Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
CMAJ ; 195(22): E800-E801, 2023 06 05.
Article in French | MEDLINE | ID: mdl-37277129
3.
CMAJ ; 195(8): E305, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36849175
4.
PLoS Pathog ; 17(10): e1009966, 2021 10.
Article in English | MEDLINE | ID: mdl-34634087

ABSTRACT

Nigeria continues to experience ever increasing annual outbreaks of Lassa fever (LF). The World Health Organization has recently declared Lassa virus (LASV) as a priority pathogen for accelerated research leading to a renewed international effort to develop relevant animal models of disease and effective countermeasures to reduce LF morbidity and mortality in endemic West African countries. A limiting factor in evaluating medical countermeasures against LF is a lack of well characterized animal models outside of those based on infection with LASV strain Josiah originating form Sierra Leone, circa 1976. Here we genetically characterize five recent LASV isolates collected from the 2018 outbreak in Nigeria. Three isolates were further evaluated in vivo and despite being closely related and from the same spatial / geographic region of Nigeria, only one of the three isolates proved lethal in strain 13 guinea pigs and non-human primates (NHP). Additionally, this isolate exhibited atypical pathogenesis characteristics in the NHP model, most notably respiratory failure, not commonly described in hemorrhagic cases of LF. These results suggest that there is considerable phenotypic heterogeneity in LASV infections in Nigeria, which leads to a multitude of pathogenesis characteristics that could account for differences between subclinical and lethal LF infections. Most importantly, the development of disease models using currently circulating LASV strains in West Africa are critical for the evaluation of potential vaccines and medical countermeasures.


Subject(s)
Disease Models, Animal , Lassa Fever/genetics , Lassa virus/genetics , Animals , Disease Outbreaks , Female , Guinea Pigs , Humans , Macaca fascicularis , Male , Nigeria , Phylogeny
5.
Microorganisms ; 9(3)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33652895

ABSTRACT

BACKGROUND: The 2014-2016 Ebola outbreak in West Africa recapitulated that nosocomial spread of Ebola virus could occur and that health care workers were at particular risk including notable cases in Europe and North America. These instances highlighted the need for centers to better prepare for potential Ebola virus cases; including understanding how the virus spreads and which interventions pose the greatest risk. METHODS: We created a fully equipped intensive care unit (ICU), within a Biosafety Level 4 (BSL4) laboratory, and infected multiple sedated non-human primates (NHPs) with Ebola virus. While providing bedside care, we sampled blood, urine, and gastric residuals; as well as buccal, ocular, nasal, rectal, and skin swabs, to assess the risks associated with routine care. We also assessed the physical environment at end-point. RESULTS: Although viral RNA was detectable in blood as early as three days post-infection, it was not detectable in the urine, gastric fluid, or swabs until late-stage disease. While droplet spread and fomite contamination were present on a few of the surfaces that were routinely touched while providing care in the ICU for the infected animal, these may have been abrogated through good routine hygiene practices. CONCLUSIONS: Overall this study has helped further our understanding of which procedures may pose the highest risk to healthcare providers and provides temporal evidence of this over the clinical course of disease.

6.
Emerg Infect Dis ; 26(12): 3020-3024, 2020 12.
Article in English | MEDLINE | ID: mdl-33219792

ABSTRACT

Hantavirus cardiopulmonary syndrome (HCPS) is a severe respiratory disease caused by Sin Nombre virus in North America (SNV). As of January 1, 2020, SNV has caused 143 laboratory-confirmed cases of HCPS in Canada. We review critical aspects of SNV virus epidemiology and the ecology, biology, and genetics of HCPS in Canada.


Subject(s)
Hantavirus Infections , Hantavirus Pulmonary Syndrome , Orthohantavirus , Sin Nombre virus , Canada/epidemiology , Orthohantavirus/genetics , Hantavirus Infections/epidemiology , Hantavirus Pulmonary Syndrome/diagnosis , Hantavirus Pulmonary Syndrome/epidemiology , Humans , North America
8.
mBio ; 10(5)2019 10 22.
Article in English | MEDLINE | ID: mdl-31641086

ABSTRACT

The 1918 influenza virus, subtype H1N1, was the causative agent of the most devastating pandemic in the history of infectious diseases. In vitro studies have confirmed that extreme virulence is an inherent property of this virus. Here, we utilized the macaque model for evaluating the efficacy of oseltamivir phosphate against the fully reconstructed 1918 influenza virus in a highly susceptible and relevant disease model. Our findings demonstrate that oseltamivir phosphate is effective in preventing severe disease in macaques but vulnerable to virus escape through emergence of resistant mutants, especially if given in a treatment regimen. Nevertheless, we conclude that oseltamivir would be highly beneficial to reduce the morbidity and mortality rates caused by a highly pathogenic influenza virus although it would be predicted that resistance would likely emerge with sustained use of the drug.IMPORTANCE Oseltamivir phosphate is used as a first line of defense in the event of an influenza pandemic prior to vaccine administration. Treatment failure through selection and replication of drug-resistant viruses is a known complication in the field and was also demonstrated in our study with spread of resistant 1918 influenza virus in multiple respiratory tissues. This emphasizes the importance of early treatment and the possibility that noncompliance may exacerbate treatment effectiveness. It also demonstrates the importance of implementing combination therapy and vaccination strategies as soon as possible in a pandemic situation.


Subject(s)
Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/drug therapy , Oseltamivir/therapeutic use , Animals , Macaca , Orthomyxoviridae Infections/virology
9.
Intensive Care Med Exp ; 7(1): 54, 2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31520194

ABSTRACT

BACKGROUND: There are currently limited data for the use of specific antiviral therapies for the treatment of Ebola virus disease (EVD). While there is anecdotal evidence that supportive care may be effective, there is a paucity of direct experimental data to demonstrate a role for supportive care in EVD. We studied the impact of ICU-level supportive care interventions including fluid resuscitation, vasoactive medications, blood transfusion, hydrocortisone, and ventilator support on the pathophysiology of EVD in rhesus macaques infected with a universally lethal dose of Ebola virus strain Makona C07. METHODS: Four NHPs were infected with a universally lethal dose Ebola virus strain Makona, in accordance with the gold standard lethal Ebola NHP challenge model. Following infection, the following therapeutic interventions were employed: continuous bedside supportive care, ventilator support, judicious fluid resuscitation, vasoactive medications, blood transfusion, and hydrocortisone as needed to treat cardiovascular compromise. A range of physiological parameters were continuously monitored to gage any response to the interventions. RESULTS: All four NHPs developed EVD and demonstrated a similar clinical course. All animals reached a terminal endpoint, which occurred at an average time of 166.5 ± 14.8 h post-infection. Fluid administration may have temporarily blunted a rise in lactate, but the effect was short lived. Vasoactive medications resulted in short-lived improvements in mean arterial pressure. Blood transfusion and hydrocortisone did not appear to have a significant positive impact on the course of the disease. CONCLUSIONS: The model employed for this study is reflective of an intramuscular infection in humans (e.g., needle stick) and is highly lethal to NHPs. Using this model, we found that the animals developed progressive severe organ dysfunction and profound shock preceding death. While the overall impact of supportive care on the observed pathophysiology was limited, we did observe some time-dependent positive responses. Since this model is highly lethal, it does not reflect the full spectrum of human EVD. Our findings support the need for continued development of animal models that replicate the spectrum of human disease as well as ongoing development of anti-Ebola therapies to complement supportive care.

10.
Sci Rep ; 8(1): 15994, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30375468

ABSTRACT

Continued outbreaks of Henipaviruses in South Asia and Australia cause severe and lethal disease in both humans and animals. Together, with evidence of human to human transmission for Nipah virus and the lack of preventative or therapeutic measures, its threat to cause a widespread outbreak and its potential for weaponization has increased. In this study we demonstrate how overexpression of the Nipah virus nucleocapsid protein regulates viral polymerase activity and viral RNA production. By overexpressing the Nipah virus nucleocapsid protein in trans viral transcription was inhibited; however, an increase in viral genome synthesis was observed. Together, the bias of polymerase activity towards genome production led to the severe inhibition of viral progeny. We identified two domains within the nucleocapsid protein, which were each independently capable of binding the viral phosphoprotein. Evident by our data, we propose that the nucleocapsid protein's ability to interact with the phosphoprotein of the polymerase complex causes a change in polymerase activity and subsequent deficiency in viral replication. This study not only provides insights into the dynamics of Henipavirus RNA synthesis and replication, but also provides insight into potential targets for antiviral drug development.


