1,473 results on '"Filovirus"'
Search Results
2. A Bivalent Adenovirus-Vectored Vaccine Induces a Robust Humoral Response, but Does Not Protect Cynomolgus Macaques Against a Lethal Challenge With Sudan Virus.
- Author
-
Tol, Sarah van, Fletcher, Paige, Feldmann, Friederike, Mukesh, Reshma K, Port, Julia R, Gallogly, Shane, Schulz, Jonathan E, Rhoderick, Joseph F, Makinson, Rebecca, Carmody, Aaron, Myers, Lara, Lovaglio, Jamie, Smith, Brian J, Okumura, Atsushi, Shaia, Carl, Saturday, Greg, Marzi, Andrea, Lambe, Teresa, Munster, Vincent J, and Doremalen, Neeltje van
- Abstract
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 immunoglobulin G responses and neutralizing antibodies. Upon challenge with SUDV, vaccinated animals showed signs of disease like those observed in control animals, and no differences in survival outcomes were measured among all 3 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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. The cellular protein phosphatase 2A is a crucial host factor for Marburg virus transcription.
- Author
-
von Creytz, Isabel, Rohde, Cornelius, and Biedenkopf, Nadine
- Subjects
- *
TRANSCRIPTION factors , *MARBURG virus , *GENETIC transcription , *REVERSE genetics , *PHOSPHOPROTEIN phosphatases - Abstract
Little is known regarding the molecular mechanisms that highly pathogenic Marburg virus (MARV) utilizes to transcribe and replicate its genome. Previous studies assumed that dephosphorylation of the filoviral transcription factor VP30 supports transcription, while phosphorylated VP30 reduces transcription. Here, we focused on the role of the host protein phosphatase 2A (PP2A) for VP30 dephosphorylation and promotion of viral transcription. We could show that MARV NP interacts with the subunit B56 of PP2A, as previously shown for the Ebola virus, and that this interaction is important for MARV transcription activity. Inhibition of the interaction between PP2A and NP either by mutating the B56 binding motif encoded on NP, or the use of a PP2A inhibitor, induced VP30 hyperphosphorylation, and as a consequence a decrease of MARV transcription as well as viral growth. These results suggest that NP plays a key role in the dephosphorylation of VP30 by recruiting PP2A. Generation of recombinant (rec) MARV lacking the PP2A-B56 interaction motif on NP was not possible suggesting an essential role of PP2A-mediated VP30 dephosphorylation for the MARV replication cycle. Likewise, we were not able to generate recMARV containing VP30 phosphomimetic mutants indicating that dynamic cycles of VP30 de- and rephosphorylation are a prerequisite for an efficient viral life cycle. As the specific binding motifs of PP2A-B56 and VP30 within NP are highly conserved among the filoviral family, our data suggest a conserved mechanism for filovirus VP30 dephosphorylation by PP2A, revealing the host factor PP2A as a promising target for pan-filoviral therapies. IMPORTANCE Our study elucidates the crucial role of host protein phosphatase 2A (PP2A) in Marburg virus (MARV) transcription. The regulatory subunit B56 of PP2A facilitates VP30 dephosphorylation, and hence transcription activation, via binding to NP. Our results, together with previous data, reveal a conserved mechanism of filovirus VP30 dephosphorylation by host factor PP2A at the NP interface and provide novel insights into potential pan-filovirus therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Enhanced broad spectrum in vitro antiviral efficacy of 3-F-4-MeO-Bn, 3-CN, and 4-CN derivatives of lipid remdesivir nucleoside monophosphate prodrugs.
- Author
-
McMillan, Rachel, Lo, Michael, Zhang, Xing-Quan, Beadle, James, Valiaeva, Nadejda, Garretson, Aaron, Clark, Alex, Freshman, Jon, Murphy, Joyce, Montgomery, Joel, Spiropoulou, Christina, Schooley, Robert, Hostetler, Karl, and Carlin, Aaron
- Subjects
Antiviral agents ,Broad spectrum antiviral ,Ebola virus ,Filovirus ,Flavivirus ,GS-441524 ,GS-5734 ,Hemorrhagic fever viruses ,Hendra virus ,Henipavirus ,Human coronavirus 229E ,Lipid prodrugs ,Nipah virus ,Paramyxovirus ,Pneumovirus ,RNA virus ,Remdesivir ,Remdesivir nucleoside ,Respiratory syncytial virus ,Respiratory viruses ,V2043 ,Zika virus ,dengue virus ,Antiviral Agents ,Prodrugs ,Nucleosides ,Glycerol ,Lipids - Abstract
Broad spectrum oral antivirals are urgently needed for the early treatment of many RNA viruses of clinical concern. We previously described the synthesis of 1-O-octadecyl-2-O-benzyl-glycero-3-phospho-RVn (V2043), an orally bioavailable lipid prodrug of remdesivir nucleoside (RVn, GS-441524) with broad spectrum antiviral activity against viruses with pandemic potential. Here we compared the relative activity of V2043 with new RVn lipid prodrugs containing sn-1 alkyl ether or sn-2 glycerol modifications. We found that 3-F-4-MeO-Bn, 3-CN-Bn, and 4-CN-Bn sn-2 glycerol modifications improved antiviral activity compared to V2043 when tested in vitro against clinically important RNA viruses from 5 virus families. These results support the continued development of V2043 and sn-2 glycerol modified RVn lipid prodrugs for the treatment of a broad range of RNA viruses for which there are limited therapies.
- Published
- 2023
5. Efficacy and Immunogenicity of a Recombinant Vesicular Stomatitis Virus-Vectored Marburg Vaccine in Cynomolgus Macaques.
- Author
-
Camargos, Vidyleison N., Rossi, Shannan L., Juelich, Terry L., Smith, Jennifer K., Vasilakis, Nikos, Freiberg, Alexander N., Nichols, Rick, and Fusco, Joan
- Subjects
- *
VESICULAR stomatitis , *MARBURG virus , *HEMORRHAGIC fever , *VACCINE effectiveness , *DEATH rate - Abstract
Filoviruses, like the Marburg (MARV) and Ebola (EBOV) viruses, have caused outbreaks associated with significant hemorrhagic morbidity and high fatality rates. Vaccines offer one of the best countermeasures for fatal infection, but to date only the EBOV vaccine has received FDA licensure. Given the limited cross protection between the EBOV vaccine and Marburg hemorrhagic fever (MHF), we analyzed the protective efficacy of a similar vaccine, rVSV-MARV, in the lethal cynomolgus macaque model. NHPs vaccinated with a single dose (as little as 1.6 × 107 pfu) of rVSV-MARV seroconverted to MARV G-protein prior to challenge on day 42. Vaccinemia was measured in all vaccinated primates, self-resolved by day 14 post vaccination. Importantly, all vaccinated NHPs survived lethal MARV challenge, and showed no significant alterations in key markers of morbid disease, including clinical signs, and certain hematological and clinical chemistry parameters. Further, apart from one primate (from which tissues were not collected and no causal link was established), no pathology associated with Marburg disease was observed in vaccinated animals. Taken together, rVSV-MARV is a safe and efficacious vaccine against MHF in cynomolgus macaques. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Non-Ebola Filoviruses: Potential Threats to Global Health Security.
- Author
-
Munyeku-Bazitama, Yannick, Edidi-Atani, Francois, and Takada, Ayato
- Subjects
- *
MARBURG virus , *VIRAL ecology , *EBOLA virus , *RNA viruses , *FILOVIRIDAE , *HEMORRHAGIC fever - Abstract
Filoviruses are negative-sense single-stranded RNA viruses often associated with severe and highly lethal hemorrhagic fever in humans and nonhuman primates, with case fatality rates as high as 90%. Of the known filoviruses, Ebola virus (EBOV), the prototype of the genus Orthoebolavirus, has been a major public health concern as it frequently causes outbreaks and was associated with an unprecedented outbreak in several Western African countries in 2013–2016, affecting 28,610 people, 11,308 of whom died. Thereafter, filovirus research mostly focused on EBOV, paying less attention to other equally deadly orthoebolaviruses (Sudan, Bundibugyo, and Taï Forest viruses) and orthomarburgviruses (Marburg and Ravn viruses). Some of these filoviruses have emerged in nonendemic areas, as exemplified by four Marburg disease outbreaks recorded in Guinea, Ghana, Tanzania, and Equatorial Guinea between 2021 and 2023. Similarly, the Sudan virus has reemerged in Uganda 10 years after the last recorded outbreak. Moreover, several novel bat-derived filoviruses have been discovered in the last 15 years (Lloviu virus, Bombali virus, Měnglà virus, and Dehong virus), most of which are poorly characterized but may display a wide host range. These novel viruses have the potential to cause outbreaks in humans. Several gaps are yet to be addressed regarding known and emerging filoviruses. These gaps include the virus ecology and pathogenicity, mechanisms of zoonotic transmission, host range and susceptibility, and the development of specific medical countermeasures. In this review, we summarize the current knowledge on non-Ebola filoviruses (Bombali virus, Bundibugyo virus, Reston virus, Sudan virus, Tai Forest virus, Marburg virus, Ravn virus, Lloviu virus, Měnglà virus, and Dehong virus) and suggest some strategies to accelerate specific countermeasure development. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Delayed treatment of cynomolgus macaques with a FVM04/ CA45 monoclonal antibody cocktail provides complete protection against lethal Sudan virus infection.
- Author
-
Mable Chan, Warner, Bryce M., Audet, Jonathan, Barker, Douglas, Tailor, Nikesh, Vendramelli, Robert, Truong, Thang, Tierney, Kevin, Boese, Amrit S., Honguy Qiu, Holtsberg, Frederick W., Aman, Javad, Kodihalli, Shantha, and Kobasa, Darwyn
- Subjects
- *
EBOLA virus , *VIRUS diseases , *VACCINE approval , *TREATMENT delay (Medicine) , *INNER cities - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. How does the polymerase of non-segmented negative strand RNA viruses commit to transcription or genome replication?
