Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Nat Microbiol ; 9(6): 1417-1426, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38783022

RESUMO

Ebola virus and other orthoebolaviruses cause severe haemorrhagic fevers in humans, with very high case fatality rates. Their non-segmented single-stranded RNA genome encodes only seven structural proteins and a small number of non-structural proteins to facilitate the virus life cycle. The basics of this life cycle are well established, but recent advances have substantially increased our understanding of its molecular details, including the viral and host factors involved. Here we provide a comprehensive overview of our current knowledge of the molecular details of the orthoebolavirus life cycle, with a special focus on proviral host factors. We discuss the multistep entry process, viral RNA synthesis in specialized phase-separated intracellular compartments called inclusion bodies, the expression of viral proteins and ultimately the assembly of new virus particles and their release at the cell surface. In doing so, we integrate recent studies into the increasingly detailed model that has developed for these fundamental aspects of orthoebolavirus biology.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Animais , Humanos , Ebolavirus/genética , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/fisiologia , Genoma Viral , Doença pelo Vírus Ebola/virologia , Interações Hospedeiro-Patógeno , RNA Viral/metabolismo , RNA Viral/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Vírion/metabolismo , Vírion/genética , Montagem de Vírus , Internalização do Vírus , Replicação Viral
2.
Nature ; 622(7983): 603-610, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37699521

RESUMO

Non-segmented negative-strand RNA viruses, including Ebola virus (EBOV), rabies virus, human respiratory syncytial virus and pneumoviruses, can cause respiratory infections, haemorrhagic fever and encephalitis in humans and animals, and are considered a substantial health and economic burden worldwide1. Replication and transcription of the viral genome are executed by the large (L) polymerase, which is a promising target for the development of antiviral drugs. Here, using the L polymerase of EBOV as a representative, we show that de novo replication of L polymerase is controlled by the specific 3' leader sequence of the EBOV genome in an enzymatic assay, and that formation of at least three base pairs can effectively drive the elongation process of RNA synthesis independent of the specific RNA sequence. We present the high-resolution structures of the EBOV L-VP35-RNA complex and show that the 3' leader RNA binds in the template entry channel with a distinctive stable bend conformation. Using mutagenesis assays, we confirm that the bend conformation of the RNA is required for the de novo replication activity and reveal the key residues of the L protein that stabilize the RNA conformation. These findings provide a new mechanistic understanding of RNA synthesis for polymerases of non-segmented negative-strand RNA viruses, and reveal important targets for the development of antiviral drugs.


Assuntos
Ebolavirus , RNA Viral , RNA Polimerase Dependente de RNA , Replicação Viral , Animais , Humanos , Antivirais/farmacologia , Ebolavirus/enzimologia , Ebolavirus/genética , Ebolavirus/crescimento & desenvolvimento , Doença pelo Vírus Ebola/virologia , RNA Viral/biossíntese , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/metabolismo , Genoma Viral , Conformação de Ácido Nucleico , Mutagênese , Estabilidade de RNA
3.
Emerg. infect. dis. (Online) ; 28: 2583-2585, dez 12, 2022. mapa, tab
Artigo em Inglês | RDSM | ID: biblio-1532401

RESUMO

We detected Bombali ebolavirus RNA in 3 free-tailed bats (Mops condylurus, Molossidae) in Mozambique. Sequencing of the large protein gene revealed 98% identity with viruses previously detected in Sierra Leone, Kenya, and Guinea. Our findings further support the suspected role of Mops condylurus bats in maintaining Bombali ebolavirus


Assuntos
Humanos , Animais , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/genética , Quirópteros , Ebolavirus/isolamento & purificação , Moçambique/epidemiologia
4.
J Virol ; 96(17): e0108322, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35993739