Subject(s)
Henipavirus Infections/genetics , Nipah Virus/genetics , Nucleocapsid Proteins/genetics , Virus Replication/genetics , Henipavirus Infections/transmission , Henipavirus Infections/virology , Humans , Nipah Virus/pathogenicity , Phosphoproteins/genetics , Protein Binding/genetics , RNA, Viral/genetics , Viral Proteins/genetics , Virion/genetics , Virion/pathogenicity
11.
Am J Trop Med Hyg ; 98(3): 872-874, 2018 03.
Article in English | MEDLINE | ID: mdl-29363462

ABSTRACT

Rift Valley fever virus (RVFV) outbreaks have considerable impact on human and animal health. Here, we are reporting a serosurvey of cattle from all regions of Mali. These demonstrated that few had been exposed to RVFV from 2005 to 2014. Recent outbreaks of RVF in Niger and a single human case in Mali provide justification for further entomological and ecological studies of this virus.


Subject(s)
Antibodies, Viral/blood , Cattle Diseases/epidemiology , Disease Outbreaks , Rift Valley Fever/epidemiology , Rift Valley fever virus/pathogenicity , Aedes/virology , Animals , Cattle , Cattle Diseases/transmission , Cattle Diseases/virology , Humans , Mali/epidemiology , Mosquito Vectors/virology , Neutralization Tests , Rift Valley Fever/transmission , Rift Valley Fever/virology , Rift Valley fever virus/immunology , Rift Valley fever virus/physiology , Seroepidemiologic Studies
12.
Methods Mol Biol ; 1628: 341-352, 2017.
Article in English | MEDLINE | ID: mdl-28573633

ABSTRACT

Reverse transcriptase polymerase chain reaction (RT-PCR)-based techniques allow for highly sensitive and specific detection of RNA viruses. Detection of the amplification products can be achieved using several methods. The following are descriptions of the detection of ebolavirus RNA using end-point RT-PCR (agarose gel visualization of amplification products) and quantitative RT-PCR (Q-RT-PCR), with fluorescent detection using an intercalating dye or detection with the use of 5' hydrolysis probe assays. All of these techniques can be used to accurately detect the presence of ebolavirus in samples.


Subject(s)
Ebolavirus/genetics , Hemorrhagic Fever, Ebola/diagnosis , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Ebolavirus/isolation & purification , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Humans , RNA, Viral/genetics , RNA, Viral/isolation & purification
13.
Sci Rep ; 7(1): 1204, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28446775

ABSTRACT

Critical care needs have been rising in recent decades as populations age and comorbidities increase. Sepsis-related admissions to critical care contribute up to 50% of volume and septic shock carries a 35-54% fatality rate. Improvements in sepsis-related care and mortality would have a significant impact of a resource-intensive area of health care delivery. Unfortunately, research has been hampered by the lack of an animal model that replicates the complex care provided to humans in an intensive care unit (ICU). We developed a protocol to provide full ICU type supportive care to Rhesus macaques. This included mechanical ventilation, continuous sedation, fluid and electrolyte management and vasopressor support in response to Ebolavirus-induced septic shock. The animals accurately recapitulated human responses to a full range of ICU interventions (e.g. fluid resuscitation). This model can overcome current animal model limitations by accurately emulating the complexity of ICU care and thereby provide a platform for testing new interventions in critical care and sepsis without placing patients at risk.


Subject(s)
Critical Care/methods , Critical Illness , Hemorrhagic Fever, Ebola/complications , Shock, Septic/therapy , Animals , Disease Models, Animal , Macaca mulatta
14.
J Infect Dis ; 214(suppl 3): S218-S221, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27571899

ABSTRACT

Personnel deployed to remote areas during infectious disease outbreaks have limited access to mechanical and chemical inactivation resources. The inactivation of infectious agents present in diagnostic samples is critical to ensure the safety of personnel and the containment of the disease. We evaluated the efficacy of thermal inactivation (exposure to 56°C for 1 hour) and chemical inactivation with 0.5% Tween-20 against a high titer of Ebola virus (species Zaire ebolavirus) variant Makona in spiked human serum samples. No surviving virus was revealed by a 50% tissue culture infective dose assay after the combined treatment under laboratory conditions. In-field use of this inactivation protocol during the 2013-2016 West Africa Ebola outbreaks demonstrated readily detectable levels of immunoglobulin G and/or immunoglobulin M in human plasma samples after treatment.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Disease Outbreaks , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/diagnosis , Virus Inactivation , Africa, Western/epidemiology , Animals , Chlorocebus aethiops , Democratic Republic of the Congo/epidemiology , Ebolavirus/isolation & purification , Enzyme-Linked Immunosorbent Assay , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Hot Temperature , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Vero Cells
15.
Clin Infect Dis ; 63(8): 1026-33, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27531847

