Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
J Virol ; 97(10): e0059023, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37750724

ABSTRACT

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


Subject(s)
Disease Outbreaks , Ebolavirus , Genetic Variation , Hemorrhagic Fever, Ebola , Humans , Disease Outbreaks/statistics & numerical data , Ebolavirus/chemistry , Ebolavirus/classification , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/transmission , Hemorrhagic Fever, Ebola/virology , Uganda/epidemiology , Contact Tracing
2.
PLoS One ; 17(2): e0263732, 2022.
Article in English | MEDLINE | ID: mdl-35143574

ABSTRACT

Filoviruses are emerging pathogens that cause acute fever with high fatality rate and present a global public health threat. During the 2013-2016 Ebola virus outbreak, genome sequencing allowed the study of virus evolution, mutations affecting pathogenicity and infectivity, and tracing the viral spread. In 2018, early sequence identification of the Ebolavirus as EBOV in the Democratic Republic of the Congo supported the use of an Ebola virus vaccine. However, field-deployable sequencing methods are needed to enable a rapid public health response. Resequencing microarrays (RMA) are a targeted method to obtain genomic sequence on clinical specimens rapidly, and sensitively, overcoming the need for extensive bioinformatic analysis. This study presents the design and initial evaluation of an ebolavirus resequencing microarray (Ebolavirus-RMA) system for sequencing the major genomic regions of four Ebolaviruses that cause disease in humans. The design of the Ebolavirus-RMA system is described and evaluated by sequencing repository samples of three Ebolaviruses and two EBOV variants. The ability of the system to identify genetic drift in a replicating virus was achieved by sequencing the ebolavirus glycoprotein gene in a recombinant virus cultured under pressure from a neutralizing antibody. Comparison of the Ebolavirus-RMA results to the Genbank database sequence file with the accession number given for the source RNA and Ebolavirus-RMA results compared to Next Generation Sequence results of the same RNA samples showed up to 99% agreement.


Subject(s)
Antibodies, Neutralizing/pharmacology , Ebolavirus/classification , Glycoproteins/genetics , Sequence Analysis, RNA/methods , Ebolavirus/drug effects , Ebolavirus/genetics , Genetic Drift , Genome, Viral , High-Throughput Nucleotide Sequencing , Oligonucleotide Array Sequence Analysis , Phylogeny , Viral Proteins/genetics
3.
Nature ; 597(7877): 539-543, 2021 09.
Article in English | MEDLINE | ID: mdl-34526718

ABSTRACT

Seven years after the declaration of the first epidemic of Ebola virus disease in Guinea, the country faced a new outbreak-between 14 February and 19 June 2021-near the epicentre of the previous epidemic1,2. Here we use next-generation sequencing to generate complete or near-complete genomes of Zaire ebolavirus from samples obtained from 12 different patients. These genomes form a well-supported phylogenetic cluster with genomes from the previous outbreak, which indicates that the new outbreak was not the result of a new spillover event from an animal reservoir. The 2021 lineage shows considerably lower divergence than would be expected during sustained human-to-human transmission, which suggests a persistent infection with reduced replication or a period of latency. The resurgence of Zaire ebolavirus from humans five years after the end of the previous outbreak of Ebola virus disease reinforces the need for long-term medical and social care for patients who survive the disease, to reduce the risk of re-emergence and to prevent further stigmatization.


Subject(s)
Disease Outbreaks , Ebolavirus/genetics , Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Models, Biological , Animals , Democratic Republic of the Congo/epidemiology , Disease Outbreaks/statistics & numerical data , Ebolavirus/classification , Female , Guinea/epidemiology , Hemorrhagic Fever, Ebola/transmission , High-Throughput Nucleotide Sequencing , Humans , Male , Persistent Infection/virology , Phylogeny , Survivors , Time Factors , Viral Zoonoses/transmission , Viral Zoonoses/virology
4.
mBio ; 12(4): e0151721, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34372693

