Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Vaccine ; 2024 May 19.
Article in English | MEDLINE | ID: mdl-38769033

ABSTRACT

The emergence and ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the need for rapid vaccine development platforms that can be updated to counteract emerging variants of currently circulating and future emerging coronaviruses. Here we report the development of a "train model" subunit vaccine platform that contains a SARS-CoV-2 Wuhan S1 protein (the "engine") linked to a series of flexible receptor binding domains (RBDs; the "cars") derived from SARS-CoV-2 variants of concern (VOCs). We demonstrate that these linked subunit vaccines when combined with Sepivac SWE™, a squalene in water emulsion (SWE) adjuvant, are immunogenic in Syrian hamsters and subsequently provide protection from infection with SARS-CoV-2 VOCs Omicron (BA.1), Delta, and Beta. Importantly, the bivalent and trivalent vaccine candidates offered protection against some heterologous SARS-CoV-2 VOCs that were not included in the vaccine design, demonstrating the potential for broad protection against a range of different VOCs. Furthermore, these formulated vaccine candidates were stable at 2-8 °C for up to 13 months post-formulation, highlighting their utility in low-resource settings. Indeed, our vaccine platform will enable the development of safe and broadly protective vaccines against emerging betacoronaviruses that pose a significant health risk for humans and agricultural animals.

2.
Front Microbiol ; 14: 1148255, 2023.
Article in English | MEDLINE | ID: mdl-37065160

ABSTRACT

The ongoing evolution of SARS-CoV-2 continues to raise new questions regarding the duration of immunity to reinfection with emerging variants. To address these knowledge gaps, controlled investigations in established animal models are needed to assess duration of immunity induced by each SARS-CoV-2 lineage and precisely evaluate the extent of cross-reactivity and cross-protection afforded. Using the Syrian hamster model, we specifically investigated duration of infection acquired immunity to SARS-CoV-2 ancestral Wuhan strain over 12 months. Plasma spike- and RBD-specific IgG titers against ancestral SARS-CoV-2 peaked at 4 months post-infection and showed a modest decline by 12 months. Similar kinetics were observed with plasma virus neutralizing antibody titers which peaked at 2 months post-infection and showed a modest decline by 12 months. Reinfection with ancestral SARS-CoV-2 at regular intervals demonstrated that prior infection provides long-lasting immunity as hamsters were protected against severe disease when rechallenged at 2, 4, 6, and 12 months after primary infection, and this coincided with the induction of high virus neutralizing antibody titers. Cross-neutralizing antibody titers against the B.1.617.2 variant (Delta) progressively waned in blood over 12 months, however, re-infection boosted these titers to levels equivalent to ancestral SARS-CoV-2. Conversely, cross-neutralizing antibodies to the BA.1 variant (Omicron) were virtually undetectable at all time-points after primary infection and were only detected following reinfection at 6 and 12 months. Collectively, these data demonstrate that infection with ancestral SARS-CoV-2 strains generates antibody responses that continue to evolve long after resolution of infection with distinct kinetics and emergence of cross-reactive and cross-neutralizing antibodies to Delta and Omicron variants and their specific spike antigens.

3.
Pathogens ; 11(4)2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35456124

ABSTRACT

Under experimental conditions, pigs infected with Ebola Virus (EBOV) develop disease and can readily transmit the virus to non-human primates or pigs. In the event of accidental or intentional EBOV infection of domestic pigs, complex and time-consuming safe depopulation and carcass disposal are expected. Delaying or preventing transmission through a reduction in viral shedding is an absolute necessity to limit the spread of the virus. In this study, we tested whether porcine interferon-α or λ3 (porIFNα or porIFNλ3) delivered by a replication-defective human type 5 adenovirus vector (Ad5-porIFNα or Ad5-porIFNλ3) could limit EBOV replication and shedding in domestic pigs. Our results show that pigs pre-treated with Ad5-porIFNα did not develop measurable clinical signs, did not shed virus RNA, and displayed strongly reduced viral RNA load in tissues. A microarray analysis of peripheral blood mononuclear cells indicated that Ad5-porIFNα treatment led to clear upregulation in immune and inflammatory responses probably involved in protection against disease. Our results indicate that administration of Ad5-porIFNα can potentially be used to limit the spread of EBOV in pigs.