ABSTRACT

BACKGROUND: The ongoing Ebola outbreak in West Africa has resulted in 28 646 suspected, probable, and confirmed Ebola virus infections. Nevertheless, malaria remains a large public health burden in the region affected by the outbreak. A joint Centers for Disease Control and Prevention/National Institutes of Health diagnostic laboratory was established in Monrovia, Liberia, in August 2014, to provide laboratory diagnostics for Ebola virus. METHODS: All blood samples from suspected Ebola virus-infected patients admitted to the Médecins Sans Frontières ELWA3 Ebola treatment unit in Monrovia were tested by quantitative real-time polymerase chain reaction for the presence of Ebola virus and Plasmodium species RNA. Clinical outcome in laboratory-confirmed Ebola virus-infected patients was analyzed as a function of age, sex, Ebola viremia, and Plasmodium species parasitemia. RESULTS: The case fatality rate of 1182 patients with laboratory-confirmed Ebola virus infections was 52%. The probability of surviving decreased with increasing age and decreased with increasing Ebola viral load. Ebola virus-infected patients were 20% more likely to survive when Plasmodium species parasitemia was detected, even after controlling for Ebola viral load and age; those with the highest levels of parasitemia had a survival rate of 83%. This effect was independent of treatment with antimalarials, as this was provided to all patients. Moreover, treatment with antimalarials did not affect survival in the Ebola virus mouse model. CONCLUSIONS: Plasmodium species parasitemia is associated with an increase in the probability of surviving Ebola virus infection. More research is needed to understand the molecular mechanism underlying this remarkable phenomenon and translate it into treatment options for Ebola virus infection.


Subject(s)
Coinfection , Ebolavirus , Hemorrhagic Fever, Ebola/complications , Hemorrhagic Fever, Ebola/mortality , Malaria/complications , Malaria/parasitology , Parasitemia , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Disease Models, Animal , Ebolavirus/genetics , Female , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/epidemiology , Humans , Infant , Infant, Newborn , Malaria/diagnosis , Malaria/epidemiology , Male , Mice , Middle Aged , Parasite Load , Plasmodium/genetics , Survival Rate , Viral Load , Young Adult
16.
J Infect Dis ; 214(suppl 3): S169-S176, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27333914

ABSTRACT

West Africa experienced the first epidemic of Ebola virus infection, with by far the greatest number of cases in Guinea, Sierra Leone, and Liberia. The unprecedented epidemic triggered an unparalleled response, including the deployment of multiple Ebola treatment units and mobile/field diagnostic laboratories. The National Institute of Allergy and Infectious Diseases and the Centers for Disease Control and Prevention deployed a joint laboratory to Monrovia, Liberia, in August 2014 to support the newly founded Ebola treatment unit at the Eternal Love Winning Africa (ELWA) campus. The laboratory operated initially out of a tent structure but quickly moved into a fixed-wall building owing to severe weather conditions, the need for increased security, and the high sample volume. Until May 2015, when the laboratory closed, the site handled close to 6000 clinical specimens for Ebola virus diagnosis and supported the medical staff in case patient management. Laboratory operation and safety, as well as Ebola virus diagnostic assays, are described and discussed; in addition, lessons learned for future deployments are reviewed.


Subject(s)
Clinical Laboratory Services/organization & administration , Ebolavirus/isolation & purification , Epidemics/prevention & control , Hemorrhagic Fever, Ebola/epidemiology , Africa, Western/epidemiology , Centers for Disease Control and Prevention, U.S. , Female , Guinea/epidemiology , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/transmission , Hemorrhagic Fever, Ebola/virology , Humans , International Cooperation , Liberia/epidemiology , Male , National Institute of Allergy and Infectious Diseases (U.S.) , Safety , Sierra Leone/epidemiology , United States
17.
Emerg Infect Dis ; 22(2): 323-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26814608

ABSTRACT

Malaria is a major public health concern in the countries affected by the Ebola virus disease epidemic in West Africa. We determined the feasibility of using molecular malaria diagnostics during an Ebola virus disease outbreak and report the incidence of Plasmodium spp. parasitemia in persons with suspected Ebola virus infection.


Subject(s)
Coinfection , Disease Outbreaks , Ebolavirus , Hemorrhagic Fever, Ebola/epidemiology , Malaria/diagnosis , Malaria/parasitology , Humans , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Parasite Load , Plasmodium falciparum/classification , Plasmodium falciparum/genetics , Prevalence
18.
Antiviral Res ; 126: 55-61, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26681387

ABSTRACT

The West Africa Ebola virus disease (EVD) outbreak has reached unprecedented magnitude and caused worldwide concerns for the spread of this deadly virus. Recent findings in nonhuman primates (NHPs) demonstrate that antibodies can be protective against EVD. However, the role of antibody response in vaccine-mediated protection is not fully understood. To address these questions quantitative serology assays are needed for measurement of the antibody response to key Ebola virus (EBOV) proteins. Serology enzyme-linked immunosorbent assays (ELISA's), using a reference detection antibody, were developed in order to standardize the quantitation of antibody levels in vaccinated NHPs or in humans exposed to EBOV or immunized with an EBOV vaccine. Critical reagents were generated to support the development of the serology ELISAs. Recombinant EBOV matrix protein (VP40) was expressed in Escherichia coli and purified. Two variants of the glycoprotein (GP), the ectodomain lacking the transmembrane domain (GPΔTM), and an engineered GP lacking the mucin-like domain (GPΔmuc) were expressed and purified from mammalian cell systems. Using these proteins, three ELISA methods were developed and optimized for reproducibility and robustness, including stability testing of critical reagents. The assay was used to determine the antibody response against VP40, GPΔTM, and GPΔmuc in a NHP vaccine study using EBOV virus-like particles (VLP) vaccine expressing GP, VP40 and the nucleoprotein. Additionally, these ELISAs were used to successfully detect antibody responses to VP40, GPΔTM and GPΔmuc in human sera from EBOV infected individuals.


Subject(s)
Antibodies, Viral/blood , Ebola Vaccines/immunology , Ebolavirus/immunology , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation , Disease Models, Animal , Ebola Vaccines/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Glycoproteins/blood , HEK293 Cells , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/virology , Humans , Macaca , Macaca fascicularis , Male , Protein Engineering , Reproducibility of Results , Viral Matrix Proteins/blood , Viral Matrix Proteins/genetics
20.
J Clin Invest ; 125(12): 4421-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26551677

ABSTRACT

BACKGROUND: Ebola virus (EBOV) causes periodic outbreaks of life-threatening EBOV disease in Africa. Historically, these outbreaks have been relatively small and geographically contained; however, the magnitude of the EBOV outbreak that began in 2014 in West Africa has been unprecedented. The aim of this study was to describe the viral kinetics of EBOV during this outbreak and identify factors that contribute to outbreak progression. METHODS: From July to December 2014, one laboratory in Sierra Leone processed over 2,700 patient samples for EBOV detection by quantitative PCR (qPCR). Viremia was measured following patient admission. Age, sex, and approximate time of symptom onset were also recorded for each patient. The data was analyzed using various mathematical models to find trends of potential interest. RESULTS: The analysis revealed a significant difference (P = 2.7 × 10(-77)) between the initial viremia of survivors (4.02 log10 genome equivalents [GEQ]/ml) and nonsurvivors (6.18 log10 GEQ/ml). At the population level, patient viral loads were higher on average in July than in November, even when accounting for outcome and time since onset of symptoms. This decrease in viral loads temporally correlated with an increase in circulating EBOV-specific IgG antibodies among individuals who were suspected of being infected but shown to be negative for the virus by PCR. CONCLUSIONS: Our results indicate that initial viremia is associated with outcome of the individual and outbreak duration; therefore, care must be taken in planning clinical trials and interventions. Additional research in virus adaptation and the impacts of host factors on EBOV transmission and pathogenesis is needed.


Subject(s)
Disease Outbreaks , Ebolavirus , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/mortality , Models, Biological , Viral Load , Antibodies, Viral/blood , Female , Humans , Immunoglobulin G/blood , Male , Sierra Leone
SELECTION OF CITATIONS
SEARCH DETAIL
...