- Author
-
Kleiner, Victoria A. and Fearns, Rachel
- Subjects
- *
RNA replicase , *MOLECULAR biology , *RESPIRATORY syncytial virus , *GENETIC transcription , *RABIES virus , *EBOLA virus - Abstract
The Mononegavirales, or non-segmented negative-sense RNA viruses (nsNSVs), includes significant human pathogens, such as respiratory syncytial virus, parainfluenza virus, measles virus, Ebola virus, and rabies virus. Although these viruses differ widely in their pathogenic properties, they are united by each having a genome consisting of a single strand of negative-sense RNA. Consistent with their shared genome structure, the nsNSVs have evolved similar ways to transcribe their genome into mRNAs and replicate it to produce new genomes. Importantly, both mRNA transcription and genome replication are performed by a single virus-encoded polymerase. A fundamental and intriguing question is: how does the nsNSV polymerase commit to being either an mRNA transcriptase or a replicase? The polymerase must become committed to one process or the other either before it interacts with the genome template or in its initial interactions with the promoter sequence at the 3´ end of the genomic RNA. This review examines the biochemical, molecular biology, and structural biology data regarding the first steps of transcription and RNA replication that have been gathered over several decades for different families of nsNSVs. These findings are discussed in relation to possible models that could explain how an nsNSV polymerase initiates and commits to either transcription or genome replication. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Comparison of Routes of Administration, Frequency, and Duration of Favipiravir Treatment in Mouse and Guinea Pig Models of Ebola Virus Disease.
- Author
-
Johnson, Dylan M., Juelich, Terry, Zhang, Lihong, Smith, Jennifer K., Kalveram, Birte K., Perez, David, Smith, Jeanon, Grimes, Michael R., Garron, Tania, Torres, Maricela, Massey, Shane, Brasel, Trevor, Beasley, David W. C., Freiberg, Alex N., and Comer, Jason E.
- Subjects
- *
EBOLA virus disease , *EBOLA virus , *TREATMENT duration , *REGULATORY approval , *PRIMATES , *GUINEA pigs - Abstract
Favipiravir is a ribonucleoside analogue that has been explored as a therapeutic for the treatment of Ebola Virus Disease (EVD). Promising data from rodent models has informed nonhuman primate trials, as well as evaluation in patients during the 2013–2016 West African EVD outbreak of favipiravir treatment. However, mixed results from these studies hindered regulatory approval of favipiravir for the indication of EVD. This study examined the influence of route of administration, duration of treatment, and treatment schedule of favipiravir in immune competent mouse and guinea pig models using rodent-adapted Zaire ebolavirus (EBOV). A dose of 300 mg/kg/day of favipiravir with an 8-day treatment was found to be fully effective at preventing lethal EVD-like disease in BALB/c mice regardless of route of administration (oral, intraperitoneal, or subcutaneous) or whether it was provided as a once-daily dose or a twice-daily split dose. Preclinical data generated in guinea pigs demonstrates that an 8-day treatment of 300 mg/kg/day of favipiravir reduces mortality following EBOV challenge regardless of route of treatment or duration of treatments for 8, 11, or 15 days. This work supports the future translational development of favipiravir as an EVD therapeutic. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Low-Input, High-Resolution 5′ Terminal Filovirus RNA Sequencing with ViBE-Seq.
- Author
-
Ross, Stephen J., Hume, Adam J., Olejnik, Judith, Turcinovic, Jacquelyn, Honko, Anna N., McKay, Lindsay G. A., Connor, John H., Griffiths, Anthony, Mühlberger, Elke, and Cifuentes, Daniel
- Subjects
- *
MARBURG virus , *RNA sequencing , *EBOLA virus , *NUCLEOTIDE sequencing , *RNA viruses - Abstract
Although next-generation sequencing (NGS) has been instrumental in determining the genomic sequences of emerging RNA viruses, de novo sequence determination often lacks sufficient coverage of the 5′ and 3′ ends of the viral genomes. Since the genome ends of RNA viruses contain the transcription and genome replication promoters that are essential for viral propagation, a lack of terminal sequence information hinders the efforts to study the replication and transcription mechanisms of emerging and re-emerging viruses. To circumvent this, we have developed a novel method termed ViBE-Seq (Viral Bona Fide End Sequencing) for the high-resolution sequencing of filoviral genome ends using a simple yet robust protocol with high fidelity. This technique allows for sequence determination of the 5′ end of viral RNA genomes and mRNAs with as little as 50 ng of total RNA. Using the Ebola virus and Marburg virus as prototypes for highly pathogenic, re-emerging viruses, we show that ViBE-Seq is a reliable technique for rapid and accurate 5′ end sequencing of filovirus RNA sourced from virions, infected cells, and tissue obtained from infected animals. We also show that ViBE-Seq can be used to determine whether distinct reverse transcriptases have terminal deoxynucleotidyl transferase activity. Overall, ViBE-Seq will facilitate the access to complete sequences of emerging viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Macaque antibodies targeting Marburg virus glycoprotein induced by multivalent immunization.
- Author
-
Janus, Benjamin M., Ruixue Wang, Cleveland IV, Thomas E., Metcalf, Matthew C., Lemmer, Aaron C., van Dyk, Nydia, Sarah Jeong, Astavans, Anagh, Class, Kenneth, Fuerst, Thomas R., and Ofek, Gilad
- Subjects
- *
MARBURG virus , *MONOCLONAL antibodies , *MACAQUES , *IMMUNOGLOBULINS , *IMMUNOLOGIC memory , *CELL receptors , *EBOLA virus , *GABA receptors - Abstract
Marburg virus infection in humans is associated with case fatality rates that can reach up to 90%, but to date, there are no approved vaccines or monoclonal antibody (mAb) countermeasures. Here, we immunized Rhesus macaques with multivalent combinations of filovirus glycoprotein (GP) antigens belonging to Marburg, Sudan, and Ebola viruses to generate monospecific and cross-reactive antibody responses against them. From the animal that developed the highest titers of Marburg virus GP-specific neutralizing antibodies, we sorted single memory B cells using a heterologous Ravn virus GP probe and cloned and characterized a panel of 34 mAbs belonging to 28 unique lineages. Antibody specificities were assessed by overlapping pepscan and binding competition analyses, revealing that roughly a third of the lineages mapped to the conserved receptor binding region, including potent neutralizing lineages that were confirmed by negative stain electron microscopy to target this region. Additional lineages targeted a protective region on GP2, while others were found to possess cross-filovirus reactivity. Our study advances the understanding of orthomar-burgvirus glycoprotein antigenicity and furthers efforts to develop candidate antibody countermeasures against these lethal viruses. IMPORTANCE Marburg viruses were the first filoviruses characterized to emerge in humans in 1967 and cause severe hemorrhagic fever with average case fatality rates of ~50%. Although mAb countermeasures have been approved for clinical use against the related Ebola viruses, there are currently no approved countermeasures against Marburg viruses. We successfully isolated a panel of orthomarburgvirus GP-specific mAbs from a macaque immunized with a multivalent combination of filovirus antigens. Our analyses revealed that roughly half of the antibodies in the panel mapped to regions on the glycoprotein shown to protect from infection, including the host cell receptor binding domain and a protective region on the membrane-anchoring subunit. Other antibodies in the panel exhibited broad filovirus GP recognition. Our study describes the discovery of a diverse panel of cross-reactive macaque antibodies targeting orthomarburgvirus and other filovirus GPs and provides candidate immunotherapeutics for further study and development. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Evidence of repeated zoonotic pathogen spillover events at ecological boundaries
- Author
-
Antoine Filion, Mekala Sundaram, John Paul Schmidt, John M. Drake, and Patrick R. Stephens
- Subjects
disease ecology ,Schmalhausen’s law ,ecological boundaries ,spillover ,filovirus ,disease emergence ,Public aspects of medicine ,RA1-1270 - Abstract
Anthropogenic modifications to the landscape have altered several ecological processes worldwide, creating new ecological boundaries at the human/wildlife interface. Outbreaks of zoonotic pathogens often occur at these ecological boundaries, but the mechanisms behind new emergences remain drastically understudied. Here, we test for the influence of two types of ecosystem boundaries on spillover risk: (1) biotic transition zones such as species range edges and transitions between ecoregions and (2) land use transition zones where wild landscapes occur in close proximity to heavily impacted areas of high human population density. Using ebolavirus as a model system and an ensemble machine learning modeling framework, we investigated the role of likely reservoir (bats) and accidental host (primates) range edges and patterns of land use (defined using SEDAC categories) on past spillover events. Our results show that overlapping species range edges and heightened habitat diversity increase ebolavirus outbreaks risk. Moreover, we show that gradual transition zones, represent by high proportion of rangelands, acts as a buffer to reduces outbreak risks. With increasing landscape changes worldwide, we provide novel ecological and evolutionary insights into our understanding of zoonotic pathogen emergence and highlight the risk of aggressively developing ecological boundaries.
- Published
- 2024
- Full Text
- View/download PDF
13. A high-throughput, polymerase-targeted RT-PCR for broad detection of mammalian filoviruses
- Author
-
Na Cui, Yael L. Perez, Adam J. Hume, B. Ethan Nunley, Kevin Kong, Margaret G. Mills, Hong Xie, and Alexander L. Greninger
- Subjects
filovirus ,RT-PCR ,broad range PCR ,pan-filovirus ,zoonosis ,Microbiology ,QR1-502 - Abstract
ABSTRACT Filoviruses are some of the most lethal viruses in the modern world, and increasing numbers of filovirus species and genera have been discovered in recent years. Despite the potential severity of filovirus outbreaks in the human population, comparably few sensitive pan-filovirus RT-PCR assays have been described that might facilitate early detection and prevention. Here, we present a new pan-filovirus RT-PCR assay targeting the L polymerase gene for detection of all known mammalian filoviruses. We demonstrate the detection of 10 synthetic filovirus RNA templates with analytical sensitivity ranging from 178 to 3,354 copies/mL, without cross-reactivity on 10 non-filoviral human viral species. We verified assay performance on 10 inactivated filovirus isolates, yielding initial sensitivities of 0.012–44.17 TCID50/mL. We coupled this broadly reactive RT-PCR with a deep sequencing workflow that is amenable to high-throughput pooling to maximize detection and discovery potential. In summary, this pan-filovirus RT-PCR assay targets the most conserved filovirus gene, offers the widest breadth of coverage to date, and may help in the detection and discovery of novel filoviruses.IMPORTANCEFiloviruses remain some of the most mysterious viruses known to the world, with extremely high lethality rates and significant pandemic potential. Yet comparably few filovirus species and genera have been discovered to date and questions surround the definitive host species for zoonotic infections. Here, we describe a novel broadly reactive RT-PCR assay targeting the conserved L polymerase gene for high-throughput screening for filoviruses in a variety of clinical and environmental specimens. We demonstrate the assay can detect all known mammalian filoviruses and determine the sensitivity and specificity of the assay on synthetic RNA sequences, inactivated filovirus isolates, and non-filoviral species.
- Published
- 2024
- Full Text
- View/download PDF
14. Identifying Paucisymptomatic or Asymptomatic and Unrecognized Ebola Virus Disease Among Close Contacts Based on Exposure Risk Assessments and Screening Algorithms.
- Author
-
Gayedyu-Dennis, Dehkontee, Fallah, Mosoka P, Drew, Clara, Badio, Moses, Moses, JS, Fayiah, Tamba, Johnson, Kumblytee, Richardson, Eugene T, Weiser, Sheri D, Porco, Travis C, Martin, Jeffrey N, Sneller, Michael C, Rutherford, George W, Reilly, Cavan, Lindan, Christina P, and Kelly, JD
- Subjects
Humans ,Hemorrhagic Fever ,Ebola ,Risk Assessment ,Cohort Studies ,Disease Outbreaks ,Ebolavirus ,Asymptomatic Infections ,Ebola virus disease ,Filovirus ,Liberia ,epidemiology ,infection control ,screening tests ,Vaccine Related ,Prevention ,Biodefense ,Emerging Infectious Diseases ,Clinical Research ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Good Health and Well Being ,Biological Sciences ,Medical and Health Sciences ,Microbiology - Abstract
BackgroundThere is limited evidence to evaluate screening algorithms with rapid antigen testing and exposure assessments as identification strategies for paucisymptomatic or asymptomatic Ebola virus (EBOV) infection and unrecognized EBOV disease (EVD).MethodsWe used serostatus and self-reported postexposure symptoms from a cohort study to classify contact-participants as having no infection, paucisymptomatic or asymptomatic infection, or unrecognized EVD. Exposure risk was categorized as low, intermediate, or high. We created hypothetical scenarios to evaluate the World Health Organization (WHO) case definition with or without rapid diagnostic testing (RDT) or exposure assessments.ResultsThis analysis included 990 EVD survivors and 1909 contacts, of whom 115 (6%) had paucisymptomatic or asymptomatic EBOV infection, 107 (6%) had unrecognized EVD, and 1687 (88%) were uninfected. High-risk exposures were drivers of unrecognized EVD (adjusted odds ratio, 3.5 [95% confidence interval, 2.4-4.9]). To identify contacts with unrecognized EVD who test negative by the WHO case definition, the sensitivity was 96% with RDT (95% confidence interval, 91%-99%), 87% with high-risk exposure (82%-92%), and 97% with intermediate- to high-risk exposures (93%-99%). The proportion of false-positives was 2% with RDT and 53%-93% with intermediate- and/or high-risk exposures.ConclusionWe demonstrated the utility and trade-offs of sequential screening algorithms with RDT or exposure risk assessments as identification strategies for contacts with unrecognized EVD.
- Published
- 2023
15. Factors associated with death in patients admitted with Ebola virus disease to Ebola Treatment Units in Guinea, Sierra Leone, and Liberia – December 2013 to March 2016 [version 1; peer review: awaiting peer review]
- Author
-
Trokon Omarley Yeabah, Ibrahima Kaba, Gomathi Ramaswamy, Prabin Dahal, Alexandre Delamou, Benjamin T. Vonhm, Ralph W. Jetoh, Laura Merson, Adam C. Levine, Pryanka Relan, Anthony D. Harries, and Ajay M.V. Kumar
- Subjects
Research Article ,Articles ,West Africa ,Ebola ,mortality ,viral haemorrhagic fever ,filovirus ,SORT IT ,operational research ,pandemic preparedness - Abstract
Background The 2013-2016 West African Ebola Virus Disease (EVD) outbreak resulted in 28,600 cases and 11,300 deaths officially reported to the World Health Organization. Previous studies investigating factors associated with death had conflicting findings, interventions showing promising outcomes had small sample sizes, studies were often single- or dual-country based and most focused on laboratory-confirmed EVD and not on clinically-suspected EVD. We used the Ebola data platform of the Infectious Disease Data Observatory (IDDO) to review individual patient records to assess factors associated with death, and particularly whether there were differences between laboratory-confirmed and clinically-suspected cases. Methods This was a cohort study involving analysis of secondary data in the IDDO database. The study population included all patients classified as having either clinically-suspected or laboratory-confirmed EVD, admitted to 22 Ebola Treatment Units (ETU) in Guinea, Liberia and Sierra Leone between December 2013 and March 2016. Baseline characteristics and treatments were documented along with ETU exit outcomes. Factors associated with death were investigated by multivariable modified Poisson regression. Results There were 14,163 patients, of whom 6,208 (43.8%) were laboratory-confirmed and 7,955 (56.2%) were clinically-suspected. Outcomes were not recorded in 2,889 (20.4%) patients. Of the 11,274 patients with known outcomes, 4,090 (36.3%) died: 2,956 (43.6%) with laboratory-confirmed EVD and 1,134 (18.8%) with clinically-suspected EVD. The strongest risk factor for death was confirmed disease status. Patients with laboratory-confirmed disease had 2.9 times higher risk of death compared to clinically-suspected patients, after adjusting for other co-variables. Other factors significantly associated with death included a higher risk for patients aged ≥60 years and a lower risk for patients in Sierra Leone. Conclusions Although laboratory-confirmed patients admitted to ETUs fared worse than clinically-suspected patients, the latter still had a substantial risk of death and more attention needs to be paid to this group in future EVD outbreaks.
- Published
- 2024
- Full Text
- View/download PDF
16. The Current Pathogenicity and Potential Risk Evaluation of Marburg Virus to Cause Mysterious "Disease X"—An Update on Recent Evidences.
- Author
-
Mitu, Rahima Akter and Islam, Md. Rabiul
- Abstract
The World Health Organization (WHO) defined Disease X as an upcoming disease with the potential to cause a pandemic. Pathogen X is responsible for Disease X. Marburg virus disease (MVD) is one of the diseases from the priority disease list published by WHO. Marburg virus is a filamentous, negative-sense RNA virus that belongs to the same filovirus family as the lethal Ebola virus. Since the first discovery of this virus in 1967, 17 outbreaks occurred sporadically till 2023. Rousettus aegyptiacus acts as the natural reservoir of the virus. With an average incubation period of 5 to 10 days, its first target is the mononuclear phagocytic system cells. It is highly contagious and can be easily transmitted from animal to human and human to human via direct contact with blood or body fluid, feces, and semen of the infected host. Although Marburg disease has a high case fatality rate of close to 90%, unfortunately, there is no approved vaccines or treatments are available. The most recent outbreak of Marburg virus in Equatorial Guinea and Tanzania in 2023 caused an alert for global health. However, based on the last global pandemic of COVID-19 and the sudden re-emerging of monkeypox around the world, we can assume that the Marburg virus has the potential to cause a global pandemic. Our modern world depends on globalization, which helps the virus transmission among countries. The Marburg virus can easily be transmitted to humans by fruit bats of the Pteropodidae family. This virus causes severe hemorrhagic disease, and there are no specific vaccines and treatments available to combat it. Therefore, community engagement and early supportive care for patients are keys to successfully controlling MVD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Kenyan Free-Tailed Bats Demonstrate Seasonal Birth Pulse Asynchrony with Implications for Virus Maintenance.
- Author
-
Lunn, Tamika J., Jackson, Reilly T., Webala, Paul W., Ogola, Joseph, and Forbes, Kristian M.
- Subjects
ZOONOSES ,EBOLA virus disease ,COMMUNICABLE diseases ,SEASONS ,EBOLA virus - Abstract
Ecological information on wildlife reservoirs is fundamental for research targeting prevention of zoonotic infectious disease, yet basic information is lacking for many species in global hotspots of disease emergence. We provide the first estimates of synchronicity, magnitude, and timing of seasonal birthing in Mops condylurus, a putative ebolavirus host, and a co-roosting species, Mops pumilus (formerly Chaerephon pumilus). We show that population-level synchronicity of M. condylurus birthing is wide (~ 8.5 weeks) and even wider in M. pumilus (> 11 weeks). This is predicted to promote the likelihood of filovirus persistence under conditions of bi-annual birthing (two births per year). Ecological features underlying the magnitude of the birth pulse—relative female abundance (higher than expected for M. condylurus and lower for M. pumilus, based on literature) and reproductive rate (lower than expected)—will have countering effects on birthing magnitude. Species-specific models are needed to interpret how identified birth pulse attributes may interact with other features of molossid ebolavirus ecology to influence infection dynamics. As a common feature of wildlife species, and a key driver of infection dynamics, detailed information on seasonal birthing will be fundamental for future research on these species and will be informative for bat-borne zoonoses generally. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Sudan virus disease – A quick review
- Author
-
Tsung-Pei Tsou
- Subjects
Ebola ,Emerging infectious diseases ,Filovirus ,Hemorrhagic fever ,Sudan virus ,Medicine (General) ,R5-920 - Abstract
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.
- Published
- 2024
- Full Text
- View/download PDF
19. Stigmatization of Ebola virus disease survivors in 2022: A cross-sectional study of survivors in Sierra Leone
- Author
-
Brayden G. Schindell, Jia B. Kangbai, Souradet Y. Shaw, and Jason Kindrachuk
- Subjects
Ebola Virus Disease ,Stigma ,Long-term complications ,Viral Hemorrhagic Fever ,Filovirus ,Infectious and parasitic diseases ,RC109-216 ,Public aspects of medicine ,RA1-1270 - Abstract
Background: Evidence has demonstrated a high proportion of Ebola virus disease (EVD) survivors experienced stigma due to the disease. This study sought to understand the longer-term effects of stigma encountered by survivors of the 2014–2016 EVD epidemic living in Sierra Leone. Methods: This was a cross-sectional study of 595 EVD survivors and 403 close contacts (n = 998) from Sierra Leone. Assessments were conducted using a three-part survey between November 2021 to March 2022. We explored the socio-demographic factors associated with stigma experienced by EVD survivors. Findings: 50·6 % (n = 301) of EVD survivors reported that they continued to experience at least one aspect of stigma. Females were disproportionately affected by stigma, with 45·2 % of females reporting isolation from friends and family compared to 33·9 % of men (p = 0·005). Multivariable logistic regression models revealed those aged 40–44, living rurally, and reporting an acute infection longer than seven days was associated with EVD-related stigma at the time of survey. Interpretation: This study demonstrates stigma is still prevalent among people who survived EVD in 2022. It also identified socio-demographic factors associated with stigma that can be used for targeting interventions. Importantly, this highlights the continued need for EVD survivors to access mental healthcare and social support systems well after disease recovery. Funding: This study was funded by the Canadian Institutes for Health Research (Grant no. PJT-175098. JK is funded by a Tier 2 Canada Research Chair in the Molecular Pathogenesis of Emerging and Re-Emerging Viruses. SS is funded by a Tier 2 Canada Research Chair in Program Science and Global Public Health.
- Published
- 2024
- Full Text
- View/download PDF
20. Isolation, characterization, and circulation sphere of a filovirus in fruit bats.
- Author
-
Biao He, Tingsong Hu, Xiaomin Yan, Yanhui Pa, Yuhang Liu, Yang Liu, Nan Li, Jing Yu, Hailin Zhang, Yonghua Liu, Jun Chai, Yue Sun, Shijiang Mi, Yan Liu, Le Yi, Zhongzhong Tu, Yiyin Wang, Sheng Sun, Ye Feng, and Wendong Zhang
- Abstract
Bats are associated with the circulation of most mammalian filoviruses (FiVs), with pathogenic ones frequently causing deadly hemorrhagic fevers in Africa. Divergent FiVs have been uncovered in Chinese bats, raising concerns about their threat to public health. Here, we describe a long-term surveillance to track bat FiVs at orchards, eventually resulting in the identification and isolation of a FiV, Dehong virus (DEHV), from Rousettus leschenaultii bats. DEHV has a typical filovirus-like morphology with a wide spectrum of cell tropism. Its entry into cells depends on the engagement of Niemann-Pick C1, and its replication is inhibited by remdesivir. DEHV has the largest genome size of filoviruses, with phylogenetic analysis placing it between the genera Dianlovirus and Orthomarburgvirus, suggesting its classification as the prototype of a new genus within the family Filoviridae. The continuous detection of viral RNA in the serological survey, together with the wide host distribution, has revealed that the region covering southern Yunnan, China, and bordering areas is a natural circulation sphere for bat FiVs. These emphasize the need for a better understanding of the pathogenicity and potential risk of FiVs in the region. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Peripheral immune responses to filoviruses in a reservoir versus spillover hosts reveal transcriptional correlates of disease.
- Author
-
Guito, Jonathan C., Arnold, Catherine E., Schuh, Amy J., Amman, Brian R., Sealy, Tara K., Spengler, Jessica R., Harmon, Jessica R., Coleman-McCray, Joann D., Sanchez-Lockhart, Mariano, Palacios, Gustavo F., Towner, Jonathan S., and Prescott, Joseph B.
- Subjects
FILOVIRIDAE ,IMMUNE response ,MARBURG virus ,DISEASE resistance of plants ,NATURAL immunity - Abstract
Several filoviruses, including Marburg virus (MARV), cause severe disease in humans and nonhuman primates (NHPs). However, the Egyptian rousette bat (ERB, Rousettus aegyptiacus), the only known MARV reservoir, shows no overt illness upon natural or experimental infection, which, like other bat hosts of zoonoses, is due to well-adapted, likely species-specific immune features. Despite advances in understanding reservoir immune responses to filoviruses, ERB peripheral blood responses to MARV and how they compare to those of diseased filovirus-infected spillover hosts remain ill-defined. We thus conducted a longitudinal analysis of ERB blood gene responses during acute MARV infection. These data were then contrasted with a compilation of published primate blood response studies to elucidate gene correlates of filovirus protection versus disease. Our work expands on previous findings in MARVinfected ERBs by supporting both host resistance and disease tolerance mechanisms, offers insight into the peripheral immunocellular repertoire during infection, and provides the most direct known cross-examination between reservoir and spillover hosts of the most prevalently-regulated response genes, pathways and activities associated with differences in filovirus pathogenesis and pathogenicity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Stigmatization of Ebola virus disease survivors in 2022: A cross-sectional study of survivors in Sierra Leone.
- Author
-
Schindell, Brayden G., Kangbai, Jia B., Shaw, Souradet Y., and Kindrachuk, Jason
- Abstract
Evidence has demonstrated a high proportion of Ebola virus disease (EVD) survivors experienced stigma due to the disease. This study sought to understand the longer-term effects of stigma encountered by survivors of the 2014–2016 EVD epidemic living in Sierra Leone. This was a cross-sectional study of 595 EVD survivors and 403 close contacts (n = 998) from Sierra Leone. Assessments were conducted using a three-part survey between November 2021 to March 2022. We explored the socio-demographic factors associated with stigma experienced by EVD survivors. 50·6 % (n = 301) of EVD survivors reported that they continued to experience at least one aspect of stigma. Females were disproportionately affected by stigma, with 45·2 % of females reporting isolation from friends and family compared to 33·9 % of men (p = 0·005). Multivariable logistic regression models revealed those aged 40–44, living rurally, and reporting an acute infection longer than seven days was associated with EVD-related stigma at the time of survey. This study demonstrates stigma is still prevalent among people who survived EVD in 2022. It also identified socio-demographic factors associated with stigma that can be used for targeting interventions. Importantly, this highlights the continued need for EVD survivors to access mental healthcare and social support systems well after disease recovery. This study was funded by the Canadian Institutes for Health Research (Grant no. PJT-175098. JK is funded by a Tier 2 Canada Research Chair in the Molecular Pathogenesis of Emerging and Re-Emerging Viruses. SS is funded by a Tier 2 Canada Research Chair in Program Science and Global Public Health. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Sudan virus disease – A quick review.
- Author
-
Tsou, Tsung-Pei
- Subjects
VIRUS diseases ,EBOLA virus disease ,COMMUNICABLE diseases ,VACCINE effectiveness ,DEATH rate ,EMERGING infectious diseases - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Development of an Immunochromatography Assay to Detect Marburg Virus and Ravn Virus.
- Author
-
Changula, Katendi, Kajihara, Masahiro, Muramatsu, Shino, Hiraoka, Koji, Yamaguchi, Toru, Yago, Yoko, Kato, Daisuke, Miyamoto, Hiroko, Mori-Kajihara, Akina, Shigeno, Asako, Yoshida, Reiko, Henderson, Corey W., Marzi, Andrea, and Takada, Ayato
- Subjects
- *
MARBURG virus , *RAPID diagnostic tests , *VIRAL antibodies , *VIRUS diseases , *TISSUE culture , *MONOCLONAL antibodies , *RAPID tooling - Abstract
The recent outbreaks of Marburg virus disease (MVD) in Guinea, Ghana, Equatorial Guinea, and Tanzania, none of which had reported previous outbreaks, imply increasing risks of spillover of the causative viruses, Marburg virus (MARV) and Ravn virus (RAVV), from their natural host animals. These outbreaks have emphasized the need for the development of rapid diagnostic tests for this disease. Using monoclonal antibodies specific to the viral nucleoprotein, we developed an immunochromatography (IC) assay for the rapid diagnosis of MVD. The IC assay was found to be capable of detecting approximately 102−4 50% tissue culture infectious dose (TCID50)/test of MARV and RAVV in the infected culture supernatants. We further confirmed that the IC assay could detect the MARV and RAVV antigens in the serum samples from experimentally infected nonhuman primates. These results indicate that the IC assay to detect MARV can be a useful tool for the rapid point-of-care diagnosis of MVD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. Characterization of the Cynomolgus Macaque Model of Marburg Virus Disease and Assessment of Timing for Therapeutic Treatment Testing.
- Author
-
Zumbrun, Elizabeth E., Garvey, Carly B., Wells, Jay B., Lynn, Ginger C., Van Tongeren, Sean, Steffens, Jesse T., Wetzel, Kelly S., Gomba, Laura M., O'Brien, Kristan A., Rossi, Franco D., Zeng, Xiankun, Lee, Eric D., Raymond, Jo Lynne W., Hoffman, Diana A., Jay, Alexandra N., Brown, Elizabeth S., Kallgren, Paul A., Norris, Sarah L., Cantey-Kiser, Jean, and Kudiya, Humza
- Subjects
- *
MARBURG virus , *MACAQUES , *VIRUS diseases , *CENTRAL venous catheters , *BLOOD collection , *TREATMENT effectiveness - Abstract
Marburg virus (MARV) causes severe disease and high mortality in humans. The objective of this study was to characterize disease manifestations and pathogenesis in cynomolgus macaques exposed to MARV. The results of this natural history study may be used to identify features of MARV disease useful in defining the ideal treatment initiation time for subsequent evaluations of investigational therapeutics using this model. Twelve cynomolgus macaques were exposed to a target dose of 1000 plaque-forming units MARV by the intramuscular route, and six control animals were mock-exposed. The primary endpoint of this study was survival to Day 28 post-inoculation (PI). Anesthesia events were minimized with the use of central venous catheters for periodic blood collection, and temperature and activity were continuously monitored by telemetry. All mock-exposed animals remained healthy for the duration of the study. All 12 MARV-exposed animals (100%) became infected, developed illness, and succumbed on Days 8–10 PI. On Day 4 PI, 11 of the 12 MARV-exposed animals had statistically significant temperature elevations over baseline. Clinically observable signs of MARV disease first appeared on Day 5 PI, when 6 of the 12 animals exhibited reduced responsiveness. Ultimately, systemic inflammation, coagulopathy, and direct cytopathic effects of MARV all contributed to multiorgan dysfunction, organ failure, and death or euthanasia of all MARV-exposed animals. Manifestations of MARV disease, including fever, systemic viremia, lymphocytolysis, coagulopathy, and hepatocellular damage, could be used as triggers for initiation of treatment in future therapeutic efficacy studies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
26. High-Avidity Anti-Filovirus IgG Elicited Using Protein Subunit Vaccines Does Not Correlate with Protection.
- Author
-
Williams, Caitlin A., Wong, Teri Ann S., Lieberman, Michael M., Yalley-Ogunro, Jake, Cabus, Mehtap, Nezami, Sara, Paz, Fabian, Andersen, Hanne, Geisbert, Thomas W., and Lehrer, Axel T.
- Subjects
- *
PEPTIDE vaccines , *EBOLA virus disease , *VACCINE effectiveness , *EBOLA virus , *HUMORAL immunity , *SPECIES specificity , *CELLULAR immunity - Abstract
Zaire ebolavirus (EBOV) poses a significant threat to public health due to its high case fatality rate and epidemic potential. This is further complicated by the lack of precise immune correlates of protection and difficulties in conducting in vivo animal studies due to species specificity of Ebola virus disease (EVD) and classification as a biosafety level 4 pathogen. Related ebolaviruses have also contributed to the public health threat; Uganda recently experienced an outbreak of Sudan ebolavirus, which also had a high case fatality rate. Vaccination targeting EBOV has demonstrated significant efficacy; however, the protective cellular and humoral responses at play are still poorly understood. Vaccination for vulnerable populations such as pregnant women, young children, and immunocompromised individuals is still limited. Understanding vaccine correlates of protection (vCOP) is key to developing alternative vaccination strategies for these groups. Components of immunity such as neutralizing antibody and cell-mediated immunity are likely responsible for protective responses; however, existing research fails to fully define their roles in protection. Here we investigated vaccine-elicited antibody avidity as a potential correlate of protection and to further characterize the contribution of antibody avidity in protective and nonprotective vaccine responses. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
27. Phylogenetic analysis of the promoter element 2 of paramyxo- and filoviruses
- Author
-
Shoichi Ashida, Shohei Kojima, Takashi Okura, Fumihiro Kato, Wakako Furuyama, Shuzo Urata, and Yusuke Matsumoto
- Subjects
paramyxovirus ,filovirus ,viral replication ,Microbiology ,QR1-502 - Abstract
ABSTRACTParamyxo- and filovirus genomes are equipped with bipartite promoters at their 3' ends to initiate RNA synthesis. The two elements, the primary promoter element 1 (PE1) and the secondary promoter element 2 (PE2), are separated by a spacer region that must be precisely a multiple of 6 nucleotides (nts), indicating these viruses adhere to the “rule of six.” However, our knowledge of PE2 has been limited to a narrow spectrum of virus species. In this study, a comparative analysis of 1,647 paramyxoviral genomes from a public database revealed that the paramyxovirus PE2 can be clearly categorized into two distinct subcategories: one marked by C repeats at every six bases (exclusive to the subfamily Orthoparamyxovirinae) and another characterized by CG repeats every 6 nts (observed in the subfamilies Avulavirinae and Rubulavirinae). This unique pattern collectively mirrors the evolutionary lineage of these subfamilies. Furthermore, we showed that PE2 of the Rubulavirinae, with the exception of mumps virus, serves as part of the gene-coding region. This may be due to the fact that the Rubulavirinae are the only paramyxoviruses that cannot propagate without RNA editing. Filoviruses have three to eight consecutive uracil repeats every six bases (UN5) in PE2, which is located in the 3' end region of the genome. We obtained PE2 sequences from 2,195 filoviruses in a public database and analyzed the sequence conservation among virus species. Our results indicate that the continuity of UN5 hexamers is consistently maintained with a high degree of conservation across virus species.IMPORTANCEThe genomic intricacies of paramyxo- and filoviruses are highlighted by the bipartite promoters—promoter element 1 (PE1) and promoter element 2 (PE2)—at their 3' termini. The spacer region between these elements follows the “rule of six,” crucial for genome replication. By a comprehensive analysis of paramyxoviral genome sequences, we identified distinct subcategories of PE2 based on C and CG repeats that were specific to Orthoparamyxovirinae and Avulavirinae/Rubulavirinae, respectively, mirroring their evolutionary lineages. Notably, the PE2 of Rubulavirinae is integrated into the gene-coding region, a unique trait potentially linked to its strict dependence on RNA editing for virus growth. This study also focused on the PE2 sequences in filovirus genomes. The strict conservation of the continuity of UN5 among virus species emphasizes its crucial role in viral genome replication.
- Published
- 2024
- Full Text
- View/download PDF
28. A novel MARV glycoprotein-specific antibody with potentials of broad-spectrum neutralization to filovirus
- Author
-
Yuting Zhang, Min Zhang, Haiyan Wu, Xinwei Wang, Hang Zheng, Junjuan Feng, Jing Wang, Longlong Luo, He Xiao, Chunxia Qiao, Xinying Li, Yuanqiang Zheng, Weijin Huang, Youchun Wang, Yi Wang, Yanchun Shi, Jiannan Feng, and Guojiang Chen
- Subjects
marburg ,neutralizing antibody ,filovirus ,glycoprotein ,NPC2 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Marburg virus (MARV) is one of the filovirus species that cause deadly hemorrhagic fever in humans, with mortality rates up to 90%. Neutralizing antibodies represent ideal candidates to prevent or treat virus disease. However, no antibody has been approved for MARV treatment to date. In this study, we identified a novel human antibody named AF-03 that targeted MARV glycoprotein (GP). AF-03 possessed a high binding affinity to MARV GP and showed neutralizing and protective activities against the pseudotyped MARV in vitro and in vivo. Epitope identification, including molecular docking and experiment-based analysis of mutated species, revealed that AF-03 recognized the Niemann-Pick C1 (NPC1) binding domain within GP1. Interestingly, we found the neutralizing activity of AF-03 to pseudotyped Ebola viruses (EBOV, SUDV, and BDBV) harboring cleaved GP instead of full-length GP. Furthermore, NPC2-fused AF-03 exhibited neutralizing activity to several filovirus species and EBOV mutants via binding to CI-MPR. In conclusion, this work demonstrates that AF-03 represents a promising therapeutic cargo for filovirus-caused disease.
- Published
- 2024
- Full Text
- View/download PDF
29. A mathematical model of Marburg virus disease outbreaks and the potential role of vaccination in control
- Author
-
George Y. Qian, W. John Edmunds, Daniel G. Bausch, and Thibaut Jombart
- Subjects
Marburg ,Marburgvirus ,Filovirus ,Vaccination ,Zoonotic ,Transmission ,Medicine - Abstract
Abstract Background Marburg virus disease is an acute haemorrhagic fever caused by Marburg virus. Marburg virus is zoonotic, maintained in nature in Egyptian fruit bats, with occasional spillover infections into humans and nonhuman primates. Although rare, sporadic cases and outbreaks occur in Africa, usually associated with exposure to bats in mines or caves, and sometimes with secondary human-to-human transmission. Outbreaks outside of Africa have also occurred due to importation of infected monkeys. Although all previous Marburg virus disease outbreaks have been brought under control without vaccination, there is nevertheless the potential for large outbreaks when implementation of public health measures is not possible or breaks down. Vaccines could thus be an important additional tool, and development of several candidate vaccines is under way. Methods We developed a branching process model of Marburg virus transmission and investigated the potential effects of several prophylactic and reactive vaccination strategies in settings driven primarily by multiple spillover events as well as human-to-human transmission. Linelist data from the 15 outbreaks up until 2022, as well as an Approximate Bayesian Computational framework, were used to inform the model parameters. Results Our results show a low basic reproduction number which varied across outbreaks, from 0.5 [95% CI 0.05–1.8] to 1.2 [95% CI 1.0–1.9] but a high case fatality ratio. Of six vaccination strategies explored, the two prophylactic strategies (mass and targeted vaccination of high-risk groups), as well as a combination of ring and targeted vaccination, were generally most effective, with a probability of potential outbreaks being terminated within 1 year of 0.90 (95% CI 0.90–0.91), 0.89 (95% CI 0.88–0.90), and 0.88 (95% CI 0.87–0.89) compared with 0.68 (0.67–0.69) for no vaccination, especially if the outbreak is driven by zoonotic spillovers and the vaccination campaign initiated as soon as possible after onset of the first case. Conclusions Our study shows that various vaccination strategies can be effective in helping to control outbreaks of MVD, with the best approach varying with the particular epidemiologic circumstances of each outbreak.
- Published
- 2023
- Full Text
- View/download PDF
30. Micro‒Global Positioning Systems for Identifying Nightly Opportunities for Marburg Virus Spillover to Humans by Egyptian Rousette Bats
- Author
-
Brian R. Amman, Amy J. Schuh, Gloria Akurut, Kilama Kamugisha, Dianah Namanya, Tara K. Sealy, James C. Graziano, Eric Enyel, Emily A. Wright, Stephen Balinandi, Julius J. Lutwama, Rebekah C. Kading, Patrick Atimnedi, and Jonathan S. Towner
- Subjects
Marburg virus ,Ravn virus ,Filovirus ,viruses ,Egyptian rousette bat ,Rousettus aegyptiacus ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
Marburg virus disease, caused by Marburg and Ravn orthomarburgviruses, emerges sporadically in sub-Saharan Africa and is often fatal in humans. The natural reservoir is the Egyptian rousette bat (ERB), which sheds virus in saliva, urine, and feces. Frugivorous ERBs discard test-bitten and partially eaten fruit, potentially leaving infectious virus behind that could be consumed by other susceptible animals or humans. Historically, 8 of 17 known Marburg virus disease outbreaks have been linked to human encroachment on ERB habitats, but no linkage exists for the other 9 outbreaks, raising the question of how bats and humans might intersect, leading to virus spillover. We used micro‒global positioning systems to identify nightly ERB foraging locations. ERBs from a known Marburg virus‒infected population traveled long distances to feed in cultivated fruit trees near homes. Our results show that ERB foraging behavior represents a Marburg virus spillover risk to humans and plausibly explains the origins of some past outbreaks.
- Published
- 2023
- Full Text
- View/download PDF
31. High-Avidity Anti-Filovirus IgG Elicited Using Protein Subunit Vaccines Does Not Correlate with Protection
- Author
-
Caitlin A. Williams, Teri Ann S. Wong, Michael M. Lieberman, Jake Yalley-Ogunro, Mehtap Cabus, Sara Nezami, Fabian Paz, Hanne Andersen, Thomas W. Geisbert, and Axel T. Lehrer
- Subjects
vaccines ,filovirus ,avidity ,nonhuman primates ,Medicine - Abstract
Zaire ebolavirus (EBOV) poses a significant threat to public health due to its high case fatality rate and epidemic potential. This is further complicated by the lack of precise immune correlates of protection and difficulties in conducting in vivo animal studies due to species specificity of Ebola virus disease (EVD) and classification as a biosafety level 4 pathogen. Related ebolaviruses have also contributed to the public health threat; Uganda recently experienced an outbreak of Sudan ebolavirus, which also had a high case fatality rate. Vaccination targeting EBOV has demonstrated significant efficacy; however, the protective cellular and humoral responses at play are still poorly understood. Vaccination for vulnerable populations such as pregnant women, young children, and immunocompromised individuals is still limited. Understanding vaccine correlates of protection (vCOP) is key to developing alternative vaccination strategies for these groups. Components of immunity such as neutralizing antibody and cell-mediated immunity are likely responsible for protective responses; however, existing research fails to fully define their roles in protection. Here we investigated vaccine-elicited antibody avidity as a potential correlate of protection and to further characterize the contribution of antibody avidity in protective and nonprotective vaccine responses.
- Published
- 2023
- Full Text
- View/download PDF
32. Efficacy and Immunogenicity of a Recombinant Vesicular Stomatitis Virus-Vectored Marburg Vaccine in Cynomolgus Macaques
- Author
-
Vidyleison N. Camargos, Shannan L. Rossi, Terry L. Juelich, Jennifer K. Smith, Nikos Vasilakis, Alexander N. Freiberg, Rick Nichols, and Joan Fusco
- Subjects
Marburg virus ,filovirus ,VSV ,vesicular stomatitis virus ,vaccine ,pseudotyped vector ,Microbiology ,QR1-502 - Abstract
Filoviruses, like the Marburg (MARV) and Ebola (EBOV) viruses, have caused outbreaks associated with significant hemorrhagic morbidity and high fatality rates. Vaccines offer one of the best countermeasures for fatal infection, but to date only the EBOV vaccine has received FDA licensure. Given the limited cross protection between the EBOV vaccine and Marburg hemorrhagic fever (MHF), we analyzed the protective efficacy of a similar vaccine, rVSV-MARV, in the lethal cynomolgus macaque model. NHPs vaccinated with a single dose (as little as 1.6 × 107 pfu) of rVSV-MARV seroconverted to MARV G-protein prior to challenge on day 42. Vaccinemia was measured in all vaccinated primates, self-resolved by day 14 post vaccination. Importantly, all vaccinated NHPs survived lethal MARV challenge, and showed no significant alterations in key markers of morbid disease, including clinical signs, and certain hematological and clinical chemistry parameters. Further, apart from one primate (from which tissues were not collected and no causal link was established), no pathology associated with Marburg disease was observed in vaccinated animals. Taken together, rVSV-MARV is a safe and efficacious vaccine against MHF in cynomolgus macaques.
- Published
- 2024
- Full Text
- View/download PDF
33. Non-Ebola Filoviruses: Potential Threats to Global Health Security
- Author
-
Yannick Munyeku-Bazitama, Francois Edidi-Atani, and Ayato Takada
- Subjects
Filovirus ,non-Ebola filovirus ,Sudan virus ,Marburg virus ,Ravn virus ,Reston virus ,Microbiology ,QR1-502 - Abstract
Filoviruses are negative-sense single-stranded RNA viruses often associated with severe and highly lethal hemorrhagic fever in humans and nonhuman primates, with case fatality rates as high as 90%. Of the known filoviruses, Ebola virus (EBOV), the prototype of the genus Orthoebolavirus, has been a major public health concern as it frequently causes outbreaks and was associated with an unprecedented outbreak in several Western African countries in 2013–2016, affecting 28,610 people, 11,308 of whom died. Thereafter, filovirus research mostly focused on EBOV, paying less attention to other equally deadly orthoebolaviruses (Sudan, Bundibugyo, and Taï Forest viruses) and orthomarburgviruses (Marburg and Ravn viruses). Some of these filoviruses have emerged in nonendemic areas, as exemplified by four Marburg disease outbreaks recorded in Guinea, Ghana, Tanzania, and Equatorial Guinea between 2021 and 2023. Similarly, the Sudan virus has reemerged in Uganda 10 years after the last recorded outbreak. Moreover, several novel bat-derived filoviruses have been discovered in the last 15 years (Lloviu virus, Bombali virus, Měnglà virus, and Dehong virus), most of which are poorly characterized but may display a wide host range. These novel viruses have the potential to cause outbreaks in humans. Several gaps are yet to be addressed regarding known and emerging filoviruses. These gaps include the virus ecology and pathogenicity, mechanisms of zoonotic transmission, host range and susceptibility, and the development of specific medical countermeasures. In this review, we summarize the current knowledge on non-Ebola filoviruses (Bombali virus, Bundibugyo virus, Reston virus, Sudan virus, Tai Forest virus, Marburg virus, Ravn virus, Lloviu virus, Měnglà virus, and Dehong virus) and suggest some strategies to accelerate specific countermeasure development.
- Published
- 2024
- Full Text
- View/download PDF
34. Comparison of Routes of Administration, Frequency, and Duration of Favipiravir Treatment in Mouse and Guinea Pig Models of Ebola Virus Disease
- Author
-
Dylan M. Johnson, Terry Juelich, Lihong Zhang, Jennifer K. Smith, Birte K. Kalveram, David Perez, Jeanon Smith, Michael R. Grimes, Tania Garron, Maricela Torres, Shane Massey, Trevor Brasel, David W. C. Beasley, Alex N. Freiberg, and Jason E. Comer
- Subjects
filovirus ,Ebola virus ,favipiravir ,T-705 ,EBOV ,animal model ,Microbiology ,QR1-502 - Abstract
Favipiravir is a ribonucleoside analogue that has been explored as a therapeutic for the treatment of Ebola Virus Disease (EVD). Promising data from rodent models has informed nonhuman primate trials, as well as evaluation in patients during the 2013–2016 West African EVD outbreak of favipiravir treatment. However, mixed results from these studies hindered regulatory approval of favipiravir for the indication of EVD. This study examined the influence of route of administration, duration of treatment, and treatment schedule of favipiravir in immune competent mouse and guinea pig models using rodent-adapted Zaire ebolavirus (EBOV). A dose of 300 mg/kg/day of favipiravir with an 8-day treatment was found to be fully effective at preventing lethal EVD-like disease in BALB/c mice regardless of route of administration (oral, intraperitoneal, or subcutaneous) or whether it was provided as a once-daily dose or a twice-daily split dose. Preclinical data generated in guinea pigs demonstrates that an 8-day treatment of 300 mg/kg/day of favipiravir reduces mortality following EBOV challenge regardless of route of treatment or duration of treatments for 8, 11, or 15 days. This work supports the future translational development of favipiravir as an EVD therapeutic.
- Published
- 2024
- Full Text
- View/download PDF
35. Viral Haemorrhagic Fevers
- Author
-
Gangopadhayya, Abhranil, Bhukya, Prudhvi Lal, Bhukya, Prudhvi Lal, editor, Mhaske, Suhas T., editor, and Sonkar, Subash C., editor
- Published
- 2023
- Full Text
- View/download PDF
36. Peripheral immune responses to filoviruses in a reservoir versus spillover hosts reveal transcriptional correlates of disease
- Author
-
Jonathan C. Guito, Catherine E. Arnold, Amy J. Schuh, Brian R. Amman, Tara K. Sealy, Jessica R. Spengler, Jessica R. Harmon, Joann D. Coleman-McCray, Mariano Sanchez-Lockhart, Gustavo F. Palacios, Jonathan S. Towner, and Joseph B. Prescott
- Subjects
viral reservoir ,spillover host ,bats ,Marburg virus ,filovirus ,immune response ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Several filoviruses, including Marburg virus (MARV), cause severe disease in humans and nonhuman primates (NHPs). However, the Egyptian rousette bat (ERB, Rousettus aegyptiacus), the only known MARV reservoir, shows no overt illness upon natural or experimental infection, which, like other bat hosts of zoonoses, is due to well-adapted, likely species-specific immune features. Despite advances in understanding reservoir immune responses to filoviruses, ERB peripheral blood responses to MARV and how they compare to those of diseased filovirus-infected spillover hosts remain ill-defined. We thus conducted a longitudinal analysis of ERB blood gene responses during acute MARV infection. These data were then contrasted with a compilation of published primate blood response studies to elucidate gene correlates of filovirus protection versus disease. Our work expands on previous findings in MARV-infected ERBs by supporting both host resistance and disease tolerance mechanisms, offers insight into the peripheral immunocellular repertoire during infection, and provides the most direct known cross-examination between reservoir and spillover hosts of the most prevalently-regulated response genes, pathways and activities associated with differences in filovirus pathogenesis and pathogenicity.
- Published
- 2024
- Full Text
- View/download PDF
37. Structural insights on the nucleoprotein C-terminal domain of Měnglà virus
- Author
-
Diego Sebastian Ferrero, Omar Tomás Gilabert, and Nuria Verdaguer
- Subjects
filovirus ,inclusion body ,Měnglà virus ,nucleoprotein ,Microbiology ,QR1-502 - Abstract
ABSTRACT Filoviruses depend on the nucleoprotein (NP) to accomplish multiple functions during the viral life cycle. NP is the most abundantly expressed viral protein in infected cells and the main component of the viral nucleocapsid. It can be structurally divided into amino- and carboxy- terminal domains (NTD and CTD). The NTD can homo-oligomerize to interact and protect the (−) ssRNA genome, forming long helical structures. The flexible CTD is responsible for the binding of other nucleocapsid proteins and is involved in the formation of inclusion bodies (IBs)—the cytoplasmic sites of nucleocapsid formation and genome replication. The CTD ends in a ~100-residue globular tail. Měnglà virus (MLAV) is the only member of the new Dianlovirus genus within the Filoviridae family. Their differential characteristics and the possibility of becoming a threat for human health justify the interest in better understanding of its structure and function. In this work, we present the structure of the globular tail of the MLAV NP CTD, showing an overall conformation closely related to that previously reported for the equivalent NP region in MARV. Moreover, analyses of the CTD-CTD interactions in the crystal asymmetric unit revealed a higher-order helicoidal structure. Mutational studies underscore the crucial role of a number of residues, located at the CTD-CTD contact interface, for IB formation. Site-directed mutagenesis of amino acids L653 and F687, involved in the formation of these helicoidal assemblies, abrogate the growth of IBs when the full-length MLAV NP was ectopically expressed in cells. Our findings confirm the role of NP CTD in IB formation, also for MLAV, and focus the attention on particular residues as starting point for further analysis. IMPORTANCE Filoviruses are the causative agents of severe and often fatal hemorrhagic disease in humans. Měnglà virus (MLAV) is a recently reported filovirus, isolated from fruit bats that is capable to replicate in human cells, representing a potential risk for human health. An in-depth structural and functional knowledge of MLAV proteins is an essential step for antiviral research on this virus that can also be extended to other emerging filoviruses. In this study, we determined the first crystal structures of the C-terminal domain (CTD) of the MLAV nucleoprotein (NP), showing important similarities to the equivalent domain in MARV. The structural data also show that the NP CTD has the ability to form large helical oligomers that may participate in the control of cytoplasmic inclusion body formation during viral replication.
- Published
- 2023
- Full Text
- View/download PDF
38. Transcriptional profiling of immune responses in NHPs after low-dose, VSV-based vaccination against Marburg virus
- Author
-
Cecilia A. Prator, Brianna M. Dorratt, Kyle L. O’Donnell, Justin Lack, Amanda N. Pinski, Stacy Ricklefs, Craig A. Martens, Ilhem Messaoudi, and Andrea Marzi
- Subjects
Filovirus ,Marburg virus Angola ,MARV ,vesicular stomatitis virus ,nonhuman primate ,host response ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
Infection with Marburg virus (MARV), the causative agent of Marburg virus disease (MVD), results in haemorrhagic disease and high case fatality rates (>40%) in humans. Despite its public health relevance, there are no licensed vaccines or therapeutics to prevent or treat MVD. A vesicular stomatitis virus (VSV)-based vaccine expressing the MARV glycoprotein (VSV-MARV) is currently in clinical development. Previously, a single 10 million PFU dose of VSV-MARV administered 1–5 weeks before lethal MARV challenge conferred uniform protection in nonhuman primates (NHPs), demonstrating fast-acting potential. Additionally, our group recently demonstrated that even a low dose VSV-MARV (1000 PFU) protected NHPs when given 7 days before MARV challenge. In this study, we longitudinally profiled the transcriptional responses of NHPs vaccinated with this low dose of VSV-MARV either 14 or 7 days before lethal MARV challenge. NHPs vaccinated 14 days before challenge presented with transcriptional changes consistent with an antiviral response before challenge. Limited gene expression changes were observed in the group vaccinated 7 days before challenge. After challenge, genes related to lymphocyte-mediated immunity were only observed in the group vaccinated 14 days before challenge, indicating that the length of time between vaccination and challenge influenced gene expression. Our results indicate that a low dose VSV-MARV elicits distinct immune responses that correlate with protection against MVD. A low dose of VSV-MARV should be evaluated in clinical rails as it may be an option to deliver beneficial public health outcomes to more people in the event of future outbreaks.
- Published
- 2023
- Full Text
- View/download PDF
39. Single-dose VSV-based vaccine protects cynomolgus macaques from disease after Taï Forest virus infection
- Author
-
Paige Fletcher, Kyle L. O’Donnell, Brianna M. Doratt, Delphine C. Malherbe, Chad S. Clancy, Joseph F. Rhoderick, Friederike Feldmann, Patrick W. Hanley, Thomas G. Ksiazek, Thomas W. Geisbert, Ilhem Messaoudi, and Andrea Marzi
- Subjects
Filovirus ,Taï forest ebolavirus ,vesicular stomatitis virus ,live-attenuated vaccine ,nonhuman primate ,transcriptomics ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
Taï Forest virus (TAFV) is a lesser-known ebolavirus that causes lethal infections in chimpanzees and is responsible for a single human case. Limited research has been done on this human pathogen; however, with the recent emergence of filoviruses in West Africa, further investigation and countermeasure development against this virus is warranted. We developed a vesicular stomatitis virus (VSV)-based vaccine expressing the TAFV glycoprotein as the viral antigen and assessed it for protective efficacy in nonhuman primates (NHPs). Following a single high-dose vaccination, NHPs developed antigen-specific binding and neutralizing antibodies as well as modest T cell responses. Importantly, all vaccinated NHPs were uniformly protected from disease after lethal TAFV challenge while the naïve control group succumbed to the disease. Histopathologic lesions consistent with filovirus disease were present in control NHPs but were not observed in vaccinated NHPs. Transcriptional analysis of whole blood samples obtained after vaccination and challenge was performed to gain insight into molecular underpinnings conferring protection. Differentially expressed genes (DEG) detected 7 days post-vaccination were enriched to processes associated with innate immunity and antiviral responses. Only a small number of DEG was detected in vaccinated NHPs post-challenge while over 1,000 DEG were detected in control NHPs at end-stage disease which mapped to gene ontology terms indicative of defense responses and inflammation. Taken together, this data demonstrates the effective single-dose protection of the VSV-TAFV vaccine, and its potential for use in outbreaks.
- Published
- 2023
- Full Text
- View/download PDF
40. N6-methyladenosine is required for efficient RNA synthesis of Ebola virus and other haemorrhagic fever viruses
- Author
-
Lisa Wendt, Matthew J. Pickin, Bianca S. Bodmer, Sven Reiche, Lucie Fénéant, Julia E. Hölper, Walter Fuchs, Allison Groseth, and Thomas Hoenen
- Subjects
Ebola virus ,Junín virus ,Crimean-Congo haemorrhagic fever virus ,filovirus ,arenavirus ,orthonairovirus ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
N6-methyladenosine (m6A) is one of the most abundant modifications of cellular RNA, where it serves various functions. m6A methylation of many viral RNA species has also been described; however, little is known about the m6A epitranscriptome of haemorrhagic fever-causing viruses like Ebola virus (EBOV). Here, we analysed the importance of the methyltransferase METTL3 for the life cycle of this virus. We found that METTL3 interacts with the EBOV nucleoprotein and the transcriptional activator VP30 to support viral RNA synthesis, and that METTL3 is recruited into EBOV inclusions bodies, where viral RNA synthesis occurs. Analysis of the m6A methylation pattern of EBOV mRNAs showed that they are methylated by METTL3. Further studies revealed that METTL3 interaction with the viral nucleoprotein, as well as its importance for RNA synthesis and protein expression, is also observed for other haemorrhagic fever viruses such as Junín virus (JUNV) and Crimean-Congo haemorrhagic fever virus (CCHFV). The negative effects on viral RNA synthesis due to loss of m6A methylation are independent of innate immune sensing, as METTL3 knockout did not affect type I interferon induction in response to viral RNA synthesis or infection. Our results suggest a novel function for m6A that is conserved among diverse haemorrhagic fever-causing viruses (i.e. EBOV, JUNV and CCHFV), making METTL3 a promising target for broadly-acting antivirals.
- Published
- 2023
- Full Text
- View/download PDF
41. In vivo characterization of the novel ebolavirus Bombali virus suggests a low pathogenic potential for humans
- Author
-
B. S. Bodmer, A. Breithaupt, M. Heung, J. E. Brunetti, C. Henkel, J. Müller-Guhl, E. Rodríguez, L. Wendt, S. L. Winter, M. Vallbracht, A. Müller, S. Römer, P. Chlanda, C. Muñoz-Fontela, T. Hoenen, and B. Escudero-Pérez
- Subjects
Bombali virus ,ebolavirus ,filovirus ,virulence ,pathogenic potential ,reverse genetics ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
ABSTRACTEbolaviruses cause outbreaks of haemorrhagic fever in Central and West Africa. Some members of this genus such as Ebola virus (EBOV) are highly pathogenic, with case fatality rates of up to 90%, whereas others such as Reston virus (RESTV) are apathogenic for humans. Bombali virus (BOMV) is a novel ebolavirus for which complete genome sequences were recently found in free-tailed bats, although no infectious virus could be isolated. Its pathogenic potential for humans is unknown. To address this question, we first determined whether proteins encoded by the available BOMV sequence found in Chaerephon pumilus were functional in in vitro assays. The correction of an apparent sequencing error in the glycoprotein based on these data then allowed us to generate infectious BOMV using reverse genetics and characterize its infection of human cells. Furthermore, we used HLA-A2-transgenic, NOD-scid-IL-2γ receptor-knockout (NSG-A2) mice reconstituted with human haematopoiesis as a model to evaluate the pathogenicity of BOMV in vivo in a human-like immune environment. These data demonstrate that not only does BOMV show a slower growth rate than EBOV in vitro, but it also shows low pathogenicity in humanized mice, comparable to previous studies using RESTV. Taken together, these findings suggest a low pathogenic potential of BOMV for humans.
- Published
- 2023
- Full Text
- View/download PDF
42. Clinical and Immunologic Correlates of Vasodilatory Shock Among Ebola Virus–Infected Nonhuman Primates in a Critical Care Model.
- Author
-
Stein, Sydney R, Platt, Andrew P, Teague, Heather L, Anthony, Scott M, Reeder, Rebecca J, Cooper, Kurt, Byrum, Russell, Drawbaugh, David J, Liu, David X, Burdette, Tracey L, Hadley, Kyra, Barr, Bobbi, Warner, Seth, Rodriguez-Hernandez, Francisco, Johnson, Cristal, Stanek, Phil, Hischak, Joseph, Kendall, Heather, Huzella, Louis M, and Strich, Jeffrey R
- Subjects
- *
MONONUCLEAR leukocytes , *EBOLA virus disease , *KILLER cells , *CRITICAL care medicine , *EBOLA virus - Abstract
Background Existing models of Ebola virus infection have not fully characterized the pathophysiology of shock in connection with daily virologic, clinical, and immunologic parameters. We implemented a nonhuman primate critical care model to investigate these associations. Methods Two rhesus macaques received a target dose of 1000 plaque-forming units of Ebola virus intramuscularly with supportive care initiated on day 3. High-dimensional spectral cytometry was used to phenotype neutrophils and peripheral blood mononuclear cells daily. Results We observed progressive vasodilatory shock with preserved cardiac function following viremia onset on day 5. Multiorgan dysfunction began on day 6 coincident with the nadir of circulating neutrophils. Consumptive coagulopathy and anemia occurred on days 7 to 8 along with irreversible shock, followed by death. The monocyte repertoire began shifting on day 4 with a decline in classical and expansion of double-negative monocytes. A selective loss of CXCR3-positive B and T cells, expansion of naive B cells, and activation of natural killer cells followed viremia onset. Conclusions Our model allows for high-fidelity characterization of the pathophysiology of acute Ebola virus infection with host innate and adaptive immune responses, which may advance host-targeted therapy design and evaluation for use after the onset of multiorgan failure. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
43. AAV-Vectored Expression of Marburg Virus–Neutralizing Antibody MR191 Provides Complete Protection From Challenge in a Guinea Pig Model.
- Author
-
Rghei, Amira D, Cao, Wenguang, He, Shihua, Lopes, Jordyn A, Zielinska, Nicole, Pei, Yanlong, Thompson, Brad, Banadyga, Logan, and Wootton, Sarah K
- Subjects
- *
GUINEA pigs , *MARBURG virus , *MONOCLONAL antibodies , *ADENO-associated virus , *B cells - Abstract
Although there are no approved countermeasures available to prevent or treat disease caused by Marburg virus (MARV), potently neutralizing monoclonal antibodies (mAbs) derived from B cells of human survivors have been identified. One such mAb, MR191, has been shown to provide complete protection against MARV in nonhuman primates. We previously demonstrated that prophylactic administration of an adeno-associated virus (AAV) expressing MR191 protected mice from MARV. Here, we modified the AAV-MR191 coding sequence to enhance efficacy and reevaluated protection in a guinea pig model. Remarkably, 4 different variants of AAV-MR191 provided complete protection against MARV, despite administration 90 days prior to challenge. Based on superior expression kinetics, AAV-MR191-io2, was selected for evaluation in a dose-reduction experiment. The highest dose provided 100% protection, while a lower dose provided ∼88% protection. These data confirm the efficacy of AAV-mediated expression of MR191 and support the further development of this promising MARV countermeasure. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
44. The Multiple Origins of Ebola Disease Outbreaks.
- Author
-
Judson, Seth D and Munster, Vincent J
- Subjects
- *
DISEASE outbreaks , *EBOLA virus disease , *EBOLA virus , *EPIDEMIOLOGY - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
45. A Single Case Observation: Is the Ebola Virus Soluble Glycoprotein an Indicator of Viral Recrudescence?
- Author
-
Furuyama, Wakako, Davey, Richard T, Chertow, Daniel S, and Marzi, Andrea
- Subjects
- *
EBOLA virus , *EBOLA virus disease , *POLYMERASE chain reaction - Abstract
This case study investigated the long-term expression dynamics of Ebola virus (EBOV) soluble glycoprotein (sGP) in the serum of a patient who was infected with EBOV in West Africa and recovered from acute Ebola virus disease (EVD) at the National Institutes of Health Clinical Center in Bethesda, Maryland. Samples from this patient were collected during acute EVD and during convalescence up to day 361 following illness onset. Although blood samples were negative by reverse transcription–quantitative polymerase chain reaction after recovery from acute EVD, we detected small amounts of EBOV sGP in the serum of the patient long after recovery, potentially indicating viral recrudescence. As this was only observed in a single patient, additional longitudinal patient samples are needed to confirm our hypothesis that EBOV sGP may be an indicator of viral recrudescence long after recovery from acute EVD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. Atypical Ebola Virus Disease in a Rhesus Macaque.
- Author
-
Marzi, Andrea, Hanley, Patrick W, Furuyama, Wakako, Haddock, Elaine, Martens, Craig A, Scott, Dana P, and Feldmann, Heinz
- Subjects
- *
EBOLA virus disease , *MEDICAL personnel , *RHESUS monkeys , *GENITALIA , *EBOLA virus , *FOREIGN bodies - Abstract
Ebola virus (EBOV)–Makona infected more than 30 000 people from 2013 to 2016 in West Africa, among them many health care workers including foreign nationals. Most of the infected foreign nationals were evacuated and treated in their respective home countries, resulting in detailed reports of the acute disease following EBOV infection as well as descriptions of symptoms now known as post-Ebola syndrome, which occurred months after the infection. Symptoms associated with this syndrome include uveitis and neurological manifestations. In 1 of our EBOV-Makona nonhuman primate (NHP) studies, 1 NHP was euthanized on day 28 after infection having completely recovered from the acute disease. During convalescence, this NHP developed neurological signs and acute respiratory distress requiring euthanasia. The organ tropism had changed with high virus titers in lungs, brain, eye, and reproductive organs but no virus in the typical target organs for acute EBOV infection. This in part reflects sequelae described for EBOV survivors albeit developing quicker after recovery from acute disease. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
47. Natural History of Nonhuman Primates After Oral Exposure to Ebola Virus Variant Makona.
- Author
-
Prasad, Abhishek N, Agans, Krystle N, Geisbert, Joan B, Borisevich, Viktoriya, Deer, Daniel J, Dobias, Natalie S, Comer, Jason E, Woolsey, Courtney, Fenton, Karla A, Geisbert, Thomas W, and Cross, Robert W
- Subjects
- *
EBOLA virus , *NATURAL history , *EBOLA virus disease , *PRIMATES , *KRA , *LYMPHOPENIA - Abstract
Background The primary route of infection by Ebola virus (EBOV) is through contact of mucosal surfaces. Few studies have explored infection of nonhuman primates (NHPs) via the oral mucosa, which is a probable portal of natural infection in humans. Methods To further characterize the pathogenesis of EBOV infection via the oral exposure route, we challenged cohorts of cynomolgus monkeys with low doses of EBOV variant Makona. Results Infection with 100 or 50 PFU of EBOV Makona via the oral route resulted in 50% and 83% lethality, respectively. Animals that progressed to fatal disease exhibited lymphopenia, marked coagulopathy, high viral loads, and increased levels of serum markers of inflammation and hepatic/renal injury. Survival in these cohorts was associated with milder fluctuations in leukocyte populations, lack of coagulopathy, and reduced or absent serum markers of inflammation and/or hepatic/renal function. Surprisingly, 2 surviving animals from the 100- and 50-PFU cohorts developed transient low-level viremia in the absence of other clinical signs of disease. Conversely, all animals in the 10 PFU cohort remained disease free and survived to the study end point. Conclusions Our observations highlight the susceptibility of NHPs, and by extension, likely humans, to relatively low doses of EBOV via the oral route. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
48. A Recombinant Vesicular Stomatitis Virus–Based Vaccine Provides Postexposure Protection Against Bundibugyo Ebolavirus Infection.
- Author
-
Woolsey, Courtney, Strampe, Jamie, Fenton, Karla A, Agans, Krystle N, Martinez, Jasmine, Borisevich, Viktoriya, Dobias, Natalie S, Deer, Daniel J, Geisbert, Joan B, Cross, Robert W, Connor, John H, and Geisbert, Thomas W
- Subjects
- *
VESICULAR stomatitis , *EBOLA virus , *VACCINES , *INFECTION , *SURVIVAL rate - Abstract
Background The filovirus Bundibugyo virus (BDBV) causes severe disease with a mortality rate of approximately 20%–51%. The only licensed filovirus vaccine in the United States, Ervebo, consists of a recombinant vesicular stomatitis virus (rVSV) vector that expresses Ebola virus (EBOV) glycoprotein (GP). Ervebo was shown to rapidly protect against fatal Ebola disease in clinical trials; however, the vaccine is only indicated against EBOV. Recent outbreaks of other filoviruses underscore the need for additional vaccine candidates, particularly for BDBV infections. Methods To examine whether the rVSV vaccine candidate rVSVΔG/BDBV-GP could provide therapeutic protection against BDBV, we inoculated seven cynomolgus macaques with 1000 plaque-forming units of BDBV, administering rVSVΔG/BDBV-GP vaccine to 6 of them 20–23 minutes after infection. Results Five of the treated animals survived infection (83%) compared to an expected natural survival rate of 21% in this macaque model. All treated animals showed an early circulating immune response, while the untreated animal did not. Surviving animals showed evidence of both GP-specific IgM and IgG production, while animals that succumbed did not produce significant IgG. Conclusions This small, proof-of-concept study demonstrated early treatment with rVSVΔG/BDBV-GP provides a survival benefit in this nonhuman primate model of BDBV infection, perhaps through earlier initiation of adaptive immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
49. Establishing a Mouse Model for Sexual Transmission and Male Reproductive Tract Persistence of Ebola Virus.
- Author
-
Clancy, Chad S, Smart, Gabrielle, Rhoderick, J Fred, O'Donnell, Kyle L, Rosenke, Rebecca, Schäfer, Alexandra, and Marzi, Andrea
- Subjects
- *
MALE reproductive organs , *EBOLA virus , *EBOLA virus disease , *GENITALIA infections , *LABORATORY mice - Abstract
Ebola virus disease (EVD) has resulted in the death of over 15 000 people since its discovery in 1976. At least 1 incident of re-emergence of EVD has been associated with persistent male reproductive tract infection in a patient surviving EVD greater than 500 days prior. To date, animal models of Ebola virus (EBOV) infection have failed to fully characterize the pathogenesis of reproductive tract infection. Furthermore, no animal model of sexual transmission of EBOV exists. In this study, we describe a roadmap to modeling sexual transmission of EBOV using a mouse-adapted EBOV isolate in immunocompetent male mice and female Ifnar −/− mice. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
50. Single-Dose Treatment With Vesicular Stomatitis Virus–Based Ebola Virus Vaccine Expressing Ebola Virus–Specific Artificial Micro-RNA Does Not Protect Mice From Lethal Disease.
- Author
-
O'Donnell, Kyle L, Callison, Julie, Feldmann, Heinz, Hoenen, Thomas, and Marzi, Andrea
- Subjects
- *
EBOLA virus , *VESICULAR stomatitis , *VIRAL vaccines , *EBOLA virus disease , *MICRORNA - Abstract
Although significant progress has been made in the development of therapeutics against Ebola virus (EBOV), we sought to expand upon existing strategies and combine an RNA interference–based intervention with the approved vesicular stomatitis virus–based Ebola virus (VSV-EBOV) vaccine to conjointly treat and vaccinate patients during an outbreak. We constructed VSV-EBOV vectors expressing artificial micro-RNAs (amiRNAs) targeting sequences of EBOV proteins. In vitro experiments demonstrated a robust decrease in EBOV replication using a minigenome system and infectious virus. For in vivo evaluation, mouse-adapted EBOV–infected CD-1 mice were treated 24 hours after infection with a single dose of the VSV-EBOV amiRNA constructs. We observed no difference in disease progression or survival compared to the control-treated mice. In summary, while amiRNAs decrease viral replication in vitro , the effect is not sufficient to protect mice from lethal disease, and this therapeutic approach requires further optimization. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.