4.
Med ; 3(6): 422-432.e3, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35437520

ABSTRACT

Background: SARS-CoV-2 Omicron variant of concern (VOC) has evolved multiple mutations within the spike protein, raising concerns of increased antibody evasion. In this study, we assessed the neutralization potential of COVID-19 convalescent sera and sera from vaccinated individuals against ancestral SARS-CoV-2 and VOCs. Methods: The neutralizing activity of sera from 65 coronavirus disease (COVID-19) vaccine recipients and convalescent individuals against clinical isolates of ancestral SARS-CoV-2 and Beta, Delta, and Omicron VOCs was assessed using a micro-neutralization assay. Findings: Convalescent sera from unvaccinated individuals infected by the ancestral virus demonstrated reduced neutralization against Beta and Omicron VOCs. Sera from individuals that received three doses of the Pfizer or Moderna vaccines demonstrated reduced neutralization of the Omicron variant relative to ancestral SARS-CoV-2. Sera from individuals that were naturally infected with ancestral SARS-CoV-2 and subsequently received two doses of the Pfizer vaccine induced significantly higher neutralizing antibody levels against ancestral virus and all VOCs. Infection alone, either with ancestral SARS-CoV-2 or the Delta variant, was not sufficient to induce high neutralizing antibody titers against Omicron. Conclusions: In summary, we demonstrate that convalescent and vaccinated sera display varying levels of SARS-CoV-2 VOC neutralization. Data from this study will inform booster vaccination strategies against SARS-CoV-2 VOCs. Funding: This research was funded by the Canadian Institutes of Health Research (CIHR). VIDO receives operational funding from the Government of Saskatchewan through Innovation Saskatchewan and the Ministry of Agriculture and from the Canada Foundation for Innovation through the Major Science Initiatives for its CL3 facility.


Subject(s)
COVID-19 , Spike Glycoprotein, Coronavirus , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19/therapy , Humans , Immunization, Passive , Membrane Glycoproteins/genetics , Neutralization Tests , SARS-CoV-2/genetics , Saskatchewan , Spike Glycoprotein, Coronavirus/genetics , Viral Envelope Proteins/genetics , COVID-19 Serotherapy
5.
PLoS Pathog ; 17(7): e1009705, 2021 07.
Article in English | MEDLINE | ID: mdl-34265022

ABSTRACT

COVID-19 (coronavirus disease 2019) caused by SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection is a disease affecting several organ systems. A model that captures all clinical symptoms of COVID-19 as well as long-haulers disease is needed. We investigated the host responses associated with infection in several major organ systems including the respiratory tract, the heart, and the kidneys after SARS-CoV-2 infection in Syrian hamsters. We found significant increases in inflammatory cytokines (IL-6, IL-1beta, and TNF) and type II interferons whereas type I interferons were inhibited. Examination of extrapulmonary tissue indicated inflammation in the kidney, liver, and heart which also lacked type I interferon upregulation. Histologically, the heart had evidence of myocarditis and microthrombi while the kidney had tubular inflammation. These results give insight into the multiorgan disease experienced by people with COVID-19 and possibly the prolonged disease in people with post-acute sequelae of SARS-CoV-2 (PASC).


Subject(s)
COVID-19/immunology , Down-Regulation/immunology , Interferon Type I/immunology , Kidney/immunology , Myocardium/immunology , Respiratory System/immunology , SARS-CoV-2/immunology , Animals , COVID-19/pathology , Cricetinae , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/pathology , Kidney/pathology , Kidney/virology , Male , Mesocricetus , Myocardium/pathology , Respiratory System/pathology , Respiratory System/virology
6.
Viruses ; 13(4)2021 04 15.
Article in English | MEDLINE | ID: mdl-33920917

ABSTRACT

Many factors impact the host response to influenza virus infection and vaccination. Ferrets have been an indispensable reagent for influenza virus research for almost one hundred years. One of the most significant and well-known factors affecting human disease after infection is host age. Another significant factor is the virus, as strain-specific disease severity is well known. Studying age-related impacts on viral infection and vaccination outcomes requires an animal model that reflects both the physiological and immunological changes that occur with human aging, and sensitivity to differentially virulent influenza viruses. The ferret is uniquely susceptible to a plethora of influenza viruses impacting humans and has proven extremely useful in studying the clinical and immunological pictures of influenza virus infection. Moreover, ferrets developmentally have several of the age-related physiological changes that occur in humans throughout infancy, adulthood, old age, and pregnancy. In this review, we discuss ferret susceptibility to influenza viruses, summarize previous influenza studies using ferrets as models of age, and finally, highlight the application of ferret age models in the pursuit of prophylactic and therapeutic agents to address age-related influenza disease severity.


Subject(s)
Ferrets/virology , Immunity , Orthomyxoviridae Infections/virology , Age Factors , Animals , Female , Humans , Influenza Vaccines , Pregnancy , Risk Factors , Vaccination
7.
Front Vet Sci ; 7: 238, 2020.
Article in English | MEDLINE | ID: mdl-32528981

ABSTRACT

Since the discovery of Rift Valley Fever virus (RVFV) in Kenya in 1930, the virus has become widespread throughout most of Africa and is characterized by sporadic outbreaks. A mosquito-borne pathogen, RVFV is poised to move beyond the African continent and the Middle East and emerge in Europe and Asia. There is a risk that RVFV could also appear in the Americas, similar to the West Nile virus. In light of this potential threat, multiple studies have been undertaken to establish international surveillance programs and diagnostic tools, develop models of transmission dynamics and risk factors for infection, and to develop a variety of vaccines as countermeasures. Furthermore, considerable efforts to establish reliable challenge models of Rift Valley fever virus have been made and platforms for testing potential vaccines and therapeutics in target species have been established. This review emphasizes the progress and insights from a North American perspective to establish challenge models in target livestock such as cattle, sheep, and goats in comparisons to other researchers' reports. A brief summary of the potential role of wildlife, such as buffalo and white-tailed deer as reservoir species will also be discussed.

8.
Front Vet Sci ; 7: 137, 2020.
Article in English | MEDLINE | ID: mdl-32411730

ABSTRACT

Rift Valley Fever virus (RVFV) is a zoonotic mosquito-borne virus that belongs to the Phenuiviridae family. Infections in animal herds cause abortion storms, high mortality rates in neonates, and mild to severe symptoms. Infected animals can also transmit the virus to people, particularly people who live or work in close contact with livestock. There is currently an ongoing effort to produce safe and efficacious veterinary vaccines against RVFV in livestock to protect against both primary infection in animals and zoonotic infections in people. To test the efficacy of these vaccines it is essential to have a reliable challenge model in relevant target species, including ruminants. In this study we evaluated three routes of inoculation (intranasal, intradermal and a combination of routes) in Holstein cattle using an infectious dose of 107 pfu/ml and a virus strain from the 2006-2007 outbreak in Kenya and Sudan. Our results demonstrated that all routes of inoculation were effective at producing viremia in all animals; however, the intranasal route induced the highest levels and longest duration of viremia, the most noticeable clinical signs, and the most widespread infection of tissues. We therefore recommend using the intranasal inoculation for future vaccine and challenge studies.

9.
Sci Rep ; 9(1): 14204, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578370

ABSTRACT

Canadian Indigenous peoples (First Nations and Inuit) exhibit a high burden of infectious diseases including tuberculosis influenced by societal factors, and biological determinants. Toll-like receptor (TLR)-mediated innate immune responses are the first line of defence against infections. We examined the production of a panel of 30 cytokines in peripheral blood-derived mononuclear cells (PBMC) isolated from Indigenous and non-Indigenous participants, following stimulation with five different TLR ligands. The levels of TLR-induced pro-inflammatory cytokines such as IL-12/23p40, IL-16, and IFN-γ, and chemokines (MCP-4, MDC and eotaxin) were different between Indigenous compared to non-Indigenous participants. Antimicrobial cationic host defence peptides (CHDP) induced by TLR activation are critical for resolution of infections and modulate the TLR-to-NFκB pathway to alter downstream cytokine responses. Therefore, we examined the expression of human CHDP defensins and cathelicidin in PBMC. mRNA expression of genes encoding for def-A1 and def-B1 were significantly higher following stimulation with TLR ligands in Indigenous compared to non-Indigenous participants. The purinergic receptor P2X7 known to be activated by ATP released following TLR stimulation, is a receptor for CHDP. Therefore, we further examined single nucleotide polymorphisms (SNP) in P2X7. Indigenous participants had a significantly higher percentage of a P2X7 SNP which is associated with reduced function and lower ability to clear infections. These results suggest that a higher frequency of non-functional P2X7 receptors may influence the activity of downstream immune mediators required for resolution of infections such as pro-inflammatory cytokines and CHDP defensins, thus contributing to higher burden of infections in Indigenous population.


Subject(s)
Indigenous Peoples/genetics , Polymorphism, Genetic/genetics , Receptors, Purinergic P2X7/genetics , Toll-Like Receptors/genetics , Canada/epidemiology , Cytokines/genetics , Defensins/genetics , Humans , Immunity, Innate/genetics , Interleukin-12/genetics , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Polymorphism, Single Nucleotide/genetics , Risk Factors , Signal Transduction/genetics
10.
Viruses ; 10(12)2018 12 12.
Article in English | MEDLINE | ID: mdl-30545088

ABSTRACT

Rift Valley fever virus (RVFV) is a zoonotic arbovirus of the Phenuiviridae family. Infection causes abortions in pregnant animals, high mortality in neonate animals, and mild to severe symptoms in both people and animals. There is currently an ongoing effort to produce safe and efficacious veterinary vaccines against RVFV in livestock to protect against both primary infection in animals and zoonotic infections in people. To test the efficacy of these vaccines, it is essential to have a reliable challenge model in relevant target species, including ruminants. We evaluated two goat breeds (Nubian and LaMancha), three routes of inoculation (intranasal, mosquito-primed subcutaneous, and subcutaneous) using an infectious dose of 107 pfu/mL, a virus strain from the 2006⁻2007 Kenyan/Sudan outbreak and compared the effect of using virus stocks produced in either mammalian or mosquito cells. Our results demonstrated that the highest and longest viremia titers were achieved in Nubian goats. The Nubian breed was also efficient at producing clinical signs, consistent viremia (peak viremia: 1.2 × 10³â»1.0 × 105 pfu/mL serum), nasal and oral shedding of viral RNA (1.5 × 10¹â»8 × 106 genome copies/swab), a systemic infection of tissues, and robust antibody responses regardless of the inoculation route. The Nubian goat breed and a needle-free intranasal inoculation technique could both be utilized in future vaccine and challenge studies. These studies are important for preventing the spread and outbreak of zoonotic viruses like RVFV and are supported by the Canadian-led BSL4ZNet network.


Subject(s)
Goats/virology , Injections, Subcutaneous/veterinary , Rift Valley Fever/blood , Zoonoses/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Breeding , Disease Models, Animal , Nasal Absorption , Phylogeny , RNA, Viral , Rift Valley Fever/pathology , Rift Valley fever virus , Viremia , Virus Shedding
11.
Synth Syst Biotechnol ; 3(1): 34-43, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29911197

ABSTRACT

All cellular functions, ranging from regular cell maintenance and homeostasis, specialized functions specific to cellular types, or generating responses due to external stimulus, are mediated by proteins within the cell. Regulation of these proteins allows the cell to alter its behavior under different circumstances. A major mechanism of protein regulation is utilizing protein kinases and phosphatases; enzymes that catalyze the transfer of phosphates between substrates [1]. Proteins involved in phosphate signaling are well studied and include kinases and phosphatases that catalyze opposing reactions regulating both structure and function of the cell. Kinomics is the study of kinases, phosphatases and their targets, and has been used to study the functional changes in numerous diseases and infectious diseases with aims to delineate the cellular functions affected. Identifying the phosphate signaling pathways changed by certain diseases or infections can lead to novel therapeutic targets. However, a daunting 518 putative protein kinase genes have been identified [2], indicating that this protein family is very large and complex. Identifying which enzymes are specific to a particular disease can be a laborious task. In this review, we will provide information on large-scale systems biology methodologies that allow global screening of the kinome to more efficiently identify which kinase pathways are pertinent for further study.

12.
Methods Mol Biol ; 1628: 273-282, 2017.
Article in English | MEDLINE | ID: mdl-28573628

ABSTRACT

Mouse models of Ebola virus (EBOV) have demonstrated their utility as important tools for screening the efficacy of candidate therapeutics and vaccines. In this chapter we explain the various mouse models that utilize either wild-type or mouse-adapted EBOV variants.


Subject(s)
Ebola Vaccines/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Animals , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Antiviral Agents/pharmacology , Disease Models, Animal , Ebolavirus/immunology , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Humans , Mice
13.
Can Fam Physician ; 63(5): 382-389, 2017 May.
Article in English | MEDLINE | ID: mdl-28500199

ABSTRACT

OBJECTIVE: To determine if the problem list (health conditions) in primary care electronic medical records (EMRs) accurately reflects the conditions for which chronic medications are prescribed in the EMR. DESIGN: A retrospective analysis of EMR data. SETTING: Eighteen primary care clinics across rural and urban Manitoba using the Accuro EMR. PARTICIPANTS: Data from the EMRs of active patients seen in an 18-month period (December 18, 2011, to June 18, 2013, or December 3, 2012, to June 3, 2014) were used. MAIN OUTCOME MEASURES: The likelihood of documentation in the EMR problem list of those specific chronic diseases for which drug prescriptions were documented in the EMR. Regression modeling was performed to determine the effect of clinic patient load and remuneration type on the completeness of EMR problem lists. RESULTS: Overall problem-list completeness was low but was highest for diabetes and lowest for insomnia. Fee-for-service clinics generally had lower problem-list completeness than salaried clinics did for all prescription medications examined. Panel size did not affect problem-list completeness rates. CONCLUSION: The low EMR problem-list completeness suggests that this field is not reliable for use in quality improvement initiatives or research until higher reliability has been demonstrated. Further research is recommended to explore the reasons for the poor quality and to support improvement efforts.


Subject(s)
Chronic Disease/drug therapy , Data Accuracy , Electronic Health Records/standards , Forms and Records Control/standards , Primary Health Care/standards , Humans , Manitoba , Odds Ratio , Primary Health Care/statistics & numerical data , Quality Improvement , Regression Analysis , Retrospective Studies
14.
Oncotarget ; 8(28): 46262-46272, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28545034

ABSTRACT

Sudan virus (SUDV) outbreaks in Africa are highly lethal; however, the development and testing of novel antivirals and vaccines for this virus has been limited by a lack of suitable animal models. Non-human primates (NHP) remain the gold standard for modeling filovirus disease, but they are not conducive to screening large numbers of experimental compounds and should only be used to test the most promising candidates. Therefore, other smaller animal models are a valuable asset. We have recently developed a guinea-pig adapted SUDV virus that is lethal in guinea pigs. In our current study, we show that ferrets are susceptible to wild-type SUDV, providing a small animal model to directly study clinical isolates, screen experimental anti-SUDV compounds and potentially study viral transmission.


Subject(s)
Disease Models, Animal , Ebola Vaccines/immunology , Ebolavirus/immunology , Ferrets , Hemorrhagic Fever, Ebola/virology , Animals , Antibodies, Viral/metabolism , Female , Guinea Pigs , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/transmission , Humans , Primates , Vaccination
15.
J Infect Dis ; 214(suppl 3): S281-S289, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27651412

ABSTRACT

Enhanced virulence and/or transmission of West African Ebola virus (EBOV) variants, which are divergent from their Central African counterparts, are suspected to have contributed to the sizable toll of the recent Ebola virus disease (EVD) outbreak. This study evaluated the pathogenicity and shedding in rhesus macaques infected with 1 of 2 West African isolates (EBOV-C05 or EBOV-C07) or a Central African isolate (EBOV-K). All animals infected with EBOV-C05 or EBOV-C07 died of EVD, whereas 2 of 3 EBOV-K-infected animals died. The viremia level was elevated 10-fold in EBOV-C05-infected animals, compared with EBOV-C07- or EBOV-K-infected animals. More-severe lung pathology was observed in 2 of 6 EBOV-C05/C07-infected macaques. This is the first detailed analysis of the recently circulating EBOV-C05/C07 in direct comparison to EBOV-K with 6 animals per group, and it showed that EBOV-C05 but not EBOV-C07 can replicate at higher levels and cause more tissue damage in some animals. Increased virus shedding from individuals who are especially susceptible to EBOV replication is possibly one of the many challenges facing the community of healthcare and policy-making responders since the beginning of the outbreak.


Subject(s)
Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Animals , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/pathology , Humans , Macaca mulatta , Species Specificity , Viremia , Virulence , Virus Shedding
16.
J Infect Dis ; 214(suppl 3): S326-S332, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27493239

ABSTRACT

A licensed vaccine against Ebola virus (EBOV) remains unavailable, despite >11 000 deaths from the 2014-2016 outbreak of EBOV disease in West Africa. Past studies have shown that recombinant vaccine viruses expressing EBOV glycoprotein (GP) are able to protect nonhuman primates (NHPs) from a lethal EBOV challenge. However, these vaccines express the viral GP-based EBOV variants found in Central Africa, which has 97.3% amino acid homology to the Makona variant found in West Africa. Our previous study showed that a recombinant adenovirus serotype 5 (Ad5)-vectored vaccine expressing the Makona EBOV GP (MakGP) was safe and immunogenic during clinical trials in China, but it is unknown whether the vaccine protects against EBOV infection. Here, we demonstrate that guinea pigs immunized with Ad5-MakGP developed robust humoral responses and were protected against exposure to guinea pig-adapted EBOV. Ad5-MakGP also elicited specific B- and T-cell immunity in NHPs and conferred 100% protection when animals were challenged 4 weeks after immunization. These results support further clinical development of this candidate and highlight the utility of Ad5-MakGP as a prophylactic measure in future outbreaks of EBOV disease.


Subject(s)
Adenovirus Vaccines/immunology , Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Immunization , Viral Proteins/immunology , Africa, Central , Africa, Western , Animals , China , Genetic Vectors , Glycoproteins/immunology , Guinea Pigs , Hemorrhagic Fever, Ebola/virology , Humans , Primates
17.
J Virol ; 90(20): 9209-23, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27489269

ABSTRACT

UNLABELLED: Bundibugyo virus (BDBV) is the etiological agent of a severe hemorrhagic fever in humans with a case-fatality rate ranging from 25 to 36%. Despite having been known to the scientific and medical communities for almost 1 decade, there is a dearth of studies on this pathogen due to the lack of a small animal model. Domestic ferrets are commonly used to study other RNA viruses, including members of the order Mononegavirales To investigate whether ferrets were susceptible to filovirus infections, ferrets were challenged with a clinical isolate of BDBV. Animals became viremic within 4 days and succumbed to infection between 8 and 9 days, and a petechial rash was observed with moribund ferrets. Furthermore, several hallmarks of human filoviral disease were recapitulated in the ferret model, including substantial decreases in lymphocyte and platelet counts and dysregulation of key biochemical markers related to hepatic/renal function, as well as coagulation abnormalities. Virological, histopathological, and immunohistochemical analyses confirmed uncontrolled BDBV replication in the major organs. Ferrets were also infected with Ebola virus (EBOV) to confirm their susceptibility to another filovirus species and to potentially establish a virus transmission model. Similar to what was seen with BDBV, important hallmarks of human filoviral disease were observed in EBOV-infected ferrets. This study demonstrates the potential of this small animal model for studying BDBV and EBOV using wild-type isolates and will accelerate efforts to understand filovirus pathogenesis and transmission as well as the development of specific vaccines and antivirals. IMPORTANCE: The 2013-2016 outbreak of Ebola virus in West Africa has highlighted the threat posed by filoviruses to global public health. Bundibugyo virus (BDBV) is a member of the genus Ebolavirus and has caused outbreaks in the past but is relatively understudied, likely due to the lack of a suitable small animal model. Such a model for BDBV is crucial to evaluating vaccines and therapies and potentially understanding transmission. To address this, we demonstrated that ferrets are susceptible models to BDBV infection as well as to Ebola virus infection and that no virus adaptation is required. Moreover, these animals develop a disease that is similar to that seen in humans and nonhuman primates. We believe that this will improve the ability to study BDBV and provide a platform to test vaccines and therapeutics.


Subject(s)
Ebolavirus/immunology , Ferrets/virology , Filoviridae Infections/microbiology , Filoviridae/immunology , Africa, Western , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Female , Filoviridae Infections/virology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Humans , Viral Vaccines/immunology
18.
Antimicrob Agents Chemother ; 60(9): 5182-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27297486

ABSTRACT

Ebola outbreaks occur on a frequent basis, with the 2014-2015 outbreak in West Africa being the largest one ever recorded. This outbreak has resulted in over 11,000 deaths in four African countries and has received international attention and intervention. Although there are currently no approved therapies or vaccines, many promising candidates are undergoing clinical trials, and several have had success in promoting recovery from Ebola. However, these prophylactics and therapeutics have been designed and tested only against the same species of Ebola virus as the one causing the current outbreak. Future outbreaks involving other species would require reformulation and possibly redevelopment. Therefore, a broad-spectrum alternative is highly desirable. We have found that a flavonoid derivative called quercetin 3-ß-O-d-glucoside (Q3G) has the ability to protect mice from Ebola even when given as little as 30 min prior to infection. Furthermore, we have demonstrated that this compound targets the early steps of viral entry. Most promisingly, antiviral activity against two distinct species of Ebola virus was seen. This study serves as a proof of principle that Q3G has potential as a prophylactic against Ebola virus infection.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/prevention & control , Quercetin/analogs & derivatives , Reassortant Viruses/drug effects , Virus Replication/drug effects , Animals , Chlorocebus aethiops , Dose-Response Relationship, Drug , Ebolavirus/growth & development , Female , Hemorrhagic Fever, Ebola/mortality , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Quercetin/pharmacology , Reassortant Viruses/growth & development , Survival Analysis , Toxicity Tests, Chronic , Treatment Outcome , Vero Cells
19.
Cell Rep ; 15(7): 1514-1526, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27160900

ABSTRACT

Previous efforts to identify cross-neutralizing antibodies to the receptor-binding site (RBS) of ebolavirus glycoproteins have been unsuccessful, largely because the RBS is occluded on the viral surface. We report a monoclonal antibody (FVM04) that targets a uniquely exposed epitope within the RBS; cross-neutralizes Ebola (EBOV), Sudan (SUDV), and, to a lesser extent, Bundibugyo viruses; and shows protection against EBOV and SUDV in mice and guinea pigs. The antibody cocktail ZMapp™ is remarkably effective against EBOV (Zaire) but does not cross-neutralize other ebolaviruses. By replacing one of the ZMapp™ components with FVM04, we retained the anti-EBOV efficacy while extending the breadth of protection to SUDV, thereby generating a cross-protective antibody cocktail. In addition, we report several mutations at the base of the ebolavirus glycoprotein that enhance the binding of FVM04 and other cross-reactive antibodies. These findings have important implications for pan-ebolavirus vaccine development and defining broadly protective antibody cocktails.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Ebolavirus/physiology , Epitopes/immunology , Glycoproteins/metabolism , Hemorrhagic Fever, Ebola/immunology , Receptors, Virus/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/ultrastructure , Antibodies, Neutralizing , Antibodies, Viral/chemistry , Binding Sites , Disease Models, Animal , Female , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/ultrastructure , Guinea Pigs , HEK293 Cells , Humans , Kinetics , Mice, Inbred BALB C , Models, Molecular , Mutation/genetics , Negative Staining , Neutralization Tests , Treatment Outcome
20.
Sci Rep ; 6: 25856, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27181584

ABSTRACT

Ebola virus infections cause a deadly hemorrhagic disease for which no vaccines or therapeutics has received regulatory approval. Here we show isolation of three (Q206, Q314 and Q411) neutralizing monoclonal antibodies (mAbs) against the surface glycoprotein (GP) of Ebola virus identified in West Africa in 2014 through sequential immunization of Chinese rhesus macaques and antigen-specific single B cell sorting. These mAbs demonstrated potent neutralizing activities against both pseudo and live Ebola virus independent of complement. Biochemical, single particle EM, and mutagenesis analysis suggested Q206 and Q411 recognized novel epitopes in the head while Q314 targeted the glycan cap in the GP1 subunit. Q206 and Q411 appeared to influence GP binding to its receptor NPC1. Treatment with these mAbs provided partial but significant protection against disease in a mouse model of Ebola virus infection. These novel mAbs could serve as promising candidates for prophylactic and therapeutic interventions against Ebola virus infection.


Subject(s)
Antibodies, Neutralizing/isolation & purification , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Macaca mulatta/immunology , Membrane Glycoproteins/immunology , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/administration & dosage , Antibodies, Viral/isolation & purification , Antibodies, Viral/pharmacology , B-Lymphocytes/immunology , Disease Models, Animal , Ebolavirus/drug effects , Epitopes/immunology , Immunization, Passive , Macaca mulatta/virology , Membrane Glycoproteins/chemistry , Mice , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...