derived MERS-CoV variants show diverse lineages circulating in domestic camels with frequent recombination. More than 90% of the available full MERS-CoV genome sequences derived from camels are from just two countries, the Kingdom of Saudi Arabia (KSA) and United Arab Emirates (UAE). In this study, we employ a novel method to amplify and sequence the partial MERS-CoV genome with high sensitivity from nasal swabs of infected camels. We recovered more than 99% of the MERS-CoV genome from field-collected samples with greater than 500 TCID50 equivalent per nasal swab from camel herds sampled in Jordan in May 2016. Our subsequent analyses of 14 camel-derived MERS-CoV genomes show a striking lack of genetic diversity circulating in Jordan camels relative to MERS-CoV genome sequences derived from large camel markets in KSA and UAE. The low genetic diversity detected in Jordan camels during our study is consistent with a lack of endemic circulation in these camel herds and reflective of data from MERS outbreaks in humans dominated by nosocomial transmission following a single introduction as reported during the 2015 MERS outbreak in South Korea. Our data suggest transmission of MERS-CoV among two camel herds in Jordan in 2016 following a single introduction event.


Assuntos
Camelus/virologia , Infecções por Coronavirus/veterinária , Variação Genética , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Zoonoses/virologia , Animais , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/virologia , Genoma Viral , Jordânia/epidemiologia , Coronavírus da Síndrome Respiratória do Oriente Médio/classificação , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Filogenia , República da Coreia/epidemiologia , Arábia Saudita/epidemiologia , Emirados Árabes Unidos/epidemiologia , Zoonoses/epidemiologia
15.
Viruses ; 13(3)2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33800270

RESUMO

Picobirnaviruses (PBVs) are small non-enveloped bisegmented double-stranded RNA viruses found in humans, mammals, and birds. Increasing molecular epidemiology studies suggest a high sequence diversity of PBVs in numerous hosts and the environment. In this study, using 229 fecal samples from dromedary camels in Dubai, 52.8% were positive for PBVs, of which 77.7% and 41.3% were positive for genogroup I and II, respectively, and 19.0% were positive for both genotypes. Phylogenetic analysis showed high diversity among the sequences of genogroup I and II dromedary PBVs. Marked nucleotide polymorphisms were observed in 75.5% and 46.0% of genogroup I and II RNA-dependent RNA polymerase (RdRp) sequences, respectively, suggesting the co-existence of multiple strains in the same specimen. Both high genetic diversity and prevalence of genogroup I and II PBV in dromedaries were observed. In fact, the prevalence of genogroup II PBV in dromedaries is the highest among all animals to date. The complete/near-complete core genomes of five genogroup I and one genogroup II dromedary PBVs and partial segment 1 and 2 of both genotypes were also sequenced. The dromedary PBV genome organizations were similar to those of other animals. Genetic reassortment and mutation are both important in the ecology and evolution of PBVs.


Assuntos
Camelus/virologia , Variação Genética , Genótipo , Picobirnavirus/classificação , Picobirnavirus/genética , Infecções por Vírus de RNA/epidemiologia , Infecções por Vírus de RNA/veterinária , Animais , Evolução Molecular , Fezes , Genoma Viral , Filogenia , Picobirnavirus/isolamento & purificação , Prevalência , RNA Viral/genética , Emirados Árabes Unidos/epidemiologia
16.
J Med Virol ; 93(9): 5328-5332, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33851740

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) is one of the recently identified zoonotic coronaviruses. The one-hump camels are believed to play important roles in the evolution and transmission of the virus. The animal-to-animal, as well as the animal-to-human transmission in the context of MERS-CoV infection, were reported. The camels shed the virus in some of their secretions, especially the nasal tract. However, there are many aspects of the transmission cycle of the virus from animals to humans that are still not fully understood. Rodents played important roles in the transmission of many pathogens, including viruses and bacteria. They have been implicated in the evolution of many human coronaviruses, especially HCoV-OC43 and HCoV-HKU1. However, the role of rodents in the transmission of MERS-CoV still requires more exploration. To achieve this goal, we identified MERS-CoV that naturally infected dromedary camel by molecular surveillance. We captured 15 of the common rodents (rats, mice, and jerboa) sharing the habitat with these animals. We collected both oral and rectal swabs from these animals and then tested them by the commercial MERS-CoV real-time-PCR kits using two targets. Despite the detection of the viral shedding in the nasal swabs of some of the dromedary camels, none of the rodents tested positive for the virus during the tenure of this study. We concluded that these species of rodents did not harbor the virus and are most unlikely to contribute to the transmission of the MERS-CoV. However, further large-scale studies are required to confirm the potential roles of rodents in the context of the MERS-CoV transmission cycle, if any.