RESUMO

Ebola virus (EBOV) VP30 regulates viral genome transcription and replication by switching its phosphorylation status. However, the importance of VP30 phosphorylation and dephosphorylation in other viral replication processes such as nucleocapsid and virion assembly is unclear. Interestingly, VP30 is predominantly dephosphorylated by cellular phosphatases in viral inclusions, while it is phosphorylated in the released virions. Thus, uncertainties regarding how VP30 phosphorylation in nucleocapsids is achieved and whether VP30 phosphorylation provides any advantages in later steps in viral replication have arisen. In the present study, to characterize the roles of VP30 phosphorylation in nucleocapsid formation, we used electron microscopic analyses and live cell imaging systems. We identified VP30 localized to the surface of protrusions surrounding nucleoprotein (NP)-forming helical structures in the nucleocapsid, suggesting the involvement in assembly and transport of nucleocapsids. Interestingly, VP30 phosphorylation facilitated its association with nucleocapsid-like structures (NCLSs). On the contrary, VP30 phosphorylation does not influence the transport characteristics and NCLS number leaving from and coming back into viral inclusions, indicating that the phosphorylation status of VP30 is not a prerequisite for NCLS departure. Moreover, the phosphorylation status of VP30 did not cause major differences in nucleocapsid transport in authentic EBOV-infected cells. In the following budding step, the association of VP30 and its phosphorylation status did not influence the budding efficiency of virus-like particles. Taken together, it is plausible that EBOV may utilize the phosphorylation of VP30 for its selective association with nucleocapsids, without affecting nucleocapsid transport and virion budding processes. IMPORTANCE Ebola virus (EBOV) causes severe fevers with unusually high case fatality rates. The nucleocapsid provides the template for viral genome transcription and replication. Thus, understanding the regulatory mechanism behind its formation is important for the development of novel therapeutic approaches. Previously, we established a live-cell imaging system based on the ectopic expression of viral fluorescent fusion proteins, allowing the visualization and characterization of intracytoplasmic transport of nucleocapsid-like structures. EBOV VP30 is an essential transcriptional factor for viral genome synthesis, and, although its role in viral genome transcription and replication is well understood, the functional importance of VP30 phosphorylation in assembly of nucleocapsids is still unclear. Our work determines the localization of VP30 at the surface of ruffled nucleocapsids, which differs from the localization of polymerase in EBOV-infected cells. This study sheds light on the novel role of VP30 phosphorylation in nucleocapsid assembly, which is an important prerequisite for virion formation.


Assuntos
Ebolavirus , Nucleocapsídeo , Fatores de Transcrição , Proteínas Virais , Montagem de Vírus , Transporte Biológico , Ebolavirus/química , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/metabolismo , Doença pelo Vírus Ebola/virologia , Humanos , Nucleocapsídeo/biossíntese , Nucleocapsídeo/metabolismo , Fosforilação , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Vírion/química , Vírion/crescimento & desenvolvimento , Vírion/metabolismo
5.
Cell Rep ; 35(2): 108984, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852862

RESUMO

Antibodies that target the glycan cap epitope on the ebolavirus glycoprotein (GP) are common in the adaptive response of survivors. A subset is known to be broadly neutralizing, but the details of their epitopes and basis for neutralization are not well understood. Here, we present cryoelectron microscopy (cryo-EM) structures of diverse glycan cap antibodies that variably synergize with GP base-binding antibodies. These structures describe a conserved site of vulnerability that anchors the mucin-like domains (MLDs) to the glycan cap, which we call the MLD anchor and cradle. Antibodies that bind to the MLD cradle share common features, including use of IGHV1-69 and IGHJ6 germline genes, which exploit hydrophobic residues and form ß-hairpin structures to mimic the MLD anchor, disrupt MLD attachment, destabilize GP quaternary structure, and block cleavage events required for receptor binding. Our results provide a molecular basis for ebolavirus neutralization by broadly reactive glycan cap antibodies.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Proteínas do Envelope Viral/química , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/química , Anticorpos Antivirais/metabolismo , Especificidade de Anticorpos , Sítios de Ligação , Microscopia Crioeletrônica , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/imunologia , Ebolavirus/patogenicidade , Epitopos/química , Epitopos/imunologia , Feminino , Células HEK293 , Células HeLa , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/patologia , Doença pelo Vírus Ebola/virologia , Humanos , Células Jurkat , Camundongos , Modelos Moleculares , Polissacarídeos/química , Polissacarídeos/imunologia , Ligação Proteica , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
6.
Clin Microbiol Rev ; 34(1)2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33055231