ABSTRACT

Ebolaviruses Bundibugyo virus (BDBV) and Ebola virus (EBOV) cause fatal hemorrhagic disease in humans and nonhuman primates. While the host response to EBOV is well characterized, less is known about BDBV infection. Moreover, immune signatures that mediate natural protection against all ebolaviruses remain poorly defined. To explore these knowledge gaps, we transcriptionally profiled BDBV-infected rhesus macaques, a disease model that results in incomplete lethality. This approach enabled us to identify prognostic indicators. As expected, survival (∼60%) correlated with reduced clinical pathology and circulating infectious virus, although peak viral RNA loads were not significantly different between surviving and nonsurviving macaques. Survivors had higher anti-BDBV antibody titers and transcriptionally derived cytotoxic T cell-, memory B cell-, and plasma cell-type quantities, demonstrating activation of adaptive immunity. Conversely, a poor prognosis was associated with lack of an appropriate adaptive response, sustained innate immune signaling, and higher expression of myeloid-derived suppressor cell (MDSC)-related transcripts (S100A8, S100A9, CEBPB, PTGS2, CXCR1, and LILRA3). MDSCs are potent immunosuppressors of cellular and humoral immunity, and therefore, they represent a potential therapeutic target. Circulating plasminogen activator inhibitor 1 (PAI-1) and tissue plasminogen activator (tPA) levels were also elevated in nonsurvivors and in survivors exhibiting severe illness, emphasizing the importance of maintaining coagulation homeostasis to control disease progression. IMPORTANCE Bundibugyo virus (BDBV) and Ebola virus (EBOV) are ebolaviruses endemic to Africa that cause severe, often fatal hemorrhagic disease. BDBV is considered a less pathogenic ebolavirus due to its reduced lethality during human outbreaks, as well as in experimentally infected nonhuman primates. The reduced mortality of BDBV in NHP models, resulting in a pool of survivors, afforded us the unique opportunity of identifying immune correlates that confer protection against ebolaviruses. In this study, we discovered that the survival of BDBV-infected nonhuman primates (NHPs) was dependent on early development of adaptive (memory) immune responses and reduced myeloid-derived suppressor cell (MDSC)-related signaling. MDSCs are a heterogenous group of immune cells implicated in a number of diseases that are powerful immunosuppressors of cellular and humoral immunity. Thus, MDSCs represent a novel therapeutic target to prevent ebolavirus disease. To our knowledge, this is the first study to link increased morbidity with recruitment of these potent immunosuppressive cells.


Subject(s)
Adaptive Immunity/genetics , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Signal Transduction/immunology , Adaptive Immunity/immunology , Africa , Animals , Antibodies, Viral/blood , Disease Progression , Ebolavirus/classification , Ebolavirus/pathogenicity , Female , Hemorrhagic Fever, Ebola/mortality , Humans , Macaca mulatta , Male , Memory B Cells/immunology , Plasminogen Activator Inhibitor 1/blood , Signal Transduction/genetics , Tissue Plasminogen Activator/blood
5.
Viruses ; 13(7)2021 07 17.
Article in English | MEDLINE | ID: mdl-34372594

ABSTRACT

Currently there is no FDA-licensed vaccine or therapeutic against Sudan ebolavirus (SUDV) infections. The largest ever reported 2014-2016 West Africa outbreak, as well as the 2021 outbreak in the Democratic Republic of Congo, highlight the critical need for countermeasures against filovirus infections. A well-characterized small animal model that is susceptible to wild-type filoviruses would greatly add to the screening of antivirals and vaccines. Here, we infected signal transducer and activator of transcription-1 knock out (STAT-1 KO) mice with five different wildtype filoviruses to determine susceptibility. SUDV and Marburg virus (MARV) were the most virulent, and caused 100% or 80% lethality, respectively. Zaire ebolavirus (EBOV), Bundibugyo ebolavirus (BDBV), and Taï Forest ebolavirus (TAFV) caused 40%, 20%, and no mortality, respectively. Further characterization of SUDV in STAT-1 KO mice demonstrated lethality down to 3.1 × 101 pfu. Viral genomic material was detectable in serum as early as 1 to 2 days post-challenge. The onset of viremia was closely followed by significant changes in total white blood cells and proportion of neutrophils and lymphocytes, as well as by an influx of neutrophils in the liver and spleen. Concomitant significant fluctuations in blood glucose, albumin, globulin, and alanine aminotransferase were also noted, altogether consistent with other models of filovirus infection. Finally, favipiravir treatment fully protected STAT-1 KO mice from lethal SUDV challenge, suggesting that this may be an appropriate small animal model to screen anti-SUDV countermeasures.


Subject(s)
Disease Models, Animal , Ebolavirus/genetics , Mice, Knockout , STAT1 Transcription Factor/genetics , Amides/therapeutic use , Animals , Antibodies, Viral/blood , Antiviral Agents/therapeutic use , Ebolavirus/classification , Ebolavirus/drug effects , Ebolavirus/pathogenicity , Female , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Male , Mice , Pyrazines/therapeutic use , Viral Proteins/genetics
7.
Virulence ; 12(1): 885-901, 2021 12.
Article in English | MEDLINE | ID: mdl-33734027

ABSTRACT

Ebola virus (EBOV), belonging to the species Zaire ebolavirus in the genus Ebolavirus, causes a severe febrile illness in humans with case fatality rates (CFRs) up to 90%. While there have been six virus species classified, which each have a single type virus in the genus Ebolavirus, CFRs of ebolavirus infections vary among viruses belonging to each distinct species. In this review, we aim to define the ebolavirus species-specific virulence on the basis of currently available laboratory and experimental findings. In addition, this review will also cover the variant-specific virulence of EBOV by referring to the unique biological and pathogenic characteristics of EBOV variant Makona, a new EBOV variant isolated from the 2013-2016 EBOV disease outbreak in West Africa. A better definition of species-specific and variant-specific virulence of ebolaviruses will facilitate our comprehensive knowledge on genus Ebolavirus biology, leading to the development of therapeutics against well-focused pathogenic mechanisms of each Ebola disease.


Subject(s)
Ebolavirus/genetics , Ebolavirus/pathogenicity , Genetic Variation , Hemorrhagic Fever, Ebola/virology , Animals , Antibodies, Viral , Disease Outbreaks , Ebolavirus/classification , Ebolavirus/immunology , Genome, Viral , Hemorrhagic Fever, Ebola/mortality , Humans , Mice , Virulence
8.
Genomics ; 113(1 Pt 2): 778-784, 2021 01.
Article in English | MEDLINE | ID: mdl-33069829

ABSTRACT

The coronavirus pandemic became a major risk in global public health. The outbreak is caused by SARS-CoV-2, a member of the coronavirus family. Though the images of the virus are familiar to us, in the present study, an attempt is made to hear the coronavirus by translating its protein spike into audio sequences. The musical features such as pitch, timbre, volume and duration are mapped based on the coronavirus protein sequence. Three different viruses Influenza, Ebola and Coronavirus were studied and compared through their auditory virus sequences by implementing Haar wavelet transform. The sonification of the coronavirus benefits in understanding the protein structures by enhancing the hidden features. Further, it makes a clear difference in the representation of coronavirus compared with other viruses, which will help in various research works related to virus sequence. This evolves as a simplified and novel way of representing the conventional computational methods.


Subject(s)
Algorithms , COVID-19/virology , Genome, Viral , Music , SARS-CoV-2/classification , SARS-CoV-2/genetics , Wavelet Analysis , Amino Acid Sequence , Cluster Analysis , Coronavirus/classification , Coronavirus/genetics , Ebolavirus/classification , Ebolavirus/genetics , Humans , Middle East Respiratory Syndrome Coronavirus/classification , Middle East Respiratory Syndrome Coronavirus/genetics , Orthomyxoviridae/classification , Orthomyxoviridae/genetics , Pandemics , RNA, Viral/genetics , Severe acute respiratory syndrome-related coronavirus/classification , Severe acute respiratory syndrome-related coronavirus/genetics , Viral Proteins/genetics
9.
Viruses ; 12(7)2020 07 14.
Article in English | MEDLINE | ID: mdl-32674252

ABSTRACT

Ongoing Ebola virus disease outbreaks in the Democratic Republic of the Congo follow the largest recorded outbreak in Western Africa (2013-2016). To combat outbreaks, testing of medical countermeasures (therapeutics or vaccines) requires a well-defined, reproducible, animal model. Here we present Ebola virus disease kinetics in 24 Chinese-origin rhesus monkeys exposed intramuscularly to a highly characterized, commercially available Kikwit Ebola virus Filovirus Animal Non-Clinical Group (FANG) stock. Until reaching predetermined clinical disease endpoint criteria, six animals underwent anesthesia for repeated clinical sampling and were compared to six that did not. Groups of three animals were euthanized and necropsied on days 3, 4, 5, and 6 post-exposure, respectively. In addition, three uninfected animals served as controls. Here, we present detailed characterization of clinical and laboratory disease kinetics and complete blood counts, serum chemistries, Ebola virus titers, and disease kinetics for future medical countermeasure (MCM) study design and control data. We measured no statistical difference in hematology, chemistry values, or time to clinical endpoint in animals that were anesthetized for clinical sampling during the acute disease compared to those that were not.


Subject(s)
Disease Models, Animal , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/physiopathology , Macaca mulatta , Animals , Disease Progression , Ebolavirus/classification , Female , Male , Viral Load , Viremia
10.
Immunobiology ; 225(3): 151949, 2020 05.
Article in English | MEDLINE | ID: mdl-32444135

ABSTRACT

Ebola virus is a highly pathogenic RNA virus that causes the Ebola haemorrhagic fever in human. This virus is considered as one of the dangerous viruses in the world with very high mortality rate. To date, no epitope-based subunit vaccine has yet been discovered to fight against Ebola although the outbreaks of this deadly virus took many lives in the past. In this study, approaches of reverse vaccinology were utilized in combination with different tools of immunoinformatics to design subunit vaccines against Ebola virus strain Mayinga-76. Three potential antigenic proteins of this virus i.e., matrix protein VP40, envelope glycoprotein and nucleoprotein were selected to construct the subunit vaccine. The MHC class-I, MHC class-II and B-cell epitopes were determined initially and after some robust analysis i.e., antigenicity, allergenicity, toxicity, conservancy and molecular docking study, EV-1, EV-2 and EV-3 were constructed as three potential vaccine constructs. These vaccine constructs are also expected to be effective on few other strains of Ebola virus since the highly conserved epitopes were used for vaccine construction. Thereafter, molecular docking study was conducted on these vaccines and EV-1 emerged as the best vaccine construct. Afterward, molecular dynamics simulation study revealed the good performances and stability of the intended vaccine protein. Finally, codon adaptation and in silico cloning were carried out to design a possible plasmid (pET-19b plasmid vector was used) for large scale production of the EV-1 vaccine. However, further in vitro and in vivo studies might be required on the predicted vaccines for final validation.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Vaccines, Subunit/immunology , Vaccinology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Computational Biology/methods , Ebolavirus/classification , Epitopes/chemistry , Epitopes/immunology , Genetic Engineering , Humans , Models, Molecular , Reproducibility of Results , Structure-Activity Relationship , Vaccinology/methods
11.
Nat Microbiol ; 5(5): 668-674, 2020 05.
Article in English | MEDLINE | ID: mdl-32341570

ABSTRACT

Virus taxonomy emerged as a discipline in the middle of the twentieth century. Traditionally, classification by virus taxonomists has been focussed on the grouping of relatively closely related viruses. However, during the past few years, the International Committee on Taxonomy of Viruses (ICTV) has recognized that the taxonomy it develops can be usefully extended to include the basal evolutionary relationships among distantly related viruses. Consequently, the ICTV has changed its Code to allow a 15-rank classification hierarchy that closely aligns with the Linnaean taxonomic system and may accommodate the entire spectrum of genetic divergence in the virosphere. The current taxonomies of three human pathogens, Ebola virus, severe acute respiratory syndrome coronavirus and herpes simplex virus 1 are used to illustrate the impact of the expanded rank structure. This new rank hierarchy of virus taxonomy will stimulate further research on virus origins and evolution, and vice versa, and could promote crosstalk with the taxonomies of cellular organisms.


Subject(s)
Classification , Viruses/classification , Viruses/genetics , Communicable Diseases/virology , Ebolavirus/classification , Ecology , Evolution, Molecular , Genes, Viral , Herpesvirus 1, Human/classification , Humans , Severe acute respiratory syndrome-related coronavirus/classification
12.
PLoS Negl Trop Dis ; 14(3): e0008117, 2020 03.
Article in English | MEDLINE | ID: mdl-32130210

ABSTRACT

Ebolaviruses pose a substantial threat to wildlife populations and to public health in Africa. Evolutionary analyses of virus genome sequences can contribute significantly to elucidate the origin of new outbreaks, which can help guide surveillance efforts. The reconstructed between-outbreak evolutionary history of Zaire ebolavirus so far has been highly consistent. By removing the confounding impact of population growth bursts during local outbreaks on the free mixing assumption that underlies coalescent-based demographic reconstructions, we find-contrary to what previous results indicated-that the circulation dynamics of Ebola virus in its animal reservoir are highly uncertain. Our findings also accentuate the need for a more fine-grained picture of the Ebola virus diversity in its reservoir to reliably infer the reservoir origin of outbreak lineages. In addition, the recent appearance of slower-evolving variants is in line with latency as a survival mechanism and with bats as the natural reservoir host.


Subject(s)
Animal Diseases/epidemiology , Chiroptera/virology , Disease Reservoirs/virology , Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/veterinary , Africa , Animal Diseases/virology , Animals , Ebolavirus/classification , Ebolavirus/genetics , Genotype , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Humans , Phylogeny
13.
Euro Surveill ; 25(3)2020 Jan.
Article in English | MEDLINE | ID: mdl-31992392

ABSTRACT

Genomic surveillance during ebolavirus outbreaks to elucidate transmission chains and develop diagnostic tests is delayed by the laborious development of variant-specific laboratory assays. We developed a new protocol combining 31 parallel PCR assays with Illumina/MinION-based sequencing, allowing generic ebolavirus genomic surveillance, validated using cell culture-derived Ebola, Reston, Sudan and Taï Forest virus at concentrations compatible with patient viral loads. Our approach enables pre-emptive genomic surveillance of ongoing and future ebolavirus outbreaks irrespective of variant divergence.


Subject(s)
DNA, Viral/analysis , Ebolavirus/genetics , Ebolavirus/isolation & purification , Genome, Viral/genetics , Hemorrhagic Fever, Ebola/diagnosis , Polymerase Chain Reaction/methods , RNA, Viral/genetics , Base Sequence , Communicable Diseases, Emerging , Ebolavirus/classification , Humans , Sensitivity and Specificity , Sequence Analysis, DNA
15.
J Clin Microbiol ; 57(9)2019 09.
Article in English | MEDLINE | ID: mdl-31315955

ABSTRACT

We applied metagenomic next-generation sequencing (mNGS) to detect Zaire Ebola virus (EBOV) and other potential pathogens from whole-blood samples from 70 patients with suspected Ebola hemorrhagic fever during a 2014 outbreak in Boende, Democratic Republic of the Congo (DRC) and correlated these findings with clinical symptoms. Twenty of 31 patients (64.5%) tested in Kinshasa, DRC, were EBOV positive by quantitative reverse transcriptase PCR (qRT-PCR). Despite partial degradation of sample RNA during shipping and handling, mNGS followed by EBOV-specific capture probe enrichment in a U.S. genomics laboratory identified EBOV reads in 22 of 70 samples (31.4%) versus in 21 of 70 (30.0%) EBOV-positive samples by repeat qRT-PCR (overall concordance = 87.1%). Reads from Plasmodium falciparum (malaria) were detected in 21 patients, of which at least 9 (42.9%) were coinfected with EBOV. Other positive viral detections included hepatitis B virus (n = 2), human pegivirus 1 (n = 2), Epstein-Barr virus (n = 9), and Orungo virus (n = 1), a virus in the Reoviridae family. The patient with Orungo virus infection presented with an acute febrile illness and died rapidly from massive hemorrhage and dehydration. Although the patient's blood sample was negative by EBOV qRT-PCR testing, identification of viral reads by mNGS confirmed the presence of EBOV coinfection. In total, 9 new EBOV genomes (3 complete genomes, and an additional 6 ≥50% complete) were assembled. Relaxed molecular clock phylogenetic analysis demonstrated a molecular evolutionary rate for the Boende strain 4 to 10× slower than that of other Ebola lineages. These results demonstrate the utility of mNGS in broad-based pathogen detection and outbreak surveillance.


Subject(s)
Coinfection/epidemiology , Disease Outbreaks , Ebolavirus/classification , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , High-Throughput Nucleotide Sequencing/methods , Metagenomics/methods , Adult , Coinfection/parasitology , Coinfection/pathology , Coinfection/virology , Democratic Republic of the Congo/epidemiology , Ebolavirus/genetics , Ebolavirus/isolation & purification , Female , Hemorrhagic Fever, Ebola/parasitology , Hemorrhagic Fever, Ebola/pathology , Humans , Infant , Male , Middle Aged , Young Adult
16.
Biochem Biophys Res Commun ; 514(3): 726-733, 2019 06 30.
Article in English | MEDLINE | ID: mdl-31078274

ABSTRACT

Microsatellites (SSRs) are ubiquitous in coding and non-coding regions of the Ebolavirus genomes. We synthetically analyzed the microsatellites in whole-genome and terminal regions of 219 Ebolavirus genomes from five species. The Ebolavirus sequences were observed with small intraspecies variations and large interspecific variations, especially in the terminal non-coding regions. Only five conserved microsatellites were detected in the complete genomes, and four of them which well base-paired to help forming conserved stem-loop structures mainly appeared in the terminal non-coding regions. These results suggest that the conserved microsatellites may be evolutionary selected to form conserved secondary structures in 5', 3' terminals of Ebolavirus genomes. It may help to understand the biological significance of microsatellites in Ebolavirus and also other virus genomes.


Subject(s)
Conserved Sequence , Ebolavirus/genetics , Genome, Viral , Inverted Repeat Sequences , Microsatellite Repeats , RNA, Viral/genetics , Base Pairing , Databases, Genetic , Ebolavirus/classification , Evolution, Molecular , Nucleic Acid Conformation , RNA, Viral/chemistry , Selection, Genetic , Sequence Alignment , Sequence Homology, Nucleic Acid
17.
Emerg Infect Dis ; 25(5): 955-957, 2019 05.
Article in English | MEDLINE | ID: mdl-31002301

ABSTRACT

Bombali virus (genus Ebolavirus) was identified in organs and excreta of an Angolan free-tailed bat (Mops condylurus) in Kenya. Complete genome analysis revealed 98% nucleotide sequence similarity to the prototype virus from Sierra Leone. No Ebola virus-specific RNA or antibodies were detected from febrile humans in the area who reported contact with bats.


Subject(s)
Chiroptera/virology , Ebolavirus , Animals , Ebolavirus/classification , Ebolavirus/genetics , Genome, Viral , Geography , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/transmission , Hemorrhagic Fever, Ebola/virology , Kenya/epidemiology , Phylogeny , Public Health Surveillance
18.
Lancet ; 393(10174): 936-948, 2019 03 02.
Article in English | MEDLINE | ID: mdl-30777297

ABSTRACT

Ebolaviruses are pathogenic agents associated with a severe, potentially fatal, systemic disease in man and great apes. Four species of ebolaviruses have been identified in west or equatorial Africa. Once the more virulent forms enter the human population, transmission occurs primarily through contact with infected body fluids and can result in major epidemics in under-resourced settings. These viruses cause a disease characterised by systemic viral replication, immune suppression, abnormal inflammatory responses, major fluid and electrolyte losses, and high mortality. Despite recent progress on vaccines, and with no licensed prophylaxis or treatment available, case management is essentially supportive with management of severe multiple organ failure resulting from immune-mediated cell damage. The 2013-16 outbreak was classified by WHO as a Public Health Emergency of International Concern, which drew attention to the challenges of diseases caused by infections with ebolaviruses and questioned scientific, clinical, and societal preparation to handle future epidemics.


Subject(s)
Disease Outbreaks/prevention & control , Ebolavirus , Hemorrhagic Fever, Ebola , Africa, Western/epidemiology , Animals , Disease Outbreaks/statistics & numerical data , Disease Progression , Ebola Vaccines/immunology , Ebolavirus/classification , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/mortality , Hemorrhagic Fever, Ebola/therapy , Hemorrhagic Fever, Ebola/transmission , Humans , International Cooperation
19.
Nat Commun ; 10(1): 105, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30631063

ABSTRACT

The 2013-2016 Ebola virus (EBOV) disease epidemic demonstrated the grave consequences of filovirus epidemics in the absence of effective therapeutics. Besides EBOV, two additional ebolaviruses, Sudan (SUDV) and Bundibugyo (BDBV) viruses, as well as multiple variants of Marburg virus (MARV), have also caused high fatality epidemics. Current experimental EBOV monoclonal antibodies (mAbs) are ineffective against SUDV, BDBV, or MARV. Here, we report that a cocktail of two broadly neutralizing ebolavirus mAbs, FVM04 and CA45, protects nonhuman primates (NHPs) against EBOV and SUDV infection when delivered four days post infection. This cocktail when supplemented by the anti-MARV mAb MR191 exhibited 100% efficacy in MARV-infected NHPs. These findings provide a solid foundation for clinical development of broadly protective immunotherapeutics for use in future filovirus epidemics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Ebolavirus/immunology , Filoviridae Infections/immunology , Marburgvirus/immunology , Primate Diseases/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Ebolavirus/classification , Ebolavirus/drug effects , Ebolavirus/physiology , Filoviridae Infections/therapy , Filoviridae Infections/virology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Immunotherapy/methods , Marburgvirus/drug effects , Marburgvirus/physiology , Primate Diseases/therapy , Primate Diseases/virology , Primates , Treatment Outcome
20.
Viruses ; 11(1)2019 01 16.
Article in English | MEDLINE | ID: mdl-30654482

ABSTRACT

We generated genome sequences from 218 cases of Ebola virus disease (EVD) in Sierra Leone (SLE) during 2014⁻2015 to complement available datasets, particularly by including cases from a period of low sequence coverage during peak transmission of Ebola virus (EBOV) in the highly-affected Western Area division of SLE. The combined dataset was utilized to produce phylogenetic and phylodynamic inferences, to study sink⁻source dynamics and virus dispersal from highly-populated transmission hotspots. We identified four districts in SLE where EBOV was introduced and transmission occurred without onward exportation to other districts. We also identified six districts that substantially contributed to the dispersal of the virus and prolonged the EVD outbreak: five of these served as major hubs, with lots of movement in and out, and one acted primarily as a source, exporting the virus to other areas of the country. Positive correlations between case numbers, inter-district transition events, and district population sizes reaffirm that population size was a driver of EBOV transmission dynamics in SLE. The data presented here confirm the role of urban hubs in virus dispersal and of a delayed laboratory response in the expansion and perpetuation of the EVD outbreak in SLE.


Subject(s)
Ebolavirus/genetics , Hemorrhagic Fever, Ebola/transmission , Phylogeny , Disease Outbreaks , Ebolavirus/classification , Genome, Viral , Hemorrhagic Fever, Ebola/epidemiology , High-Throughput Nucleotide Sequencing , Humans , Sierra Leone/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...