Assuntos
Camelus/virologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/veterinária , Monitoramento Epidemiológico/veterinária , RNA Viral/genética , Animais , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/virologia , Humanos , Camundongos , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , Cavidade Nasal/virologia , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reto/virologia , Roedores/virologia , Arábia Saudita/epidemiologia
17.
Trop Anim Health Prod ; 53(2): 195, 2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33666802

RESUMO

This study reports the monitoring of several emerging viral pathogens in Mauritania, which was carried out by the analysis of bovine and camel samples taken at the slaughterhouse of Nouakchott. Blood and serum were collected by random sampling from 159 camels and 118 cattle in March 2013 at the large animals abattoir in Nouakchott. Serological tests for Rift Valley Fever (RVF), Peste des Petits Ruminants (PPR), West Nile disease (WND), epizootic haemorrhagic disease (EHD) and African horse sickness (AHS) were carried out using commercial ELISA kits. The samples, which resulted positives for PPR, WND and AHS, were tested with the confirmatory virus neutralization test (VNT). According to ELISA results, serological prevalence of RVF was 45% (95% CI 52.3-37.7) in camels and 16% (95% CI 22.6-9.4) in cattle. The difference between the observed prevalences in camels and in cattle was significant (p value ≤ 0.01). PPR was absent in camels and had 12% prevalence (95% CI, 17.86-6.14) in cattle. Furthermore, camels showed 92% (95% CI, 96.1-87.9) prevalence of WNV, 73% (95% CI, 82.3-63.64) of EHD and 3% (95% CI, 5.6-0.4) of AHS. This data are of relevance since provided useful feedbacks on the circulation of the pathogens in field. Moreover, this survey provided new information on the susceptibility of camels to several emerging pathogens and on the possible use of this species as sentinel animal.


Assuntos
Matadouros , Camelus/virologia , Doenças dos Bovinos/epidemiologia , Viroses/veterinária , Doença Equina Africana/epidemiologia , Doença Equina Africana/virologia , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/imunologia , Bovinos , Doenças dos Bovinos/virologia , Ensaio de Imunoadsorção Enzimática/veterinária , Vírus da Doença Hemorrágica Epizoótica/imunologia , Vírus da Doença Hemorrágica Epizoótica/isolamento & purificação , Mauritânia/epidemiologia , Vírus da Peste dos Pequenos Ruminantes/imunologia , Vírus da Peste dos Pequenos Ruminantes/isolamento & purificação , Febre do Vale de Rift/epidemiologia , Febre do Vale de Rift/virologia , Estudos Soroepidemiológicos , Viroses/epidemiologia , Viroses/virologia , Febre do Nilo Ocidental/epidemiologia , Febre do Nilo Ocidental/veterinária , Febre do Nilo Ocidental/virologia
18.
BMC Res Notes ; 14(1): 96, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33691761

RESUMO

OBJECTIVE: The Middle East respiratory syndrome coronavirus (MERS-CoV) is one of the zoonotic coronaviruses [Hemida Peer J 7:e7556, 2019; Hemida et al. One Health 8:100102, 2019]. The dromedary camels remained the only known animal reservoir for this virus. Several aspects of the transmission cycle of the virus between animals, including arthropod-borne infection, is still largely unknown. The main objective of the current work was to study the possibility of MERS-CoV transmission through some arthropod vectors, particularly the hard ticks. To achieve this objective, we identified a positive MERS-CoV dromedary camel herd using the commercial available real-time PCR kits. We collected some arthropods, particularly the ticks from these positive animals as well as from the animal habitats. We tested these arthropods for the presence of MERS-CoV viral RNAs. RESULTS: Our results showing the absence of any detectable MERS-CoV-RNAs in these arthropods despite these animals were actively shedding the virus in their nasal secretions. Our results are confirming for the first the failure of detection of the MERS-CoV in ticks infesting dromedary camels. Failure of the detection of MERS-CoV in ticks infesting positive naturally infected MERS-CoV camels is strongly suggesting that ticks do not play roles in the transmission of the virus among the animals and close contact humans.


Assuntos
Vetores Artrópodes/virologia , Camelus/virologia , Infecções por Coronavirus/transmissão , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Carrapatos/virologia , Animais , Infecções por Coronavirus/epidemiologia , Ecossistema , Feminino , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Arábia Saudita/epidemiologia
19.
Zoonoses Public Health ; 68(4): 353-357, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33565270

RESUMO

Dromedary camels are playing essential roles in the evolution and transmission of MERS-CoV. MERS-CoV shedding in some dromedary camel secretions, particularly nasal swabs, were studied in more detail. However, the roles of viral shedding in saliva and ocular secretions are still required further detailed studies. We performed a longitudinal study on a farm of dromedary camel herd from 10th March until 7th April, 2019, in eastern Saudi Arabia. This is a closed management herd including a large number of colour-based breed animals and include animals of both sexes. We collected saliva and ocular swabs from 18% of the target animal population. Detection of the MERS-CoV-RNAs in these samples was conducted by the real-time PCR technique. We detected the viral RNAs in the saliva of and conjunctival swabs of some of the tested animals at 33%, 77% and 88% during the three-time points, respectively. Moreover, we also detected the viral RNAs in the conjunctival swabs at 11%, 22% and 33% at similar time intervals. Our results are suggesting the possibility of MERS-CoV shedding in the saliva and the ocular discharges of the infected dromedary camels. This explains, at least in part, the mechanism of transmission of MERS-CoV from animals to humans. More studies are needed for a better understanding of the transmission of MERS-CoV from animals to humans; thus, the risk of virus spread can be mitigated.


Assuntos
Camelus/virologia , Túnica Conjuntiva/química , Infecções por Coronavirus/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , RNA Viral/análise , Saliva/química , Animais , Filogenia , Arábia Saudita/epidemiologia , Eliminação de Partículas Virais
20.
J Epidemiol Glob Health ; 11(2): 155-159, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33605109

RESUMO

Countries in the Middle-East (ME) are tackling two corona virus outbreaks simultaneously, Middle-Eastern Respiratory Syndrome Coronavirus (MERS-CoV) and the current Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Both viruses infect the same host (humans) and the same cell (type-II alveolar cells) causing lower respiratory illnesses such as pneumonia. Molecularly, MERS-CoV and SARS-CoV-2 enter alveolar cells via spike proteins recognizing dipeptidyl peptidase-4 and angiotensin converting enzyme-II, respectively. Intracellularly, both viruses hide in organelles to generate negative RNA strands and initiate replication using very similar mechanisms. At the transcription level, both viruses utilise identical Transcription Regulatory Sequences (TRSs), which are known recombination cross-over points during replication, to transcribe genes. Using whole genome alignments of both viruses, we identify clusters of high sequence homology at ORF1a and ORF1b. Given the high recombination rates detected in SARS-CoV-2, we speculate that in co-infections recombination is feasible via TRS and/or clusters of homologies. Accordingly, here we recommend mitigation measure and testing for both MERS-CoV and SARS-CoV-2 in ME countries.


Assuntos
COVID-19/epidemiologia , Coinfecção/epidemiologia , Infecções por Coronavirus/epidemiologia , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Recombinação Genética , SARS-CoV-2/genética , Animais , COVID-19/virologia , Camelus/virologia , Humanos , Oriente Médio/epidemiologia , Zoonoses Virais/epidemiologia , Zoonoses Virais/transmissão , Zoonoses Virais/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...