RESUMO

Patients and physicians worldwide are facing tremendous health care hazards that are caused by the ongoing severe acute respiratory distress syndrome coronavirus 2 (SARS-CoV-2) pandemic. Remdesivir (GS-5734) is the first approved treatment for severe coronavirus disease 2019 (COVID-19). It is a novel nucleoside analog with a broad antiviral activity spectrum among RNA viruses, including ebolavirus (EBOV) and the respiratory pathogens Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV, and SARS-CoV-2. First described in 2016, the drug was derived from an antiviral library of small molecules intended to target emerging pathogenic RNA viruses. In vivo, remdesivir showed therapeutic and prophylactic effects in animal models of EBOV, MERS-CoV, SARS-CoV, and SARS-CoV-2 infection. However, the substance failed in a clinical trial on ebolavirus disease (EVD), where it was inferior to investigational monoclonal antibodies in an interim analysis. As there was no placebo control in this study, no conclusions on its efficacy in EVD can be made. In contrast, data from a placebo-controlled trial show beneficial effects for patients with COVID-19. Remdesivir reduces the time to recovery of hospitalized patients who require supplemental oxygen and may have a positive impact on mortality outcomes while having a favorable safety profile. Although this is an important milestone in the fight against COVID-19, approval of this drug will not be sufficient to solve the public health issues caused by the ongoing pandemic. Further scientific efforts are needed to evaluate the full potential of nucleoside analogs as treatment or prophylaxis of viral respiratory infections and to develop effective antivirals that are orally bioavailable.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Antivirais/farmacologia , Infecções por Coronavirus/tratamento farmacológico , Doença pelo Vírus Ebola/tratamento farmacológico , Pneumonia Viral/tratamento farmacológico , Síndrome Respiratória Aguda Grave/tratamento farmacológico , Monofosfato de Adenosina/farmacocinética , Monofosfato de Adenosina/farmacologia , Alanina/farmacocinética , Alanina/farmacologia , Antivirais/farmacocinética , Betacoronavirus/efeitos dos fármacos , Betacoronavirus/crescimento & desenvolvimento , Betacoronavirus/patogenicidade , COVID-19 , Ensaios Clínicos como Assunto , Ensaios de Uso Compassivo/métodos , Infecções por Coronavirus/mortalidade , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Esquema de Medicação , Ebolavirus/efeitos dos fármacos , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/mortalidade , Doença pelo Vírus Ebola/patologia , Doença pelo Vírus Ebola/virologia , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/crescimento & desenvolvimento , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , Pandemias , Segurança do Paciente , Pneumonia Viral/mortalidade , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/crescimento & desenvolvimento , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/mortalidade , Síndrome Respiratória Aguda Grave/patologia , Síndrome Respiratória Aguda Grave/virologia , Análise de Sobrevida , Resultado do Tratamento
7.
J Cell Mol Med ; 24(12): 6988-6999, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32374474

RESUMO

Outbreaks of infections with viruses like Sars-CoV-2, Ebola virus and Zika virus lead to major global health and economic problems because of limited treatment options. Therefore, new antiviral drug candidates are urgently needed. The promising new antiviral drug candidate silvestrol effectively inhibited replication of Corona-, Ebola-, Zika-, Picorna-, Hepatis E and Chikungunya viruses. Besides a direct impact on pathogens, modulation of the host immune system provides an additional facet to antiviral drug development because suitable immune modulation can boost innate defence mechanisms against the pathogens. In the present study, silvestrol down-regulated several pro- and anti-inflammatory cytokines (IL-6, IL-8, IL-10, CCL2, CCL18) and increased TNF-α during differentiation and activation of M1-macrophages, suggesting that the effects of silvestrol might cancel each other out. However, silvestrol amplified the anti-inflammatory potential of M2-macrophages by increasing expression of anti-inflammatory surface markers CD206, TREM2 and reducing release of pro-inflammatory IL-8 and CCL2. The differentiation of dendritic cells in the presence of silvestrol is characterized by down-regulation of several surface markers and cytokines indicating that differentiation is impaired by silvestrol. In conclusion, silvestrol influences the inflammatory status of immune cells depending on the cell type and activation status.


Assuntos
Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Citocinas/genética , Células Dendríticas/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Macrófagos/efeitos dos fármacos , Triterpenos/farmacologia , Betacoronavirus/crescimento & desenvolvimento , Betacoronavirus/imunologia , Diferenciação Celular/efeitos dos fármacos , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/crescimento & desenvolvimento , Vírus Chikungunya/imunologia , Citocinas/classificação , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Ebolavirus/efeitos dos fármacos , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Vírus da Hepatite E/efeitos dos fármacos , Vírus da Hepatite E/crescimento & desenvolvimento , Vírus da Hepatite E/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/virologia , Especificidade de Órgãos , Picornaviridae/efeitos dos fármacos , Picornaviridae/crescimento & desenvolvimento , Picornaviridae/imunologia , Cultura Primária de Células , SARS-CoV-2 , Transdução de Sinais , Zika virus/efeitos dos fármacos , Zika virus/crescimento & desenvolvimento , Zika virus/imunologia
8.
BMC Res Notes ; 12(1): 639, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31570108

RESUMO

OBJECTIVE: Survivors of Ebola virus disease (EVD) are at risk of developing blinding intraocular inflammation-or uveitis-which is associated with retinal pigment epithelial (RPE) scarring and persistence of live Zaire ebolavirus (EBOV) within the eye. As part of a large research project aimed at defining the human RPE cell response to being infected with EBOV, this work focused on the microRNAs (miRNAs) associated with the infection. RESULTS: Using RNA-sequencing, we detected 13 highly induced and 2 highly repressed human miRNAs in human ARPE-19 RPE cells infected with EBOV, including hsa-miR-1307-5p, hsa-miR-29b-3p and hsa-miR-33a-5p (up-regulated), and hsa-miR-3074-3p and hsa-miR-27b-5p (down-regulated). EBOV-miR-1-5p was also found in infected RPE cells. Through computational identification of putative miRNA targets, we predicted a broad range of regulatory activities, including effects on innate and adaptive immune responses, cellular metabolism, cell cycle progression, apoptosis and autophagy. The most highly-connected molecule in the miR-target network was leucine-rich repeat kinase 2, which is involved in neuroinflammation and lysosomal processing. Our findings should stimulate new studies on the impact of miRNA changes in EBOV-infected RPE cells to further understanding of intraocular viral persistence and the pathogenesis of uveitis in EVD survivors.


Assuntos
Ebolavirus/genética , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Interações Hospedeiro-Patógeno/genética , MicroRNAs/genética , Imunidade Adaptativa/genética , Apoptose/genética , Autofagia/genética , Ciclo Celular/genética , Linhagem Celular , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/patogenicidade , Células Epiteliais/imunologia , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/genética , MicroRNAs/classificação , MicroRNAs/imunologia , Pigmentos da Retina , Transdução de Sinais
9.
Artigo em Inglês | MEDLINE | ID: mdl-31307979

RESUMO

Quinacrine hydrochloride is a small-molecule, orally bioavailable drug that has been used clinically as an antimalarial and for many other applications. A machine learning model trained on Ebola virus (EBOV) screening data identified quinacrine as a potent (nanomolar) in vitro inhibitor. In the current study, quinacrine 25 mg/kg was shown to protect 70% of mice (statistically significant) from a lethal challenge with mouse-adapted EBOV with once-daily intraperitoneal dosing for 8 days.


Assuntos
Antimaláricos/farmacologia , Antivirais/farmacologia , Reposicionamento de Medicamentos , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Quinacrina/farmacologia , Animais , Células CACO-2 , Chlorocebus aethiops , Modelos Animais de Doenças , Ebolavirus/crescimento & desenvolvimento , Células HeLa , Doença pelo Vírus Ebola/mortalidade , Doença pelo Vírus Ebola/patologia , Doença pelo Vírus Ebola/virologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Análise de Sobrevida , Tilorona/farmacologia , Células Vero , Carga Viral/efeitos dos fármacos
10.
Nat Rev Microbiol ; 17(10): 593-606, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31341272

RESUMO

Following the Ebola virus disease epidemic in west Africa, there has been increased awareness of the need for improved therapies for emerging diseases, including viral haemorrhagic fevers such as those caused by Ebola virus and other filoviruses. Our continually improving understanding of the virus life cycle coupled with the increased availability of 'omics' analyses and high-throughput screening technologies has enhanced our ability to identify potential viral and host factors and aspects involved in the infection process that might be intervention targets. In this Review we address compounds that have shown promise to various degrees in interfering with the filovirus life cycle, including monoclonal antibodies such as ZMapp, mAb114 and REGN-EB3 and inhibitors of viral RNA synthesis such as remdesivir and TKM-Ebola. Furthermore, we discuss the general potential of targeting aspects of the virus life cycle such as the entry process, viral RNA synthesis and gene expression, as well as morphogenesis and budding.


Assuntos
Antivirais/isolamento & purificação , Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Ebolavirus/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos
11.
Virology ; 532: 22-29, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30999160

RESUMO

Priming of the viral glycoprotein (GP) by the cellular proteases cathepsin B and L (CatB, CatL) is believed to be essential for cell entry of filoviruses. However, pseudotyping systems that predominantly produce non-filamentous particles have frequently been used to prove this concept. Here, we report that GP-mediated entry of retroviral-, rhabdoviral and filoviral particles depends on CatB/CatL activity and that this effect is cell line-independent. Moreover, we show that the human cell line Calu-3, which expresses low amounts of CatL, is largely resistant to entry driven by diverse filovirus GPs. Finally, we demonstrate that Calu-3 cell entry mediated by certain filovirus GPs can be rescued upon directed expression of CatL or DC-SIGN. Our results identify Calu-3 cells as largely resistant to filovirus GP-driven entry and demonstrate that entry is limited at the stage of virion attachment and GP priming.


Assuntos
Catepsina L/genética , Moléculas de Adesão Celular/genética , Ebolavirus/genética , Células Epiteliais/imunologia , Lectinas Tipo C/genética , Receptores de Superfície Celular/genética , Proteínas Virais/genética , Células A549 , Animais , Catepsina B/antagonistas & inibidores , Catepsina B/genética , Catepsina B/imunologia , Catepsina B/metabolismo , Catepsina L/antagonistas & inibidores , Catepsina L/imunologia , Catepsina L/metabolismo , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Regulação da Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Humanos , Lectinas Tipo C/antagonistas & inibidores , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Leucina/análogos & derivados , Leucina/farmacologia , Marburgvirus/genética , Marburgvirus/crescimento & desenvolvimento , Marburgvirus/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Células Vero , Vesiculovirus/genética , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/metabolismo , Proteínas Virais/metabolismo , Vírion/genética , Vírion/crescimento & desenvolvimento , Vírion/metabolismo , Internalização do Vírus/efeitos dos fármacos
12.
Epidemics ; 26: 128-133, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30880169

RESUMO

On August 1, 2018, the Democratic Republic of Congo declared its 10th and largest outbreak of Ebola inflicting North Khivu and Ituri provinces. The spread of Ebola to Congolese urban centers along with deliberate attacks on the health care workers has hindered epidemiological surveillance activities, leading to substantial reporting delays. Reporting delays distort the epidemic incidence pattern misrepresenting estimates of epidemic potential and the outbreak trajectory. To assess the impact of reporting delays, we conducted a real-time analysis of the dynamics of the ongoing Ebola outbreak in the DRC using epidemiological data retrieved from the World Health Organization Situation Reports and Disease Outbreak News. We analyzed temporal trends in reporting delays, epidemic curves of crude and reporting-delay adjusted incidences and changes in the effective reproduction number, Rt. As of January 15, 2019, 663 Ebola cases have been reported in the Democratic Republic of Congo. The average reporting delay exhibited 81.1% decline from a mean of 17.4 weeks (95% CI 13-24.1) in May, 2018 to 3.3 weeks (95% CI 2.7-4.2) in September, 2018 (F-test statistic = 44.9, p = 0.0067). The Ebola epidemic has shown a two-wave pattern with the first surge in cases occurring between July 30 and August 13, 2018 and the second on September 24, 2018. During the last 4 generation intervals, the trend in the mean Rt has exhibited a slight decline (rho = -0.37, p < 0.001), fluctuating around 0.9 (range: 0-1.8). Our most recent estimate of R is at 0.9 (95% CI: 0.4, 1.1) during the last generation interval. Our most recent analysis of the Ebola outbreak in DRC indicates that the Ebola virus still active although transmission is characterized by a low fluctuating reproduction number. Yet, this pattern does not imply that the epidemic can be easily controlled particularly in the context of unstable epidemiological surveillance efforts hindered by unpredictable local violence.


Assuntos
Ebolavirus/crescimento & desenvolvimento , Epidemias/estatística & dados numéricos , Doença pelo Vírus Ebola/epidemiologia , República Democrática do Congo/epidemiologia , Feminino , Humanos , Tempo
13.
Viruses ; 10(11)2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30453499

RESUMO

The filoviruses Ebola virus (EBOV) and Sudan virus (SUDV) can cause severe diseases, and there are currently no licensed countermeasures available for use against them. Transmission occurs frequently via contact with bodily fluids from infected individuals. However, it can be difficult to determine when or how someone became infected, or the quantity of infectious virus to which they were exposed. Evidence suggests the infectious dose is low, but the majority of published studies use high exposure doses. This study characterized the outcome of exposure to a low dose of EBOV or SUDV, using a Macaca fascicularis model. Further, because the effect of virus passage in cell culture may be more pronounced when lower exposure doses are used, viruses that possessed either the characteristics of wild type viruses (possessing predominantly 7-uridine (7U) genotype and a high particle-to-plaque forming unit (PFU) ratio) or cell culture-passaged viruses (predominantly 8-uridine (8U) genotype, a lower particle-to-PFU ratio) were used. The time to death after a low dose exposure was delayed in comparison to higher exposure doses. These data demonstrated that an extremely low dose of EBOV or SUDV is sufficient to cause lethal disease. A low dose exposure model can help inform studies on pathogenesis, transmission, and optimization of prevention strategies.


Assuntos
Modelos Animais de Doenças , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/patologia , Animais , Injeções Intramusculares , Macaca fascicularis , Análise de Sobrevida
14.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30021890

RESUMO

The Zaire ebolavirus (EBOV) glycoprotein (GP) is cleaved into two subunits (GP1 and GP2) that are both required for virus attachment and entry into cells. Sequence changes in the GP have been proposed to increase pathogenesis and to alter virus growth properties. Mutations in GP acquired during EBOV tissue culture passage have also been reported to change virus growth properties. Here, we report the isolation of six amino acid mutations in EBOV GP that spontaneously appeared during recovery and passage of an EBOV-Makona GP-pseudotyped vesicular stomatitis virus (VSV), two of which also occur during passage of EBOV clinical isolates in tissue culture. Each of the six mutations resulted in increased virus growth in monkey and human cell lines. All mutations are located in the GP2 fusion subunit and increase entry kinetics of EBOV virus-like particles (VLPs). The gain-of-entry function mapped to two mechanistic phenotypes. Mutations in heptad repeat 1 (HR1) decreased the requirement for cathepsin B activity for viral infection. Mutations directly within the fusion loop increased entry kinetics without altering the cathepsin B dependence. Several mutations in the fusion loop were substitutions of residues present in other ebolavirus glycoproteins, illustrating the evolutionary paths for maintaining an optimally functioning fusion loop under selection pressure.IMPORTANCEZaire ebolavirus (EBOV) is the causative agent of the highly lethal Ebola virus disease and poses a significant threat to the global health community. Approved antivirals against EBOV are lacking; however, promising therapies targeting the EBOV glycoprotein are being developed. Efficacy testing of these candidate therapeutics relies on EBOV laboratory stocks, which when grown in tissue culture may acquire mutations in the glycoprotein. These mutations can produce inaccurate results in therapeutic testing. Until recently, distinguishing between tissue culture mutations and naturally occurring polymorphisms in EBOV GP was difficult in the absence of consensus clinical GP sequences. Here, we utilize recombinant VSV (rVSV) pseudotyped with the consensus clinical EBOV Makona GP to identify several mutations that have emerged or have potential to emerge in EBOV GP during tissue culture passage. Identifying these mutations informs the EBOV research community as to which mutations may arise during preparation of laboratory virus stocks.


Assuntos
Catepsina B/metabolismo , Ebolavirus , Mutação , Proteínas do Envelope Viral , Internalização do Vírus , Animais , Catepsina B/genética , Chlorocebus aethiops , Ebolavirus/genética , Ebolavirus/crescimento & desenvolvimento , Humanos , Estrutura Secundária de Proteína , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
15.
Curr Med Chem ; 25(38): 5177-5190, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29032747

RESUMO

Ebola virus has caused 26 outbreaks in 10 different countries since its identification in 1976, making it one of the deadliest emerging viral pathogens. The most recent outbreak in West Africa from 2014-16 was the deadliest yet and culminated in 11,310 deaths out of 28,616 confirmed cases. Currently, there are no FDA-approved therapeutics or vaccines to treat Ebola virus infections. The slow development of effective vaccines combined with the severity of past outbreaks emphasizes the need to accelerate research into understanding the virus lifecycle and the development of therapeutics for post exposure treatment. Here we present a summary of the major findings on the Ebola virus replication cycle and the therapeutic approaches explored to treat this devastating disease. The major focus of this review is on small molecule inhibitors.


Assuntos
Antivirais/uso terapêutico , Desenvolvimento de Medicamentos/métodos , Ebolavirus , Doença pelo Vírus Ebola/tratamento farmacológico , Animais , Ebolavirus/efeitos dos fármacos , Ebolavirus/genética , Ebolavirus/crescimento & desenvolvimento , Doença pelo Vírus Ebola/etiologia , Humanos , Transcrição Gênica/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
16.
Curr Top Microbiol Immunol ; 419: 113-150, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28710692

RESUMO

Ebola virus (EBOV) is a highly pathogenic emerging virus that represents a serious threat to global public health and a major priority for biodefense. The 2014 West African outbreak demonstrated the potential of EBOV to cause an epidemic affecting thousands of people. The severity of disease and high case fatality rate of EBOV is largely due to the host response elicited by the virus. EBOV infection hijacks a number of host pathways to carry out replication and stimulate potent inflammatory responses, while simultaneously subverting the host antiviral immune response. Together, these events trigger a complex, systemic, often lethal febrile disease characterized by high levels of inflammatory cytokines, acute hepatitis and liver dysfunction, immune antagonism, gastrointestinal distress, and, in some cases, hemorrhage caused by coagulopathy and vascular leakage. This review presents current knowledge about the particular host responses induced and disrupted by EBOV infection and how these contribute to virus replication, immune evasion, pathogenesis, and disease outcome.


Assuntos
Ebolavirus/crescimento & desenvolvimento , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/virologia , Interações Hospedeiro-Patógeno/fisiologia , Replicação Viral , Citocinas/metabolismo , Ebolavirus/imunologia , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/patologia , Humanos , Evasão da Resposta Imune
17.
J Infect Dis ; 217(1): 58-63, 2017 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-29087482

RESUMO

Both Ebola virus (EBOV) and Reston virus (RESTV) cause disease in nonhuman primates, yet only EBOV causes disease in humans. To investigate differences in viral pathogenicity, humanized mice (hu-NSG-SGM3) were inoculated with EBOV or RESTV. Consistent with differences in disease in human infection, pronounced weight loss and markers of hepatic damage and disease were observed exclusively in EBOV-infected mice. These abnormalities were associated with significantly higher EBOV replication in the liver but not in the spleen, suggesting that in this model, efficiency of viral replication in select tissues early in infection may contribute to differences in viral pathogenicity.


Assuntos
Ebolavirus/crescimento & desenvolvimento , Doença pelo Vírus Ebola/virologia , Fígado/virologia , Replicação Viral , Animais , Peso Corporal , Modelos Animais de Doenças , Doença pelo Vírus Ebola/patologia , Humanos , Testes de Função Hepática , Camundongos , Camundongos SCID
18.
Curr Top Microbiol Immunol ; 411: 293-322, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28685291

RESUMO

This chapter describes the various strategies filoviruses use to escape host immune responses with a focus on innate immune and cell death pathways. Since filovirus replication can be efficiently blocked by interferon (IFN), filoviruses have evolved mechanisms to counteract both type I IFN induction and IFN response signaling pathways. Intriguingly, marburg- and ebolaviruses use different strategies to inhibit IFN signaling. This chapter also summarizes what is known about the role of IFN-stimulated genes (ISGs) in filovirus infection. These fall into three categories: those that restrict filovirus replication, those whose activation is inhibited by filoviruses, and those that have no measurable effect on viral replication. In addition to innate immunity, mammalian cells have evolved strategies to counter viral infections, including the induction of cell death and stress response pathways, and we summarize our current knowledge of how filoviruses interact with these pathways. Finally, this chapter delves into the interaction of EBOV with myeloid dendritic cells and macrophages and the associated inflammatory response, which differs dramatically between these cell types when they are infected with EBOV. In summary, we highlight the multifaceted nature of the host-viral interactions during filoviral infections.


Assuntos
Filoviridae/imunologia , Imunidade Inata/imunologia , Replicação Viral/imunologia , Animais , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/imunologia , Filoviridae/patogenicidade , Filoviridae/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Interferons/imunologia
19.
Methods Mol Biol ; 1628: 177-188, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28573619

RESUMO

Reverse genetics systems encompass a wide array of tools aimed at recapitulating some or all of the virus life cycle. In their most complete form, full-length clone systems allow us to use plasmid-encoded versions of the ribonucleoprotein (RNP) components to initiate the transcription and replication of a plasmid-encoded version of the complete viral genome, thereby initiating the complete virus life cycle and resulting in infectious virus. As such this approach is ideal for the generation of tailor-made recombinant filoviruses, which can be used to study virus biology. In addition, the generation of tagged and particularly fluorescent or luminescent viruses can be applied as tools for both diagnostic applications and for screening to identify novel countermeasures. Here we describe the generation and basic characterization of recombinant Ebola viruses rescued from cloned cDNA using a T7-driven system.


Assuntos
Ebolavirus/genética , Doença pelo Vírus Ebola/genética , Genética Reversa/métodos , Replicação Viral/genética , DNA Complementar/genética , Ebolavirus/crescimento & desenvolvimento , Genoma Viral , Doença pelo Vírus Ebola/virologia , Humanos
20.
Methods Mol Biol ; 1628: 203-210, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28573622

RESUMO

Assessment of small molecule compounds against filoviruses, such as Ebola virus, has identified numerous compounds that appear to have antiviral activity and should presumably be further investigated in animal efficacy trials. However, despite the many compounds that are purported to have good antiviral activity in in vitro studies, there are few instances where any efficacy has been reported in nonhuman primate models. Many of the high-throughput screening assays use reporter systems that only recapitulate a portion of the virus life cycle, while other assays only assess antiviral activity at relatively early time points. Moreover, many assays do not assess virus progeny production. A more in-depth evaluation of small numbers of test compounds is useful to economize resources and to generate higher quality antiviral hits. Assessing virus progeny production as late as 5 days post-infection allows for the elimination of compounds that have initial antiviral effects that are not sustained or where the virus rapidly develops resistance. While this eliminates many potential lead compounds that may be worthy of further structure-activity relationship (SAR) development, it also quickly excludes compounds that in their current form are unlikely to be effective in animal models. In addition, the inclusion of multiple assays that assess both cell viability and cell cytotoxicity, via different mechanisms, provides a more thorough assessment to exclude compounds that are not direct-acting antivirals.


Assuntos
Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Ensaios de Triagem em Larga Escala/métodos , Sobrevivência Celular/efeitos dos fármacos , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/virologia , Humanos , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA