Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
PLoS Biol ; 18(12): e3001016, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33347434

RESUMO

SARS Coronavirus 2 (SARS-CoV-2) emerged in late 2019, leading to the Coronavirus Disease 2019 (COVID-19) pandemic that continues to cause significant global mortality in human populations. Given its sequence similarity to SARS-CoV, as well as related coronaviruses circulating in bats, SARS-CoV-2 is thought to have originated in Chiroptera species in China. However, whether the virus spread directly to humans or through an intermediate host is currently unclear, as is the potential for this virus to infect companion animals, livestock, and wildlife that could act as viral reservoirs. Using a combination of surrogate entry assays and live virus, we demonstrate that, in addition to human angiotensin-converting enzyme 2 (ACE2), the Spike glycoprotein of SARS-CoV-2 has a broad host tropism for mammalian ACE2 receptors, despite divergence in the amino acids at the Spike receptor binding site on these proteins. Of the 22 different hosts we investigated, ACE2 proteins from dog, cat, and cattle were the most permissive to SARS-CoV-2, while bat and bird ACE2 proteins were the least efficiently used receptors. The absence of a significant tropism for any of the 3 genetically distinct bat ACE2 proteins we examined indicates that SARS-CoV-2 receptor usage likely shifted during zoonotic transmission from bats into people, possibly in an intermediate reservoir. Comparison of SARS-CoV-2 receptor usage to the related coronaviruses SARS-CoV and RaTG13 identified distinct tropisms, with the 2 human viruses being more closely aligned. Finally, using bioinformatics, structural data, and targeted mutagenesis, we identified amino acid residues within the Spike-ACE2 interface, which may have played a pivotal role in the emergence of SARS-CoV-2 in humans. The apparently broad tropism of SARS-CoV-2 at the point of viral entry confirms the potential risk of infection to a wide range of companion animals, livestock, and wildlife.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Tropismo Viral , Ligação Viral , Substituição de Aminoácidos , Animais , Sítios de Ligação , Gatos , Bovinos , Cães , Cobaias , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Coelhos , Ratos , Zoonoses Virais/virologia
2.
Hepatology ; 71(3): 794-807, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31400152

RESUMO

BACKGROUND AND AIMS: The lack of immunocompetent small animal models for hepatitis C virus (HCV) has greatly hindered the development of effective vaccines. Using rodent hepacivirus (RHV), a homolog of HCV that shares many characteristics of HCV infection, we report the development and application of an RHV outbred rat model for HCV vaccine development. APPROACH AND RESULTS: Simian adenovirus (ChAdOx1) encoding a genetic immune enhancer (truncated shark class II invariant chain) fused to the nonstructural (NS) proteins NS3-NS5B from RHV (ChAd-NS) was used to vaccinate Sprague-Dawley rats, resulting in high levels of cluster of differentiation 8-positive (CD8+ ) T-cell responses. Following RHV challenge (using 10 or 100 times the minimum infectious dose), 42% of vaccinated rats cleared infection within 6-8 weeks, while all mock vaccinated controls became infected with high-level viremia postchallenge. A single, 7-fold higher dose of ChAd-NS increased efficacy to 67%. Boosting with ChAd-NS or with a plasmid encoding the same NS3-NS5B antigens increased efficacy to 100% and 83%, respectively. A ChAdOx1 vector encoding structural antigens (ChAd-S) was also constructed. ChAd-S alone showed no efficacy. Strikingly, when combined with ChAd-NS, ChAD-S produced 83% efficacy. Protection was associated with a strong CD8+ interferon gamma-positive recall response against NS4. Next-generation sequencing of a putative RHV escape mutant in a vaccinated rat identified mutations in both identified immunodominant CD8+ T-cell epitopes. CONCLUSIONS: A simian adenovirus vector vaccine strategy is effective at inducing complete protective immunity in the rat RHV model. The RHV Sprague-Dawley rat challenge model enables comparative testing of vaccine platforms and antigens and identification of correlates of protection and thereby provides a small animal experimental framework to guide the development of an effective vaccine for HCV in humans.


Assuntos
Hepacivirus/imunologia , Vacinação , Vacinas contra Hepatite Viral/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Epitopos de Linfócito T , Interferon gama/sangue , Masculino , Ratos , Ratos Sprague-Dawley , Vacinas Sintéticas/imunologia , Proteínas não Estruturais Virais/imunologia
3.
Nucleic Acids Res ; 47(15): 8061-8083, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31276592

RESUMO

Zinc finger antiviral protein (ZAP) is a powerful restriction factor for viruses with elevated CpG dinucleotide frequencies. We report that ZAP similarly mediates antiviral restriction against echovirus 7 (E7) mutants with elevated frequencies of UpA dinucleotides. Attenuation of both CpG- and UpA-high viruses and replicon mutants was reversed in ZAP k/o cell lines, and restored by plasmid-derived reconstitution of expression in k/o cells. In pull-down assays, ZAP bound to viral RNA transcripts with either CpG- and UpA-high sequences inserted in the R2 region. We found no evidence that attenuation of CpG- or UpA-high mutants was mediated through either translation inhibition or accelerated RNA degradation. Reversal of the attenuation of CpG-high, and UpA-high E7 viruses and replicons was also achieved through knockout of RNAseL and oligodenylate synthetase 3 (OAS3), but not OAS1. WT levels of replication of CpG- and UpA-high mutants were observed in OAS3 k/o cells despite abundant expression of ZAP, indicative of synergy or complementation of these hitherto unconnected pathways. The dependence on expression of ZAP, OAS3 and RNAseL for CpG/UpA-mediated attenuation and the variable and often low level expression of these pathway proteins in certain cell types, such as those of the central nervous system, has implications for the use of CpG-elevated mutants as attenuated live vaccines against neurotropic viruses.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , Endorribonucleases/metabolismo , Regulação da Expressão Gênica , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , 2',5'-Oligoadenilato Sintetase/genética , Células A549 , Linhagem Celular Tumoral , Ilhas de CpG/genética , Fosfatos de Dinucleosídeos/genética , Endorribonucleases/genética , Enterovirus Humano B/genética , Técnicas de Inativação de Genes , Humanos , Mutação , Ligação Proteica , RNA Viral/genética , RNA Viral/metabolismo , Proteínas de Ligação a RNA/genética , Replicon/genética
4.
Emerg Infect Dis ; 26(12): 3030-3033, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33219787

RESUMO

We report a unique outbreak of Rift Valley fever in the Eldamar area, Sudan, May-July 2019, that resulted in 1,129 case-patients and 19 (1.7%) deaths. Patients exhibited clinical signs including fever (100%), headache (79%), and bleeding (4%). Most (98%) patients also reported death and abortions among their livestock.


Assuntos
Aborto Espontâneo , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Surtos de Doenças , Feminino , Humanos , Gado , Gravidez , Febre do Vale de Rift/diagnóstico , Febre do Vale de Rift/epidemiologia , Vírus da Febre do Vale do Rift/genética , Sudão/epidemiologia
5.
Proc Natl Acad Sci U S A ; 112(2): E176-85, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25548172

RESUMO

Arboviruses are transmitted by mosquitoes and other arthropods to humans and animals. The risk associated with these viruses is increasing worldwide, including new emergence in Europe and the Americas. Anopheline mosquitoes are vectors of human malaria but are believed to transmit one known arbovirus, o'nyong-nyong virus, whereas Aedes mosquitoes transmit many. Anopheles interactions with viruses have been little studied, and the initial antiviral response in the midgut has not been examined. Here, we determine the antiviral immune pathways of the Anopheles gambiae midgut, the initial site of viral infection after an infective blood meal. We compare them with the responses of the post-midgut systemic compartment, which is the site of the subsequent disseminated viral infection. Normal viral infection of the midgut requires bacterial flora and is inhibited by the activities of immune deficiency (Imd), JAK/STAT, and Leu-rich repeat immune factors. We show that the exogenous siRNA pathway, thought of as the canonical mosquito antiviral pathway, plays no detectable role in antiviral defense in the midgut but only protects later in the systemic compartment. These results alter the prevailing antiviral paradigm by describing distinct protective mechanisms in different body compartments and infection stages. Importantly, the presence of the midgut bacterial flora is required for full viral infectivity to Anopheles, in contrast to malaria infection, where the presence of the midgut bacterial flora is required for protection against infection. Thus, the enteric flora controls a reciprocal protection tradeoff in the vector for resistance to different human pathogens.


Assuntos
Anopheles/imunologia , Anopheles/virologia , Arbovírus/imunologia , Arbovírus/patogenicidade , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/transmissão , Animais , Anopheles/genética , Infecções por Arbovirus/imunologia , Infecções por Arbovirus/transmissão , Arbovírus/genética , Sistema Digestório/imunologia , Sistema Digestório/microbiologia , Sistema Digestório/virologia , Feminino , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Insetos Vetores/genética , Insetos Vetores/imunologia , Insetos Vetores/virologia , Janus Quinases/imunologia , Microbiota , Vírus O'nyong-nyong/genética , Vírus O'nyong-nyong/imunologia , Vírus O'nyong-nyong/patogenicidade , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais/imunologia
6.
J Gen Virol ; 95(Pt 3): 517-530, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24343914

RESUMO

Arthropod-borne viruses (arboviruses) pose a considerable threat to human and animal health, yet effective control measures have proven difficult to implement, and novel means of controlling their replication in arthropod vectors, such as mosquitoes, are urgently required. One of the most exciting approaches to emerge from research on arthropods is the use of the endosymbiotic intracellular bacterium Wolbachia to control arbovirus transmission from mosquito to vertebrate. These α-proteobacteria propagate through insects, in part through modulation of host reproduction, thus ensuring spread through species and maintenance in nature. Since it was discovered that Wolbachia endosymbiosis inhibits insect virus replication in Drosophila species, these bacteria have also been shown to inhibit arbovirus replication and spread in mosquitoes. Importantly, it is not clear how these antiviral effects are mediated. This review will summarize recent work and discuss determinants of antiviral effectiveness that may differ between individual Wolbachia/vector/arbovirus interactions. We will also discuss the application of this approach to field settings and the associated risks.


Assuntos
Aedes/microbiologia , Aedes/virologia , Arbovírus/fisiologia , Controle de Doenças Transmissíveis , Insetos Vetores/virologia , Wolbachia/fisiologia , Aedes/fisiologia , Animais , Doenças Transmissíveis/virologia , Humanos , Insetos Vetores/microbiologia , Insetos Vetores/fisiologia
7.
J Biol Chem ; 287(20): 16289-99, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22451671

RESUMO

The obligate intracellular and promiscuous protozoan parasite Toxoplasma gondii needs an extensive membrane biogenesis that must be satisfied irrespective of its host-cell milieu. We show that the synthesis of the major lipid in T. gondii, phosphatidylcholine (PtdCho), is initiated by a novel choline kinase (TgCK). Full-length (∼70-kDa) TgCK displayed a low affinity for choline (K(m) ∼0.77 mM) and harbors a unique N-terminal hydrophobic peptide that is required for the formation of enzyme oligomers in the parasite cytosol but not for activity. Conditional mutagenesis of the TgCK gene in T. gondii attenuated the protein level by ∼60%, which was abolished in the off state of the mutant (Δtgck(i)). Unexpectedly, the mutant was not impaired in its growth and exhibited a normal PtdCho biogenesis. The parasite compensated for the loss of full-length TgCK by two potential 53- and 44-kDa isoforms expressed through a cryptic promoter identified within exon 1. TgCK-Exon1 alone was sufficient in driving the expression of GFP in E. coli. The presence of a cryptic promoter correlated with the persistent enzyme activity, PtdCho synthesis, and susceptibility of T. gondii to a choline analog, dimethylethanolamine. Quite notably, the mutant displayed a regular growth in the off state despite a 35% decline in PtdCho content and lipid synthesis, suggesting a compositional flexibility in the membranes of the parasite. The observed plasticity of gene expression and membrane biogenesis can ensure a faithful replication and adaptation of T. gondii in disparate host or nutrient environments.


Assuntos
Colina Quinase/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Mutagênese , Fosfatidilcolinas/biossíntese , Proteínas de Protozoários/biossíntese , Toxoplasma/enzimologia , Sequência de Bases , Colina Quinase/genética , Deanol/metabolismo , Dados de Sequência Molecular , Mutação , Fosfatidilcolinas/genética , Multimerização Proteica/fisiologia , Proteínas de Protozoários/genética , Toxoplasma/genética
8.
J Gen Virol ; 94(Pt 5): 917-932, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23364195

RESUMO

Almost 30 years after its initial discovery, infection with the human immunodeficiency virus-1 (HIV-1) remains incurable and the virus persists due to reservoirs of latently infected CD4(+) memory T-cells and sanctuary sites within the infected individual where drug penetration is poor. Reactivating latent viruses has been a key strategy to completely eliminate the virus from the host, but many difficulties and unanswered questions remain. In this review, the latest developments in HIV-persistence and latency research are presented.


Assuntos
Fármacos Anti-HIV/farmacocinética , Infecções por HIV/virologia , HIV-1/fisiologia , Latência Viral , Linfócitos T CD4-Positivos/virologia , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Humanos , Modelos Biológicos , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos
9.
Pathogens ; 12(4)2023 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-37111448

RESUMO

The ongoing global emergence of arthropod-borne (arbo) viruses has accelerated research into the interactions of these viruses with the immune systems of their vectors. Only limited information exists on how bunyaviruses, such as Rift Valley fever virus (RVFV), are sensed by mosquito immunity or escape detection. RVFV is a zoonotic phlebovirus (Bunyavirales; Phenuiviridae) of veterinary and human public health and economic importance. We have shown that the infection of mosquitoes with RVFV triggers the activation of RNA interference pathways, which moderately restrict viral replication. Here, we aimed to better understand the interactions between RVFV and other vector immune signaling pathways that might influence RVFV replication and transmission. For this, we used the immunocompetent Aedes aegypti Aag2 cell line as a model. We found that bacteria-induced immune responses restricted RVFV replication. However, virus infection alone did not alter the gene expression levels of immune effectors. Instead, it resulted in the marked enhancement of immune responses to subsequent bacterial stimulation. The gene expression levels of several mosquito immune pattern recognition receptors were altered by RVFV infection, which may contribute to this immune priming. Our findings imply that there is a complex interplay between RVFV and mosquito immunity that could be targeted in disease prevention strategies.

10.
J Virol ; 85(12): 5840-52, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21490095

RESUMO

Domestic cats endure infections by all three subfamilies of the retroviridae: lentiviruses (feline immunodeficiency virus [FIV]), gammaretroviruses (feline leukemia virus [FeLV]), and spumaretroviruses (feline foamy virus [FFV]). Thus, cats present an insight into the evolution of the host-retrovirus relationship and the development of intrinsic/innate immune mechanisms. Tetherin (BST-2) is an interferon-inducible transmembrane protein that inhibits the release of enveloped viruses from infected cells. Here, we characterize the feline homologue of tetherin and assess its effects on the replication of FIV. Tetherin was expressed in many feline cell lines, and expression was induced by interferons, including alpha interferon (IFN-α), IFN-ω, and IFN-γ. Like human tetherin, feline tetherin displayed potent inhibition of FIV and HIV-1 particle release; however, this activity resisted antagonism by either HIV-1 Vpu or the FIV Env and "OrfA" proteins. Further, as overexpression of complete FIV genomes in trans could not overcome feline tetherin, these data suggest that FIV lacks a functional tetherin antagonist. However, when expressed stably in feline cell lines, tetherin did not abrogate the replication of FIV; indeed, syncytium formation was significantly enhanced in tetherin-expressing cells infected with cell culture-adapted (CD134-independent) strains of FIV (FIV Fca-F14 and FIV Pco-CoLV). Thus, while tetherin may prevent the release of nascent viral particles, cell-to-cell spread remains efficient in the presence of abundant viral receptors and tetherin upregulation may enhance syncytium formation. Accordingly, tetherin expression in vivo may promote the selective expansion of viral variants capable of more efficient cell-to-cell spread.


Assuntos
Antígenos CD/farmacologia , Proteínas Ligadas por GPI/farmacologia , Vírus da Imunodeficiência Felina/fisiologia , Vírus da Imunodeficiência Felina/patogenicidade , Liberação de Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Antígenos CD/química , Antígenos CD/efeitos dos fármacos , Antígenos CD/genética , Antígenos CD/metabolismo , Gatos , Linhagem Celular , Cães , Fibroblastos/virologia , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/efeitos dos fármacos , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Células Gigantes/fisiologia , Humanos , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Interferons/farmacologia , Camundongos , Dados de Sequência Molecular , Receptores CXCR4/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Replicação Viral
11.
Microorganisms ; 10(5)2022 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-35630372

RESUMO

Crimean-Congo hemorrhagic fever (CCHF) is a zoonotic arboviral disease that poses a great threat to global health in the Old World, and it is endemic in Europe, Asia, and Africa, including Sudan. In this retrospective study, we reviewed previous epidemiological reports about the major epidemics of CCHF throughout Sudan between 2010 and 2020. During these epidemics, the infection of humans with Crimean-Congo hemorrhagic fever virus (CCHFV), the causative agent of CCHF, was diagnosed using qRT-PCR. We have identified 88 cases of CCHF, including 13 fatalities reported during five epidemics that occurred in 2010, 2011, 2015, 2019, and 2020. The two epidemics in 2010 and 2011 were by far the largest, with 51 and 27 cases reported, respectively. The majority of cases (78%) were reported in the endemic region of Kordofan. Here, we document that the first emergence of CCHFV in the Darfur region, West Sudan, occurred in 2010. We were not able to investigate outbreak dynamics through phylogenetic analysis due to the limited diagnostic capacity and the lack of sequencing services in the country. These findings call for establishing a genomic-based integrated One Health surveillance and response system for the early preparedness, prevention, and control of CCHF in the country.

12.
J Virol ; 84(17): 8980-5, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20554781

RESUMO

A synthetic feline TRIM5-cyclophilin A fusion protein (feTRIMCyp) was generated and transduced into feline cells. feTRIMCyp was highly efficient at preventing infection with human (HIV) and feline (FIV) immunodeficiency virus pseudotypes, and feTRIMCyp-expressing cells resisted productive infection with either FIV-Fca or FIV-Pco. The restriction of FIV infection by feTRIMCyp was reversed by the cyclosporine (Cs) derivatives NIM811 and Debio-025 but less so by Cs itself. FeTRIMCyp and FIV infections of the cat offer a unique opportunity to evaluate TRIMCyp-based approaches to genetic therapy for HIV infection and the treatment of AIDS.


Assuntos
Proteínas de Transporte/metabolismo , Ciclofilina A/metabolismo , Síndrome de Imunodeficiência Adquirida Felina/virologia , Infecções por HIV/virologia , HIV-1/fisiologia , Vírus da Imunodeficiência Felina/fisiologia , Internalização do Vírus , Animais , Fatores de Restrição Antivirais , Proteínas de Transporte/síntese química , Proteínas de Transporte/genética , Gatos , Linhagem Celular , Ciclofilina A/síntese química , Ciclofilina A/genética , Modelos Animais de Doenças , Síndrome de Imunodeficiência Adquirida Felina/prevenção & controle , Infecções por HIV/prevenção & controle , Humanos , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
13.
Int J Infect Dis ; 108: 513-516, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34044142

RESUMO

OBJECTIVES: This study aimed to investigate an outbreak of a non-malaria, undifferentiated febrile illness, among internally displaced persons (IDPs) living in humanitarian camps in North Darfur, Sudan, in 2019. METHODS: An investigation team was deployed to North Darfur to identify suspected cases and collect blood samples, and clinical and demographical data. Blood samples were examined microscopically for Plasmodium spp and tested for dengue (DENV) and yellow fever viruses by reverse transcriptase-quantitative polymerase chain reaction. RESULTS: Between September 7 and December 18, 2019, we clinically identified 18 (24%), 41 (54%), and 17 (22%) cases of dengue fever, dengue with warning signs, and severe dengue, respectively. Blood samples were collected from 22% of patients, and 47% of these tested positive for DENV-1 RNA. We confirmed 32 malaria cases with 5 co-infections with DENV. This outbreak of dengue was the first among IDPs in the humanitarian camps. CONCLUSIONS: Our findings indicate that dengue has become endemic or that there has been a new introduction. Further epidemiological, entomological, and phylogenetic studies are needed to understand disease transmission in the area. An early warning and response system and an effective health policy are crucial for preventing and controlling arboviruses in Sudan.


Assuntos
Coinfecção , Vírus da Dengue , Dengue , Epidemias , Malária , Refugiados , Coinfecção/epidemiologia , Dengue/epidemiologia , Vírus da Dengue/genética , Surtos de Doenças , Humanos , Malária/epidemiologia , Filogenia , Sudão/epidemiologia
14.
Nat Commun ; 12(1): 542, 2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33483491

RESUMO

There is need for effective and affordable vaccines against SARS-CoV-2 to tackle the ongoing pandemic. In this study, we describe a protein nanoparticle vaccine against SARS-CoV-2. The vaccine is based on the display of coronavirus spike glycoprotein receptor-binding domain (RBD) on a synthetic virus-like particle (VLP) platform, SpyCatcher003-mi3, using SpyTag/SpyCatcher technology. Low doses of RBD-SpyVLP in a prime-boost regimen induce a strong neutralising antibody response in mice and pigs that is superior to convalescent human sera. We evaluate antibody quality using ACE2 blocking and neutralisation of cell infection by pseudovirus or wild-type SARS-CoV-2. Using competition assays with a monoclonal antibody panel, we show that RBD-SpyVLP induces a polyclonal antibody response that recognises key epitopes on the RBD, reducing the likelihood of selecting neutralisation-escape mutants. Moreover, RBD-SpyVLP is thermostable and can be lyophilised without losing immunogenicity, to facilitate global distribution and reduce cold-chain dependence. The data suggests that RBD-SpyVLP provides strong potential to address clinical and logistic challenges of the COVID-19 pandemic.


Assuntos
Anticorpos Antivirais/imunologia , Vacinas contra COVID-19/imunologia , COVID-19/prevenção & controle , Peptídeos/imunologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Enzima de Conversão de Angiotensina 2/imunologia , Animais , Anticorpos Bloqueadores/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , COVID-19/imunologia , Linhagem Celular , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Suínos
15.
Viruses ; 12(1)2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31936607

RESUMO

The risk of emergence and/or re-emergence of arthropod-borne viral (arboviral) infections is rapidly growing worldwide, particularly in Africa. The burden of arboviral infections and diseases is not well scrutinized because of the inefficient surveillance systems in endemic countries. Furthermore, the health systems are fully occupied by the burden of other co-existing febrile illnesses, especially malaria. In this review we summarize the epidemiology and risk factors associated with the major human arboviral diseases and highlight the gap in knowledge, research, and control in Sudan. Published data in English up to March 2019 were reviewed and are discussed to identify the risks and challenges for the control of arboviruses in the country. In addition, the lack of suitable diagnostic tools such as viral genome sequencing, and the urgent need for establishing a genomic database of the circulating viruses and potential sources of entry are discussed. Moreover, the research and healthcare gaps and global health threats are analyzed, and suggestions for developing strategic health policy for the prevention and control of arboviruses with focus on building the local diagnostic and research capacity and establishing an early warning surveillance system for the early detection and containment of arboviral epidemics are offered.


Assuntos
Infecções por Arbovirus/epidemiologia , Infecções por Arbovirus/prevenção & controle , Vetores Artrópodes/virologia , Epidemias/prevenção & controle , Animais , Arbovírus/genética , Pesquisa Biomédica , Controle de Doenças Transmissíveis , Genoma Viral , Humanos , Fatores de Risco , Sudão/epidemiologia
16.
mSphere ; 5(2)2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32269152

RESUMO

Arboviruses are pathogens of humans and animals. A better understanding of the interactions between these pathogens and the arthropod vectors, such as mosquitoes, that transmit them is necessary to develop novel control measures. A major antiviral pathway in the mosquito vector is the exogenous small interfering RNA (exo-siRNA) pathway, which is induced by arbovirus-derived double-stranded RNA in infected cells. Although recent work has shown the key role played by Argonaute-2 (Ago-2) and Dicer-2 (Dcr-2) in this pathway, the regulatory mechanisms that govern these pathways have not been studied in mosquitoes. Here, we show that the Domino ortholog p400 has antiviral activity against the alphavirus Semliki Forest virus (Togaviridae) both in Aedes aegypti-derived cells and in vivo Antiviral activity of p400 was also demonstrated against chikungunya virus (Togaviridae) and Bunyamwera virus (Peribunyaviridae) but not Zika virus (Flaviviridae). p400 was found to be expressed across mosquito tissues and regulated ago-2 but not dcr-2 transcript levels in A. aegypti mosquitoes. These findings provide novel insights into the regulation of an important aedine exo-siRNA pathway effector protein, Ago-2, by the Domino ortholog p400. They add functional insights to previous observations of this protein's antiviral and RNA interference regulatory activities in Drosophila melanogasterIMPORTANCE Female Aedes aegypti mosquitoes are vectors of human-infecting arthropod-borne viruses (arboviruses). In recent decades, the incidence of arthropod-borne viral infections has grown dramatically. Vector competence is influenced by many factors, including the mosquito's antiviral defenses. The exogenous small interfering RNA (siRNA) pathway is a major antiviral response restricting arboviruses in mosquitoes. While the roles of the effectors of this pathway, Argonaute-2 and Dicer-2 are well characterized, nothing is known about its regulation in mosquitoes. In this study, we demonstrate that A. aegypti p400, whose ortholog Domino in Drosophila melanogaster is a chromatin-remodeling ATPase member of the Tip60 complex, regulates siRNA pathway activity and controls ago-2 expression levels. In addition, we found p400 to have antiviral activity against different arboviruses. Therefore, our study provides new insights into the regulation of the antiviral response in A. aegypti mosquitoes.


Assuntos
Aedes/genética , Proteínas Argonautas/genética , Proteínas de Insetos/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Aedes/virologia , Animais , Arbovírus/fisiologia , Feminino , Regulação da Expressão Gênica , Mosquitos Vetores/genética , Mosquitos Vetores/virologia
17.
Viruses ; 12(4)2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32295109

RESUMO

The inland floodwater mosquito Aedes vexans (Meigen, 1830) is a competent vector of numerous arthropod-borne viruses such as Rift Valley fever virus (Phenuiviridae) and Zika virus (Flaviviridae). Aedes vexans spp. have widespread Afrotropical distribution and are common European cosmopolitan mosquitoes. We examined the virome of Ae. vexans arabiensis samples from Barkédji village, Senegal, with small RNA sequencing, bioinformatic analysis, and RT-PCR screening. We identified a novel 9494 nt iflavirus (Picornaviridae) designated here as Aedes vexans iflavirus (AvIFV). Annotation of the AvIFV genome reveals a 2782 amino acid polyprotein with iflavirus protein domain architecture and typical iflavirus 5' internal ribosomal entry site and 3' poly-A tail. Aedes vexans iflavirus is most closely related to a partial virus sequence from Venturia canescens (a parasitoid wasp) with 56.77% pairwise amino acid identity. Analysis of AvIFV-derived small RNAs suggests that AvIFV is targeted by the exogenous RNA interference pathway but not the PIWI-interacting RNA response, as ~60% of AvIFV reads corresponded to 21 nt Dicer-2 virus-derived small RNAs and the 24-29 nt AvIFV read population did not exhibit a "ping-pong" signature. The RT-PCR screens of archival and current (circa 2011-2020) Ae. vexans arabiensis laboratory samples and wild-caught mosquitoes from Barkédji suggest that AvIFV is ubiquitous in these mosquitoes. Further, we screened wild-caught European Ae. vexans samples from Germany, the United Kingdom, Italy, and Sweden, all of which tested negative for AvIFV RNA. This report provides insight into the diversity of commensal Aedes viruses and the host RNAi response towards iflaviruses.


Assuntos
Aedes/virologia , Picornaviridae/genética , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Viral , Animais , Biologia Computacional/métodos , Genoma Viral , Anotação de Sequência Molecular , Mosquitos Vetores/virologia , Análise de Sequência de RNA
18.
NPJ Vaccines ; 5(1): 69, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32793398

RESUMO

Clinical development of the COVID-19 vaccine candidate ChAdOx1 nCoV-19, a replication-deficient simian adenoviral vector expressing the full-length SARS-CoV-2 spike (S) protein was initiated in April 2020 following non-human primate studies using a single immunisation. Here, we compared the immunogenicity of one or two doses of ChAdOx1 nCoV-19 in both mice and pigs. Whilst a single dose induced antigen-specific antibody and T cells responses, a booster immunisation enhanced antibody responses, particularly in pigs, with a significant increase in SARS-CoV-2 neutralising titres.

19.
Viruses ; 10(2)2018 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-29463033

RESUMO

Mosquitoes transmit several human- and animal-pathogenic alphaviruses (Togaviridae family). In alphavirus-infected mosquito cells two different types of virus-specific small RNAs are produced as part of the RNA interference response: short-interfering (si)RNAs and PIWI-interacting (pi)RNAs. The siRNA pathway is generally thought to be the main antiviral pathway. Although an antiviral activity has been suggested for the piRNA pathway its role in host defences is not clear. Knock down of key proteins of the piRNA pathway (Ago3 and Piwi5) in Aedesaegypti-derived cells reduced the production of alphavirus chikungunya virus (CHIKV)-specific piRNAs but had no effect on virus replication. In contrast, knock down of the siRNA pathway key protein Ago2 resulted in an increase in virus replication. Similar results were obtained when expression of Piwi4 was silenced. Knock down of the helicase Spindle-E (SpnE), an essential co-factor of the piRNA pathway in Drosophila melanogaster, resulted in increased virus replication indicating that SpnE acts as an antiviral against alphaviruses such as CHIKV and the related Semliki Forest virus (SFV). Surprisingly, this effect was found to be independent of the siRNA and piRNA pathways in Ae. aegypti cells and specific for alphaviruses. This suggests a small RNA-independent antiviral function for this protein in mosquitoes.


Assuntos
Infecções por Alphavirus/virologia , Alphavirus , Culicidae/virologia , Animais , Células Cultivadas , Proteínas de Insetos/genética , Interferência de RNA , Pequeno RNA não Traduzido/genética , Replicação Viral
20.
Elife ; 62017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28960178

RESUMO

Most vertebrate and plant RNA and small DNA viruses suppress genomic CpG and UpA dinucleotide frequencies, apparently mimicking host mRNA composition. Artificially increasing CpG/UpA dinucleotides attenuates viruses through an entirely unknown mechanism. Using the echovirus 7 (E7) model in several cell types, we show that the restriction in E7 replication in mutants with increased CpG/UpA dinucleotides occurred immediately after viral entry, with incoming virions failing to form replication complexes. Sequences of CpG/UpA-high virus stocks showed no evidence of increased mutational errors that would render them replication defective, these viral RNAs were not differentially sequestered in cytoplasmic stress granules nor did they induce a systemic antiviral state. Importantly, restriction was not mediated through effects on translation efficiency since replicons with high CpG/UpA sequences inserted into a non-coding region were similarly replication defective. Host-cells thus possess intrinsic defence pathways that prevent replication of viruses with increased CpG/UpA frequencies independently of codon usage.


Assuntos
Enterovirus Humano B/genética , Enterovirus Humano B/fisiologia , Interações Hospedeiro-Patógeno , Nucleotídeos/genética , RNA Viral/genética , Replicação Viral , Animais , Linhagem Celular , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA