Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Cell ; 185(6): 995-1007.e18, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35303429

RESUMO

Several ebolaviruses cause outbreaks of severe disease. Vaccines and monoclonal antibody cocktails are available to treat Ebola virus (EBOV) infections, but not Sudan virus (SUDV) or other ebolaviruses. Current cocktails contain antibodies that cross-react with the secreted soluble glycoprotein (sGP) that absorbs virus-neutralizing antibodies. By sorting memory B cells from EBOV infection survivors, we isolated two broadly reactive anti-GP monoclonal antibodies, 1C3 and 1C11, that potently neutralize, protect rodents from disease, and lack sGP cross-reactivity. Both antibodies recognize quaternary epitopes in trimeric ebolavirus GP. 1C11 bridges adjacent protomers via the fusion loop. 1C3 has a tripartite epitope in the center of the trimer apex. One 1C3 antigen-binding fragment anchors simultaneously to the three receptor-binding sites in the GP trimer, and separate 1C3 paratope regions interact differently with identical residues on the three protomers. A cocktail of both antibodies completely protected nonhuman primates from EBOV and SUDV infections, indicating their potential clinical value.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Ebolavirus , Doença pelo Vírus Ebola , Animais , Epitopos , Glicoproteínas/química , Subunidades Proteicas
2.
J Virol ; : e0124223, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39012096

RESUMO

Sudan ebolavirus (SUDV) is a member of the genus Ebolavirus (Family Filoviridae) and has caused sporadic outbreaks of Ebola disease (EBOD), or more specifically Sudan virus disease (SVD), with high mortality rates in Africa. Current vaccines and therapies that have been developed for filoviruses are almost all specific for Ebola virus (EBOV; of the species Zaire ebolavirus), and there is a current lack of therapeutics specific for SUDV. The recent SUDV outbreak in Uganda, which was distributed across multiple districts, including Kampala, a densely populated urban center, highlights the critical need for the development of novel SUDV-specific or pan-Ebola virus therapeutics. Previous work has characterized two monoclonal antibodies, FVM04 and CA45, which have neutralization capabilities against both EBOV and SUDV and have shown protective efficacy in animal challenge studies. Here, we expand upon this work, showing that treatment with a monoclonal antibody cocktail consisting of FVM04 and CA45 provides full protection against lethal SUDV infection in cynomolgus macaques. Studies that evaluate outcomes at late time points after infection, once clinical signs of illness are apparent, are vital for assessing the therapeutic efficacy of antibody therapeutics. We have shown that when treatment is initiated as late as 5 days after infection, with a second dose given on day 8, that treated groups showed few clinical signs or morbidity, with complete survival. This work provides further evidence that FVM04 and CA45 have strong therapeutic potential against SUDV and their development as a pan-Ebola virus therapeutic should be pursued. IMPORTANCE: There are currently no approved vaccines or therapeutics for Sudan virus, a filovirus which is highly related to Ebola virus and causes similar disease and outbreaks. In this study, a cocktail of two potent monoclonal antibodies that effectively neutralize Sudan virus was tested in a nonhuman primate model of Sudan virus disease. Treatment was highly effective, even when initiated as late as 5 days after infection, when clinical signs of infection were already evident. All treated animals showed complete recovery from infection, with little evidence of disease, while all animals that received a control treatment succumbed to infection within 8 days. The study further demonstrated the strong therapeutic potential of the antibody treatment and supported further development for use in Sudan virus outbreaks.

3.
J Infect Dis ; 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38487996

RESUMO

The most recent Sudan virus (SUDV) outbreak in Uganda was first detected in September 2022 and resulted in 164 laboratory-confirmed cases and 77 deaths. There are no approved vaccines against SUDV. Here, we investigated the protective efficacy of ChAdOx1-biEBOV in cynomolgus macaques using a prime or a prime-boost regimen. ChAdOx1-biEBOV is a replication-deficient simian adenovirus vector encoding SUDV and Ebola virus (EBOV) glycoproteins (GPs). Intramuscular vaccination induced SUDV and EBOV GP-specific IgG responses and neutralizing antibodies. Upon challenge with SUDV, vaccinated animals showed signs of disease like those observed in control animals, and no difference in survival outcomes were measured among all three groups. Viral load in blood samples and in tissue samples obtained after necropsy were not significantly different between groups. Overall, this study highlights the importance of evaluating vaccines in multiple animal models and demonstrates the importance of understanding protective efficacy in both animal models and human hosts.

4.
J Virol ; 97(10): e0059023, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37750724

RESUMO

IMPORTANCE: Ebola disease (EBOD) is a public health threat with a high case fatality rate. Most EBOD outbreaks have occurred in remote locations, but the 2013-2016 Western Africa outbreak demonstrated how devastating EBOD can be when it reaches an urban population. Here, the 2022 Sudan virus disease (SVD) outbreak in Mubende District, Uganda, is summarized, and the genetic relatedness of the new variant is evaluated. The Mubende variant exhibited 96% amino acid similarity with historic SUDV sequences from the 1970s and a high degree of conservation throughout the outbreak, which was important for ongoing diagnostics and highly promising for future therapy development. Genetic differences between viruses identified during the Mubende SVD outbreak were linked with epidemiological data to better interpret viral spread and contact tracing chains. This methodology should be used to better integrate discrete epidemiological and sequence data for future viral outbreaks.


Assuntos
Surtos de Doenças , Ebolavirus , Variação Genética , Doença pelo Vírus Ebola , Humanos , Surtos de Doenças/estatística & dados numéricos , Ebolavirus/química , Ebolavirus/classificação , Ebolavirus/genética , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/transmissão , Doença pelo Vírus Ebola/virologia , Uganda/epidemiologia , Busca de Comunicante
5.
BMC Infect Dis ; 24(1): 543, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816800

RESUMO

BACKGROUND: In 2022, an Ebola disease outbreak caused by Sudan virus (SUDV) occurred in Uganda, primarily affecting Mubende and Kassanda districts. We determined risk factors for SUDV infection among household members (HHM) of cases. METHODS: We conducted a case-control and retrospective cohort study in January 2023. Cases were RT-PCR-confirmed SUDV infection in residents of Mubende or Kassanda districts during the outbreak. Case-households housed a symptomatic, primary case-patient for ≥ 24 h and had ≥ 1 secondary case-patient with onset < 2 weeks after their last exposure to the primary case-patient. Control households housed a case-patient and other HHM but no secondary cases. A risk factor questionnaire was administered to the primary case-patient or another adult who lived at home while the primary case-patient was ill. We conducted a retrospective cohort study among case-household members and categorized their interactions with primary case-patients during their illnesses as none, minimal, indirect, and direct contact. We conducted logistic regression to explore associations between exposures and case-household status, and Poisson regression to identify risk factors for SUDV infection among HHM. RESULTS: Case- and control-households had similar median sizes. Among 19 case-households and 51 control households, primary case-patient death (adjusted odds ratio [ORadj] = 7.6, 95% CI 1.4-41) and ≥ 2 household bedrooms (ORadj=0.19, 95% CI 0.056-0.71) were associated with case-household status. In the cohort of 76 case-HHM, 44 (58%) were tested for SUDV < 2 weeks from their last contact with the primary case-patient; 29 (38%) were positive. Being aged ≥ 18 years (adjusted risk ratio [aRRadj] = 1.9, 95%CI: 1.01-3.7) and having direct or indirect contact with the primary case-patient (aRRadj=3.2, 95%CI: 1.1-9.7) compared to minimal or no contact increased risk of Sudan virus disease (SVD). Access to a handwashing facility decreased risk (aRRadj=0.52, 95%CI: 0.31-0.88). CONCLUSION: Direct contact, particularly providing nursing care for and sharing sleeping space with SVD patients, increased infection risk among HHM. Risk assessments during contact tracing may provide evidence to justify closer monitoring of some HHM. Health messaging should highlight the risk of sharing sleeping spaces and providing nursing care for persons with Ebola disease symptoms and emphasize hand hygiene to aid early case identification and reduce transmission.


Assuntos
Surtos de Doenças , Características da Família , Doença pelo Vírus Ebola , Humanos , Uganda/epidemiologia , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/transmissão , Fatores de Risco , Masculino , Adulto , Feminino , Estudos Retrospectivos , Estudos de Casos e Controles , Adolescente , Adulto Jovem , Pessoa de Meia-Idade , Criança , Pré-Escolar , Ebolavirus , Lactente
6.
BMC Infect Dis ; 24(1): 520, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38783244

RESUMO

BACKGROUND: On 20 September 2022, Uganda declared its fifth Sudan virus disease (SVD) outbreak, culminating in 142 confirmed and 22 probable cases. The reproductive rate (R) of this outbreak was 1.25. We described persons who were exposed to the virus, became infected, and they led to the infection of an unusually high number of cases during the outbreak. METHODS: In this descriptive cross-sectional study, we defined a super-spreader person (SSP) as any person with real-time polymerase chain reaction (RT-PCR) confirmed SVD linked to the infection of ≥ 13 other persons (10-fold the outbreak R). We reviewed illness narratives for SSPs collected through interviews. Whole-genome sequencing was used to support epidemiologic linkages between cases. RESULTS: Two SSPs (Patient A, a 33-year-old male, and Patient B, a 26-year-old male) were identified, and linked to the infection of one probable and 50 confirmed secondary cases. Both SSPs lived in the same parish and were likely infected by a single ill healthcare worker in early October while receiving healthcare. Both sought treatment at multiple health facilities, but neither was ever isolated at an Ebola Treatment Unit (ETU). In total, 18 secondary cases (17 confirmed, one probable), including three deaths (17%), were linked to Patient A; 33 secondary cases (all confirmed), including 14 (42%) deaths, were linked to Patient B. Secondary cases linked to Patient A included family members, neighbours, and contacts at health facilities, including healthcare workers. Those linked to Patient B included healthcare workers, friends, and family members who interacted with him throughout his illness, prayed over him while he was nearing death, or exhumed his body. Intensive community engagement and awareness-building were initiated based on narratives collected about patients A and B; 49 (96%) of the secondary cases were isolated in an ETU, a median of three days after onset. Only nine tertiary cases were linked to the 51 secondary cases. Sequencing suggested plausible direct transmission from the SSPs to 37 of 39 secondary cases with sequence data. CONCLUSION: Extended time in the community while ill, social interactions, cross-district travel for treatment, and religious practices contributed to SVD super-spreading. Intensive community engagement and awareness may have reduced the number of tertiary infections. Intensive follow-up of contacts of case-patients may help reduce the impact of super-spreading events.


Assuntos
Surtos de Doenças , Humanos , Uganda/epidemiologia , Masculino , Estudos Transversais , Adulto , Feminino , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/virologia , Sequenciamento Completo do Genoma , Ebolavirus/genética , Ebolavirus/isolamento & purificação
7.
J Formos Med Assoc ; 123(1): 16-22, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37355438

RESUMO

The recent Sudan virus disease (SVD) outbreak in Uganda is a reminder of threat from Filovirus diseases. Unlike Ebola virus disease, no effective antiviral and vaccine is available for SVD. The outbreak was declared over after 115 days, with 142 confirmed cases and case fatality rate of 39%, before any dose of candidate vaccine could be used on contacts. We provide a quick review of up-to-date information on the Uganda outbreak, summary of previous outbreaks, and detail the existing SVD treatment and vaccine candidates. Evolution of disease attributes and the impact on public health were also discussed. For high consequence infectious disease like SVD, it takes international collaboration to be better prepared for the next outbreak.


Assuntos
Doença pelo Vírus Ebola , Vacinas , Humanos , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/prevenção & controle , Surtos de Doenças/prevenção & controle , Uganda/epidemiologia
8.
J Infect Dis ; 228(Suppl 7): S465-S473, 2023 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-37592878

RESUMO

BACKGROUND: The origins of Ebola disease outbreaks remain enigmatic. Historically outbreaks have been attributed to spillover events from wildlife. However, recent data suggest that some outbreaks may originate from human-to-human transmission of prior outbreak strains instead of spillover. Clarifying the origins of Ebola disease outbreaks could improve detection and mitigation of future outbreaks. METHODS: We reviewed the origins of all Ebola disease outbreaks from 1976 to 2022 to analyze the earliest cases and characteristics of each outbreak. The epidemiology and phylogenetic relationships of outbreak strains were used to further identify the likely source of each outbreak. RESULTS: From 1976 to 2022 there were 35 Ebola disease outbreaks with 48 primary/index cases. While the majority of outbreaks were associated with wildlife spillover, resurgence of human-to-human transmission could account for roughly a quarter of outbreaks caused by Ebola virus. Larger outbreaks were more likely to lead to possible resurgence, and nosocomial transmission was associated with the majority of outbreaks. CONCLUSIONS: While spillover from wildlife has been a source for many Ebola disease outbreaks, multiple outbreaks may have originated from flare-ups of prior outbreak strains. Improving access to diagnostics as well as identifying groups at risk for resurgence of ebolaviruses will be crucial to preventing future outbreaks.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Animais , Humanos , Filogenia , Ebolavirus/genética , Surtos de Doenças/prevenção & controle , Animais Selvagens
9.
J Infect Dis ; 228(Suppl 7): S691-S700, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37288609

RESUMO

Filoviruses, including ebolaviruses and marburgviruses, can cause severe and often fatal disease in humans. Over the past several years, antibody therapy has emerged as a promising strategy for the treatment of filovirus disease. Here, we describe 2 distinct cross-reactive monoclonal antibodies (mAbs) isolated from mice immunized with recombinant vesicular stomatitis virus-based filovirus vaccines. Both mAbs recognized the glycoproteins of multiple different ebolaviruses and exhibited broad but differential in vitro neutralization activities against these viruses. By themselves, each mAb provided partial to full protection against Ebola virus in mice, and in combination, the mAbs provided 100% protection against Sudan virus challenge in guinea pigs. This study identified novel mAbs that were elicited through immunization and able to provide protection from ebolavirus infection, thus enriching the pool of candidate therapeutics for treating Ebola disease.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Humanos , Animais , Cobaias , Camundongos , Anticorpos Monoclonais , Terapia Combinada de Anticorpos , Anticorpos Neutralizantes , Anticorpos Antivirais
10.
J Infect Dis ; 228(Suppl 7): S660-S670, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37171813

RESUMO

BACKGROUND: The family Filoviridae consists of several virus members known to cause significant mortality and disease in humans. Among these, Ebola virus (EBOV), Marburg virus (MARV), Sudan virus (SUDV), and Bundibugyo virus (BDBV) are considered the deadliest. The vaccine, Ervebo, was shown to rapidly protect humans against Ebola disease, but is indicated only for EBOV infections with limited cross-protection against other filoviruses. Whether multivalent formulations of similar recombinant vesicular stomatitis virus (rVSV)-based vaccines could likewise confer rapid protection is unclear. METHODS: Here, we tested the ability of an attenuated, quadrivalent panfilovirus VesiculoVax vaccine (rVSV-Filo) to elicit fast-acting protection against MARV, EBOV, SUDV, and BDBV. Groups of cynomolgus monkeys were vaccinated 7 days before exposure to each of the 4 viral pathogens. All subjects (100%) immunized 1 week earlier survived MARV, SUDV, and BDBV challenge; 80% survived EBOV challenge. Survival correlated with lower viral load, higher glycoprotein-specific immunoglobulin G titers, and the expression of B-cell-, cytotoxic cell-, and antigen presentation-associated transcripts. CONCLUSIONS: These results demonstrate multivalent VesiculoVax vaccines are suitable for filovirus outbreak management. The highly attenuated nature of the rVSV-Filo vaccine may be preferable to the Ervebo "delta G" platform, which induced adverse events in a subset of recipients.


Assuntos
Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Marburgvirus , Vacinas Virais , Humanos , Animais , Vacinas Atenuadas , Macaca fascicularis , Vesiculovirus/genética , Vírus da Estomatite Vesicular Indiana , Anticorpos Antivirais
11.
J Infect Dis ; 228(Suppl 6): S446-S459, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37849404

RESUMO

Viruses in the family Filoviridae, including the commonly known Ebola (EBOV) and Marburg (MARV) viruses, can cause severe hemorrhagic fever in humans and nonhuman primates. Sporadic outbreaks of filovirus disease occur in sub-Saharan Africa with reported case fatality rates ranging from 25% to 90%. The high mortality and increasing frequency and magnitude of recent outbreaks along with the increased potential for spread from rural to urban areas highlight the importance of pandemic preparedness for these viruses. Despite their designation as high-priority pathogens, numerous scientific gaps exist in critical areas. In this review, these gaps and an assessment of potential prototype pathogen candidates are presented for this important virus family.


Assuntos
Ebolavirus , Filoviridae , Doença pelo Vírus Ebola , Marburgvirus , Animais , Humanos , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/prevenção & controle , Surtos de Doenças
12.
Emerg Infect Dis ; 29(7): 1429-1432, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37347815

RESUMO

We estimated the mean serial interval for Sudan virus in Uganda to be 11.7 days (95 CI% 8.2-15.8 days). Estimates for the 2022 outbreak indicate a mean basic reproduction number of 2.4-2.7 (95% CI 1.7-3.5). Estimated net reproduction numbers across districts suggest a marked spatial heterogeneity.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Humanos , Doença pelo Vírus Ebola/epidemiologia , Uganda/epidemiologia , Surtos de Doenças , Número Básico de Reprodução
13.
Proc Natl Acad Sci U S A ; 117(7): 3768-3778, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32015126

RESUMO

Antibody-based therapies are a promising treatment option for managing ebolavirus infections. Several Ebola virus (EBOV)-specific and, more recently, pan-ebolavirus antibody cocktails have been described. Here, we report the development and assessment of a Sudan virus (SUDV)-specific antibody cocktail. We produced a panel of SUDV glycoprotein (GP)-specific human chimeric monoclonal antibodies (mAbs) using both plant and mammalian expression systems and completed head-to-head in vitro and in vivo evaluations. Neutralizing activity, competitive binding groups, and epitope specificity of SUDV mAbs were defined before assessing protective efficacy of individual mAbs using a mouse model of SUDV infection. Of the mAbs tested, GP base-binding mAbs were more potent neutralizers and more protective than glycan cap- or mucin-like domain-binding mAbs. No significant difference was observed between plant and mammalian mAbs in any of our in vitro or in vivo evaluations. Based on in vitro and rodent testing, a combination of two SUDV-specific mAbs, one base binding (16F6) and one glycan cap binding (X10H2), was down-selected for assessment in a macaque model of SUDV infection. This cocktail, RIID F6-H2, provided protection from SUDV infection in rhesus macaques when administered at 50 mg/kg on days 4 and 6 postinfection. RIID F6-H2 is an effective postexposure SUDV therapy and provides a potential treatment option for managing human SUDV infection.


Assuntos
Anticorpos Antivirais/administração & dosagem , Ebolavirus/imunologia , Doença pelo Vírus Ebola/tratamento farmacológico , Animais , Anticorpos Monoclonais/administração & dosagem , Modelos Animais de Doenças , Ebolavirus/genética , Feminino , Glicoproteínas/imunologia , Doença pelo Vírus Ebola/virologia , Humanos , Imunoterapia , Macaca mulatta , Masculino , Camundongos , Proteínas Virais/imunologia
14.
J Virol ; 95(18): e0107321, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34232742

RESUMO

Sudan virus (SUDV) is one of five filoviruses that compose the genus Ebolavirus that has been responsible for episodic outbreaks in Central Africa. While the SUDV glycoprotein (GP) structure has been solved, GP residues that affect SUDV entry have not been extensively examined; many of the entry characteristics of SUDV GP are inferred from studies with the Zaire Ebola virus (EBOV) GP. Here, we investigate the effect on virus entry of a naturally occurring polymorphism in SUDV GP. Two of the earliest SUDV isolates contain glutamine at residue 95 (Q95) within the base region of GP1, whereas more recent SUDV isolates and GPs from all other ebolaviruses carry lysine at this position (K95). A K95Q change dramatically decreased titers of pseudovirions bearing SUDV GP, whereas the K95Q substitution in EBOV GP had no effect on titer. We evaluated virus entry to identify SUDV GP Q95-specific entry defects. The presence of Q95 in either EBOV or SUDV GP resulted in enhanced sensitivity of GP to proteolytic processing, yet this could not account for the SUDV-specific decrease in GP Q95 infectivity. We found that SUDV GP Q95 pseudovirions were more sensitive to imipramine, a GP-destabilizing antiviral. In contrast, SUDV GP K95 was more stable, requiring elevated temperatures to inhibit virus infection. Thus, the residue present at GP 95 has a critical role in stabilizing the SUDV glycoprotein, whereas this polymorphism has no effect on EBOV GP stability. These results provide novel insights into filovirus species-specific GP structure that affects virus infectivity. IMPORTANCE Filovirus outbreaks are associated with significant morbidity and mortality. Understanding the structural constraints of filoviral GPs that control virus entry into cells is critical for rational development of novel antivirals to block infection. Here, we identify a naturally occurring glutamine (Q) to lysine (K) polymorphism at residue 95 as a critical determinant of Sudan virus GP stability but not Zaire Ebola virus GP stability. We propose that glutamine at residue 95 in Sudan virus GP mediates decreased virus entry, thereby reducing infectivity. Our findings highlight a unique structural characteristic of Sudan virus GP that affects GP-mediated functionality. Further, it provides a cautionary note for the development of future broad-spectrum filovirus antivirals.


Assuntos
Ebolavirus/fisiologia , Glicoproteínas/química , Doença pelo Vírus Ebola/virologia , Especificidade de Hospedeiro , Polimorfismo Genético , Proteínas do Envelope Viral/química , Internalização do Vírus , Sequência de Aminoácidos , Animais , Células CHO , Chlorocebus aethiops , Cricetulus , Feminino , Glicoproteínas/genética , Doença pelo Vírus Ebola/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Estabilidade Proteica , Homologia de Sequência , Células Vero , Proteínas do Envelope Viral/genética
15.
J Virol ; 95(17): e0071421, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34160256

RESUMO

Ebola virus (EBOV), of the family Filoviridae, is an RNA virus that can cause a hemorrhagic fever with a high mortality rate. Defective viral genomes (DVGs) are truncated genomes that have been observed during multiple RNA virus infections, including in vitro EBOV infection, and have previously been associated with viral persistence and immunostimulatory activity. As DVGs have been detected in cells persistently infected with EBOV, we hypothesized that DVGs may also accumulate during viral replication in filovirus-infected hosts. Therefore, we interrogated sequence data from serum and tissue samples using a bioinformatics tool in order to identify the presence of DVGs in nonhuman primates (NHPs) infected with EBOV, Sudan virus (SUDV), or Marburg virus (MARV). Multiple 5' copy-back DVGs (cbDVGs) were detected in NHP serum during the acute phase of filovirus infection. While the relative abundance of total DVGs in most animals was low, serum collected during acute EBOV and SUDV infections, but not MARV infections, contained a higher proportion of short trailer sequence cbDVGs than the challenge stock. This indicated an accumulation of these DVGs throughout infection, potentially due to the preferential replication of short DVGs over the longer viral genome. Using reverse transcriptase PCR (RT-PCR) and deep sequencing, we also confirmed the presence of 5' cbDVGs in EBOV-infected NHP testes, which is of interest due to EBOV persistence in semen of male survivors of infection. This work suggests that DVGs play a role in EBOV infection in vivo and that further study will lead to a better understanding of EBOV pathogenesis. IMPORTANCE The study of filovirus pathogenesis is critical for understanding the consequences of infection and for the development of strategies to ameliorate future outbreaks. Defective viral genomes (DVGs) have been detected during EBOV infections in vitro; however, their presence in in vivo infections remains unknown. In this study, DVGs were detected in samples collected from EBOV- and SUDV-infected nonhuman primates (NHPs). The accumulation of these DVGs in the trailer region of the genome during infection indicates a potential role in EBOV and SUDV pathogenesis. In particular, the presence of DVGs in the testes of infected NHPs requires further investigation as it may be linked to the establishment of persistence.


Assuntos
Vírus Defeituosos/genética , Ebolavirus/genética , Genoma Viral , Doença pelo Vírus Ebola/virologia , Interações Hospedeiro-Patógeno , Macaca mulatta/virologia , Replicação Viral , Animais , Feminino , Masculino
16.
J Virol ; 94(9)2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32075939

RESUMO

The 2013-2016 Ebola outbreak in West Africa led to accelerated efforts to develop vaccines against these highly virulent viruses. A live, recombinant vesicular stomatitis virus-based vaccine has been deployed in outbreak settings and appears highly effective. Vaccines based on replication-deficient adenovirus vectors either alone or in combination with a multivalent modified vaccinia Ankara (MVA) Ebola vaccine also appear promising and are progressing in clinical evaluation. However, the ability of current live vector-based approaches to protect against multiple pathogenic species of Ebola is not yet established, and eliciting durable responses may require additional booster vaccinations. Here, we report the development of a bivalent, spherical Ebola virus-like particle (VLP) vaccine that incorporates glycoproteins (GPs) from Zaire Ebola virus (EBOV) and Sudan Ebola virus (SUDV) and is designed to extend the breadth of immunity beyond EBOV. Immunization of rabbits with bivalent Ebola VLPs produced antibodies that neutralized all four pathogenic species of Ebola viruses and elicited antibody-dependent cell-mediated cytotoxicity (ADCC) responses against EBOV and SUDV. Vaccination of rhesus macaques with bivalent VLPs generated strong humoral immune responses, including high titers of binding, as well as neutralizing antibodies and ADCC responses. VLP vaccination led to a significant increase in the frequency of Ebola GP-specific CD4 and CD8 T cell responses. These results demonstrate that a novel bivalent Ebola VLP vaccine elicits strong humoral and cellular immune responses against pathogenic Ebola viruses and support further evaluation of this approach as a potential addition to Ebola vaccine development efforts.IMPORTANCE Ebola outbreaks result in significant morbidity and mortality in affected countries. Although several leading candidate Ebola vaccines have been developed and advanced in clinical testing, additional vaccine candidates may be needed to provide protection against different Ebola species and to extend the durability of protection. A novel approach demonstrated here is to express two genetically diverse glycoproteins on a spherical core, generating a vaccine that can broaden immune responses against known pathogenic Ebola viruses. This approach provides a new method to broaden and potentially extend protective immune responses against Ebola viruses.


Assuntos
Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , África Ocidental , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Feminino , Glicoproteínas/imunologia , Imunização , Macaca mulatta , Masculino , Vacinação , Vacinas Atenuadas , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia
17.
Emerg Infect Dis ; 24(1): 114-117, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29260678

RESUMO

To determine whether fruit bats in Singapore have been exposed to filoviruses, we screened 409 serum samples from bats of 3 species by using a multiplex assay that detects antibodies against filoviruses. Positive samples reacted with glycoproteins from Bundibugyo, Ebola, and Sudan viruses, indicating filovirus circulation among bats in Southeast Asia.


Assuntos
Quirópteros/sangue , Quirópteros/virologia , Ebolavirus , Marburgvirus , Proteínas do Envelope Viral/sangue , Animais , Glicoproteínas/sangue , Glicoproteínas/genética , Glicoproteínas/isolamento & purificação , Estudos Soroepidemiológicos , Singapura/epidemiologia
18.
J Infect Dis ; 215(1): 64-69, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27226206

RESUMO

Here we describe clinicopathologic features of Ebola virus disease in pregnancy. One woman infected with Sudan virus in Gulu, Uganda, in 2000 had a stillbirth and survived, and another woman infected with Bundibugyo virus had a live birth with maternal and infant death in Isiro, the Democratic Republic of the Congo in 2012. Ebolavirus antigen was seen in the syncytiotrophoblast and placental maternal mononuclear cells by immunohistochemical analysis, and no antigen was seen in fetal placental stromal cells or fetal organs. In the Gulu case, ebolavirus antigen localized to malarial parasite pigment-laden macrophages. These data suggest that trophoblast infection may be a mechanism of transplacental ebolavirus transmission.


Assuntos
Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/patologia , Doença pelo Vírus Ebola/virologia , Complicações Infecciosas na Gravidez/patologia , Complicações Infecciosas na Gravidez/virologia , Adulto , Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Antígenos Virais/isolamento & purificação , República Democrática do Congo , Ebolavirus/química , Ebolavirus/genética , Ebolavirus/imunologia , Feminino , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/transmissão , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Imuno-Histoquímica , Macrófagos/parasitologia , Macrófagos/ultraestrutura , Macrófagos/virologia , Malária/complicações , Malária/imunologia , Malária/virologia , Microscopia Eletrônica de Transmissão , Placenta/ultraestrutura , Placenta/virologia , Reação em Cadeia da Polimerase , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/parasitologia , Natimorto , Células Estromais/ultraestrutura , Células Estromais/virologia , Trofoblastos/parasitologia , Trofoblastos/ultraestrutura , Trofoblastos/virologia
19.
J Infect Dis ; 212 Suppl 2: S295-304, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25920319

RESUMO

Sudan virus (SUDV), like the closely related Ebola virus (EBOV), is a filovirus that causes severe hemorrhagic disease. They both contain an RNA editing site in the glycoprotein gene that controls expression of soluble and full-length protein. We tested the consequences of cell culture passage on the genome sequence at the SUDV editing site locus and determined whether this affected virulence. Passage resulted in expansion of the SUDV editing site, similar to that observed with EBOV. We compared viruses possessing either the wild-type or expanded editing site, using a nonhuman primate model of disease. Despite differences in virus serum titer at one time point, there were no significant differences in time to death or any other measured parameter. These data imply that changes at this locus were not important for SUDV lethality.


Assuntos
Ebolavirus/genética , Ebolavirus/patogenicidade , Glicoproteínas/genética , Doença pelo Vírus Ebola/virologia , Edição de RNA/genética , Animais , Chlorocebus aethiops , Genoma Viral/genética , Haplorrinos , Inoculações Seriadas/métodos , Sudão , Células Vero/virologia , Carga Viral/métodos , Virulência/genética
20.
J Infect Dis ; 212 Suppl 2: S282-94, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25943199

RESUMO

A major obstacle in ebolavirus research is the lack of a small-animal model for Sudan virus (SUDV), as well as other wild-type (WT) ebolaviruses. Here, we expand on research by Bray and by Lever et al suggesting that WT ebolaviruses are pathogenic in mice deficient for the type 1 interferon (IFN) α/ß receptor (IFNα/ßR-/-). We examined the disease course of several WT ebolaviruses: Boneface (SUDV/Bon) and Gulu variants of SUDV, Ebola virus (EBOV), Bundibugyo virus (BDBV), Taï Forest virus, and Reston virus (RESTV). We determined that exposure to WT SUDV or EBOV results in reproducible signs of disease in IFNα/ßR-/- mice, as measured by weight loss and partial lethality. Vaccination with the SUDV or EBOV glycoprotein (GP)-expressing Venezuelan equine encephalitis viral replicon particle vaccine protected these mice from SUDV/Bon and EBOV challenge, respectively. Treatment with SUDV- or EBOV-specific anti-GP antibodies protected mice from challenge when delivered 1-3 days after infection. Serial sampling experiments revealed evidence of disseminated intravascular coagulation in the livers of mice infected with the Boneface variant of SUDV, EBOV, and BDBV. Taken together, these data solidify the IFNα/ßR-/- mouse as an important and useful model for the study of WT EBOV disease.


Assuntos
Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/virologia , Receptor de Interferon alfa e beta/deficiência , Virulência/fisiologia , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Vacinas contra Ebola/imunologia , Ebolavirus/metabolismo , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Replicon/imunologia , Vacinação/métodos , Células Vero/virologia , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Virulência/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa