Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Nat Commun ; 15(1): 3589, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678025

ABSTRACT

The black rat (Rattus rattus) is a globally invasive species that has been widely introduced across Africa. Within its invasive range in West Africa, R. rattus may compete with the native rodent Mastomys natalensis, the primary reservoir host of Lassa virus, a zoonotic pathogen that kills thousands annually. Here, we use rodent trapping data from Sierra Leone and Guinea to show that R. rattus presence reduces M. natalensis density within the human dwellings where Lassa virus exposure is most likely to occur. Further, we integrate infection data from M. natalensis to demonstrate that Lassa virus zoonotic spillover risk is lower at sites with R. rattus. While non-native species can have numerous negative effects on ecosystems, our results suggest that R. rattus invasion has the indirect benefit of decreasing zoonotic spillover of an endemic pathogen, with important implications for invasive species control across West Africa.


Subject(s)
Disease Reservoirs , Introduced Species , Lassa Fever , Lassa virus , Murinae , Zoonoses , Animals , Lassa virus/pathogenicity , Lassa virus/physiology , Lassa Fever/transmission , Lassa Fever/epidemiology , Lassa Fever/virology , Lassa Fever/veterinary , Disease Reservoirs/virology , Humans , Rats , Murinae/virology , Zoonoses/virology , Zoonoses/transmission , Zoonoses/epidemiology , Sierra Leone/epidemiology , Guinea/epidemiology , Ecosystem , Rodent Diseases/virology , Rodent Diseases/epidemiology , Rodent Diseases/transmission
2.
J Virol ; 96(16): e0075422, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35913216

ABSTRACT

Lassa virus (LASV) is a mammarenavirus that can cause lethal Lassa fever disease with no FDA-approved vaccine and limited treatment options. Fatal LASV infections are associated with innate immune suppression. We have previously shown that the small matrix Z protein of LASV, but not of a nonpathogenic arenavirus Pichinde virus (PICV), can inhibit the cellular RIG-I-like receptors (RLRs), but its biological significance has not been evaluated in an infectious virus due to the multiple essential functions of the Z protein required for the viral life cycle. In this study, we developed a stable HeLa cell line (HeLa-iRIGN) that could be rapidly and robustly induced by doxycycline (Dox) treatment to express RIG-I N-terminal effector, with concomitant production of type I interferons (IFN-Is). We also generated recombinant tri-segmented PICVs, rP18tri-LZ, and rP18tri-PZ, which encode LASV Z and PICV Z, respectively, as an extra mScarlet fusion protein that is nonessential for the viral life cycle. Upon infection, rP18tri-LZ consistently expressed viral genes at a higher level than rP18tri-PZ. rP18tri-LZ also showed a higher level of a viral infection than rP18tri-PZ did in HeLa-iRIGN cells, especially upon Dox induction. The heterologous Z gene did not alter viral growth in Vero and A549 cells by growth curve analysis, while LASV Z strongly increased and prolonged viral gene expression, especially in IFN-competent A549 cells. Our study provides important insights into the biological role of LASV Z-mediated RIG-I inhibition and implicates LASV Z as a potential virulence factor. IMPORTANCE Lassa virus (LASV) can cause lethal hemorrhagic fever disease in humans but other arenaviruses, such as Pichinde virus (PICV), do not cause obvious disease. We have previously shown that the Z protein of LASV but not of PICV can inhibit RIG-I, a cytosolic innate immune receptor. In this study, we developed a stable HeLa cell line that can be induced to express the RIG-I N-terminal effector domain, which allows for timely control of RIG-I activation. We also generated recombinant PICVs encoding LASV Z or PICV Z as an extra gene that is nonessential for the viral life cycle. Compared to PICV Z, LASV Z could increase viral gene expression and viral infection in an infectious arenavirus system, especially when RIG-I signaling is activated. Our study presented a convenient cell system to characterize RIG-I signaling and its antagonists and revealed LASV Z as a possible virulence factor and a potential antiviral target.


Subject(s)
Lassa virus , Viral Proteins/metabolism , HeLa Cells , Humans , Lassa Fever/virology , Lassa virus/pathogenicity , Lassa virus/physiology , Pichinde virus/genetics , Virulence Factors
3.
Nature ; 603(7899): 174-179, 2022 03.
Article in English | MEDLINE | ID: mdl-35173332

ABSTRACT

Lassa virus (LASV) is a human pathogen, causing substantial morbidity and mortality1,2. Similar to other Arenaviridae, it presents a class-I spike complex on its surface that facilitates cell entry. The virus's cellular receptor is matriglycan, a linear carbohydrate that is present on α-dystroglycan3,4, but the molecular mechanism that LASV uses to recognize this glycan is unknown. In addition, LASV and other arenaviruses have a unique signal peptide that forms an integral and functionally important part of the mature spike5-8; yet the structure, function and topology of the signal peptide in the membrane remain uncertain9-11. Here we solve the structure of a complete native LASV spike complex, finding that the signal peptide crosses the membrane once and that its amino terminus is located in the extracellular region. Together with a double-sided domain-switching mechanism, the signal peptide helps to stabilize the spike complex in its native conformation. This structure reveals that the LASV spike complex is preloaded with matriglycan, suggesting the mechanism of binding and rationalizing receptor recognition by α-dystroglycan-tropic arenaviruses. This discovery further informs us about the mechanism of viral egress and may facilitate the rational design of novel therapeutics that exploit this binding site.


Subject(s)
Dystroglycans , Lassa virus , Receptors, Virus , Viral Envelope Proteins , Dystroglycans/chemistry , Dystroglycans/metabolism , Humans , Lassa Fever/virology , Lassa virus/chemistry , Lassa virus/metabolism , Protein Conformation , Protein Sorting Signals , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization
4.
Emerg Microbes Infect ; 10(1): 2313-2325, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34792436

ABSTRACT

Lassa virus (LASV), a Risk Group-4 zoonotic haemorrhagic fever virus, affects sub-Saharan African countries. Lassa fever, caused by LASV, results in thousands of annual deaths. Although decades have elapsed since the identification of the Natal multimammate mouse (Mastomys natalensis) as a natural reservoir of LASV, little effort has been made to characterize LASV infection in its reservoir. The natural route of infection and transmission of LASV within M. natalensis remains unknown, and the clinical impact of LASV in M. natalensis is mostly undescribed. Herein, using an outbred colony of M. natalensis, we investigate the replication and dissemination dynamics of LASV in this reservoir following various inoculation routes. Inoculation with LASV, regardless of route, resulted in a systemic infection and accumulation of abundant LASV-RNA in many tissues. LASV infection in the Natal multimammate mice was subclinical, however, clinical chemistry values were transiently altered and immune infiltrates were observed histologically in lungs, spleens and livers, indicating a minor disease with coordinated immune responses are elicited, controlling infection. Intranasal infection resulted in unique virus tissue dissemination dynamics and heightened LASV shedding, compared to subcutaneous inoculation. Our study provides important insights into LASV infection in its natural reservoir using a contemporary infection system, demonstrating that specific inoculation routes result in disparate dissemination outcomes, suggesting intranasal inoculation is important in the maintenance of LASV in the natural reservoir, and emphasizes that selection of the appropriate inoculation route is necessary to examine aspects of viral replication, transmission and responses to zoonotic viruses in their natural reservoirs.


Subject(s)
Disease Reservoirs/veterinary , Lassa Fever/veterinary , Lassa virus/physiology , Murinae/virology , Rodent Diseases/virology , Viral Zoonoses/virology , Virus Shedding , Animals , Disease Reservoirs/virology , Female , Humans , Lassa Fever/transmission , Lassa Fever/virology , Lassa virus/genetics , Male , Murinae/physiology , Rodent Diseases/transmission , Viral Zoonoses/transmission
5.
Nat Commun ; 12(1): 5759, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599162

ABSTRACT

Lassa fever is a longstanding public health concern in West Africa. Recent molecular studies have confirmed the fundamental role of the rodent host (Mastomys natalensis) in driving human infections, but control and prevention efforts remain hampered by a limited baseline understanding of the disease's true incidence, geographical distribution and underlying drivers. Here, we show that Lassa fever occurrence and incidence is influenced by climate, poverty, agriculture and urbanisation factors. However, heterogeneous reporting processes and diagnostic laboratory access also appear to be important drivers of the patchy distribution of observed disease incidence. Using spatiotemporal predictive models we show that including climatic variability added retrospective predictive value over a baseline model (11% decrease in out-of-sample predictive error). However, predictions for 2020 show that a climate-driven model performs similarly overall to the baseline model. Overall, with ongoing improvements in surveillance there may be potential for forecasting Lassa fever incidence to inform health planning.


Subject(s)
Disease Reservoirs/virology , Epidemiological Monitoring , Lassa Fever/epidemiology , Lassa virus/pathogenicity , Murinae/virology , Animals , Climate , Geography , Humans , Incidence , Lassa Fever/transmission , Lassa Fever/virology , Nigeria/epidemiology , Poverty , Retrospective Studies , Spatio-Temporal Analysis , Urbanization
6.
Sci Rep ; 11(1): 20698, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667210

ABSTRACT

Lassa fever (LF) is a viral haemorrhagic fever endemic in West Africa and spread primarily by the multimammate rat, Mastomys natalensis. As there is no vaccine, reduction of rodent-human transmission is essential for disease control. As the household is thought to be a key site of transmission, understanding domestic risk factors for M. natalensis abundance is crucial. Rodent captures in conjunction with domestic surveys were carried out in 6 villages in an area of rural Upper Guinea with high LF endemicity. 120 rodent traps were set in rooms along a transect in each village for three nights, and the survey was administered in each household on the transects. This study was able to detect several domestic risk factors for increased rodent abundance in rural Upper Guinea. Regression analysis demonstrated that having > 8 holes (RR = 1.8 [1.0004-3.2, p = 0.048), the presence of rodent burrows (RR = 2.3 [1.6-3.23, p = 0.000003), and being in a multi-room square building (RR = 2.0 [1.3-2.9], p = 0.001) were associated with increased rodent abundance. The most addressable of these may be rodent burrows, as burrow patching is a relatively simple process that may reduce rodent entry. Further study is warranted to explicitly link domestic rodent abundance to LF risk, to better characterize domestic risk factors, and to evaluate how household rodent-proofing interventions could contribute to LF control.


Subject(s)
Lassa Fever/epidemiology , Lassa Fever/transmission , Rodentia/virology , Adult , Animals , Disease Reservoirs/virology , Female , Guinea/epidemiology , Humans , Lassa Fever/virology , Lassa virus/pathogenicity , Male , Middle Aged , Rats , Risk Factors , Rural Population
7.
Viruses ; 13(9)2021 08 25.
Article in English | MEDLINE | ID: mdl-34578260

ABSTRACT

Lassa fever virus (LASV) can cause life-threatening hemorrhagic fevers for which there are currently no vaccines or targeted treatments. The late Prof. Stefan Kunz, along with others, showed that the high-affinity host receptor for LASV, and other Old World and clade-C New World mammarenaviruses, is matriglycan-a linear repeating disaccharide of alternating xylose and glucuronic acid that is polymerized uniquely on α-dystroglycan by like-acetylglucosaminyltransferase-1 (LARGE1). Although α-dystroglycan is ubiquitously expressed, LASV preferentially infects vascular endothelia and professional phagocytic cells, which suggests that viral entry requires additional cell-specific factors. In this review, we highlight the work of Stefan Kunz detailing the molecular mechanism of LASV binding and discuss the requirements of receptors, such as tyrosine kinases, for internalization through apoptotic mimicry.


Subject(s)
Dystroglycans/metabolism , Glucuronic Acid/chemistry , Lassa virus/metabolism , Polymers/metabolism , Virus Attachment , Xylose/chemistry , Animals , Dystroglycans/chemistry , Glucuronic Acid/metabolism , Humans , Lassa Fever/virology , Lassa virus/genetics , Mice , Polymers/chemistry , Receptors, Virus , Virus Internalization , Xylose/metabolism
8.
Viruses ; 13(9)2021 09 03.
Article in English | MEDLINE | ID: mdl-34578344

ABSTRACT

Lassa virus (LASV)-a member of the family Arenaviridae-causes Lassa fever in humans and is endemic in West Africa. Currently, no approved drugs are available. We screened 2480 small compounds for their potential antiviral activity using pseudotyped vesicular stomatitis virus harboring the LASV glycoprotein (VSV-LASVGP) and a related prototypic arenavirus, lymphocytic choriomeningitis virus (LCMV). Follow-up studies confirmed that CP100356 hydrochloride (CP100356), a specific P-glycoprotein (P-gp) inhibitor, suppressed VSV-LASVGP, LCMV, and LASV infection with half maximal inhibitory concentrations of 0.52, 0.54, and 0.062 µM, respectively, without significant cytotoxicity. Although CP100356 did not block receptor binding at the cell surface, it inhibited low-pH-dependent membrane fusion mediated by arenavirus glycoproteins. P-gp downregulation did not cause a significant reduction in either VSV-LASVGP or LCMV infection, suggesting that P-gp itself is unlikely to be involved in arenavirus entry. Finally, our data also indicate that CP100356 inhibits the infection by other mammarenaviruses. Thus, our findings suggest that CP100356 can be considered as an effective virus entry inhibitor for LASV and other highly pathogenic mammarenaviruses.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Arenaviridae/metabolism , Isoquinolines/pharmacology , Lassa virus/drug effects , Quinazolines/pharmacology , Virus Internalization/drug effects , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Humans , Lassa Fever/drug therapy , Lassa Fever/virology , Lymphocytic choriomeningitis virus , Receptors, Virus , Vero Cells , Vesicular Stomatitis/virology , Viral Fusion Protein Inhibitors/pharmacology
9.
Viruses ; 13(8)2021 08 13.
Article in English | MEDLINE | ID: mdl-34452470

ABSTRACT

While investigating a signal of adaptive evolution in humans at the gene LARGE, we encountered an intriguing finding by Dr. Stefan Kunz that the gene plays a critical role in Lassa virus binding and entry. This led us to pursue field work to test our hypothesis that natural selection acting on LARGE-detected in the Yoruba population of Nigeria-conferred resistance to Lassa Fever in some West African populations. As we delved further, we conjectured that the "emerging" nature of recently discovered diseases like Lassa fever is related to a newfound capacity for detection, rather than a novel viral presence, and that humans have in fact been exposed to the viruses that cause such diseases for much longer than previously suspected. Dr. Stefan Kunz's critical efforts not only laid the groundwork for this discovery, but also inspired and catalyzed a series of events that birthed Sentinel, an ambitious and large-scale pandemic prevention effort in West Africa. Sentinel aims to detect and characterize deadly pathogens before they spread across the globe, through implementation of its three fundamental pillars: Detect, Connect, and Empower. More specifically, Sentinel is designed to detect known and novel infections rapidly, connect and share information in real time to identify emerging threats, and empower the public health community to improve pandemic preparedness and response anywhere in the world. We are proud to dedicate this work to Stefan Kunz, and eagerly invite new collaborators, experts, and others to join us in our efforts.


Subject(s)
Disaster Planning , Lassa Fever/epidemiology , Lassa virus/physiology , Africa, Western/epidemiology , Disaster Planning/methods , Humans , Lassa Fever/genetics , Lassa Fever/prevention & control , Lassa Fever/virology , Lassa virus/genetics , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/immunology , Nigeria/epidemiology , Pandemics , Polymorphism, Genetic , Receptors, Virus/genetics , Receptors, Virus/immunology
10.
PLoS Negl Trop Dis ; 15(7): e0009522, 2021 07.
Article in English | MEDLINE | ID: mdl-34237063

ABSTRACT

Ribavirin is the only available Lassa fever treatment. The rationale for using ribavirin is based on one clinical study conducted in the early 1980s. However, reanalysis of previous unpublished data reveals that ribavirin may actually be harmful in some Lassa fever patients. An urgent reevaluation of ribavirin is therefore needed.


Subject(s)
Antiviral Agents/administration & dosage , Lassa Fever/drug therapy , Ribavirin/administration & dosage , Humans , Lassa Fever/virology
11.
Ann Clin Microbiol Antimicrob ; 20(1): 29, 2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33894784

ABSTRACT

Lassa fever (LF), a zoonotic illness, represents a public health burden in West African countries where the Lassa virus (LASV) circulates among rodents. Human exposure hinges significantly on LASV ecology, which is in turn shaped by various parameters such as weather seasonality and even virus and rodent-host genetics. Furthermore, human behaviour, despite playing a key role in the zoonotic nature of the disease, critically affects either the spread or control of human-to-human transmission. Previous estimations on LF burden date from the 80s and it is unclear how the population expansion and the improvement on diagnostics and surveillance methods have affected such predictions. Although recent data have contributed to the awareness of epidemics, the real impact of LF in West African communities will only be possible with the intensification of interdisciplinary efforts in research and public health approaches. This review discusses the causes and consequences of LF from a One Health perspective, and how the application of this concept can improve the surveillance and control of this disease in West Africa.


Subject(s)
Disease Reservoirs/virology , Lassa Fever/epidemiology , Lassa Fever/transmission , Lassa Fever/virology , Lassa virus , One Health , Rodentia/virology , Africa, Western/epidemiology , Animals , Humans , Lassa Fever/prevention & control , Public Health
12.
Immunobiology ; 226(3): 152076, 2021 05.
Article in English | MEDLINE | ID: mdl-33689957

ABSTRACT

BACKGROUND: The increasing trends of morbidity and mortality of Lassa fever is becoming more alarming in Nigeria. Information about immune response to the virus is limited. At exposure, the level of immunity plays a vital role in the vulnerability of individuals infected. OBJECTIVE: Investigating the immune status of health workers, infected cases and contacts of infected cases of Lassa fever in Ondo State. STUDY DESIGN: Blood samples were collected from 233 individuals comprising 102 health workers, 22 infected cases and 109 contacts of infected cases from Owo and Ose Local Government Areas and transported in triple level packaging. Plasma samples were analyzed for IgG and IgM markers using ReLASV® Pan-Lassa NP IgG/IgM ELISA Kit (Zalgen Labs, LLC, USA) while RNAs extracted from IgM positive samples were analyzed for LASV RNA according to manufacturers' instructions. RESULT: Among the health workers, 20/102 (19.6%) and 2/102 (2.0%) were IgG and IgM positive respectively. While 16/22 (72.7%) and 14/22 (63.6%) were IgG and IgM positive respectively among the infected cases. Of the contacts of infected cases screened, 64/109 (58.7%) were IgG positive while 4/109 (3.7%) were positive for IgM. There was no detectable LASV RNA in the samples analyzed. CONCLUSION: These findings suggest that majority of the health workers are naïve to the virus and hence may be prone to the viral infection. It could also be suggestive that a good personal protective procedure is been practiced by the health workers, hence the low exposure. However, most of the contacts of infected cases show exposure to the virus.


Subject(s)
Contact Tracing , Health Personnel , Lassa Fever/epidemiology , Lassa Fever/virology , Lassa virus , Antibodies, Viral/blood , Antibodies, Viral/immunology , Enzyme-Linked Immunosorbent Assay/methods , Humans , Lassa Fever/diagnosis , Lassa Fever/transmission , Lassa virus/immunology , Mass Screening , Nigeria/epidemiology , Public Health Surveillance
13.
PLoS Comput Biol ; 17(3): e1008811, 2021 03.
Article in English | MEDLINE | ID: mdl-33657095

ABSTRACT

Forecasting the risk of pathogen spillover from reservoir populations of wild or domestic animals is essential for the effective deployment of interventions such as wildlife vaccination or culling. Due to the sporadic nature of spillover events and limited availability of data, developing and validating robust, spatially explicit, predictions is challenging. Recent efforts have begun to make progress in this direction by capitalizing on machine learning methodologies. An important weakness of existing approaches, however, is that they generally rely on combining human and reservoir infection data during the training process and thus conflate risk attributable to the prevalence of the pathogen in the reservoir population with the risk attributed to the realized rate of spillover into the human population. Because effective planning of interventions requires that these components of risk be disentangled, we developed a multi-layer machine learning framework that separates these processes. Our approach begins by training models to predict the geographic range of the primary reservoir and the subset of this range in which the pathogen occurs. The spillover risk predicted by the product of these reservoir specific models is then fit to data on realized patterns of historical spillover into the human population. The result is a geographically specific spillover risk forecast that can be easily decomposed and used to guide effective intervention. Applying our method to Lassa virus, a zoonotic pathogen that regularly spills over into the human population across West Africa, results in a model that explains a modest but statistically significant portion of geographic variation in historical patterns of spillover. When combined with a mechanistic mathematical model of infection dynamics, our spillover risk model predicts that 897,700 humans are infected by Lassa virus each year across West Africa, with Nigeria accounting for more than half of these human infections.


Subject(s)
Disease Reservoirs/virology , Lassa Fever , Lassa virus , Models, Biological , Africa, Western , Animals , Animals, Wild/virology , Computational Biology , Ecology , Humans , Lassa Fever/epidemiology , Lassa Fever/transmission , Lassa Fever/veterinary , Lassa Fever/virology , Machine Learning , Models, Statistical , Risk , Rodentia/virology
14.
PLoS Negl Trop Dis ; 15(3): e0009212, 2021 03.
Article in English | MEDLINE | ID: mdl-33730025

ABSTRACT

As a consequence of the Ebola outbreak, human-animal contact has gained importance for zoonotic transmission surveillance. In Faranah (Upper Guinea), daily life is intertwined with rodents, such as the Natal multimammate mouse, Mastomys natalensis; a reservoir for Lassa virus (LASV). However, this contact is rarely perceived as a health risk by residents, although Lassa fever (LF) is known to be endemic to this region. Conversely, these observations remain a great concern for global health agendas. Drawing on ethnographic research involving interviews, focus group discussions, participant observations, and informal discussions over four months, we first identified factors that motivated children to hunt and consume rodents in Faranah villages, and thereafter, explored the knowledge of LF infection in children and their parents. Furthermore, we studied two dimensions of human-rodent encounters: 1) space-time of interaction and 2) factors that allowed the interaction to occur and their materiality. This approach allowed us to contextualize child-rodent contacts beyond domestic limits in the fallow fields, swamps, and at other times for this practice. A close look at these encounters provided information on rodent trapping, killing, and manipulation of cooking techniques and the risk these activities posed for the primary transmission of LASV. This research facilitated the understanding of children's exposure to M. natalensis during hunting sessions and the importance of rodent hunting, which is a part of their boyish identity in rural areas. Determination of when, where, why, and how children, rodents, and environments interacted allowed us to understand the exposures and risks important for human and animal surveillance programs in the Lassa-endemic region.


Subject(s)
Disease Reservoirs/veterinary , Endemic Diseases , Lassa Fever/epidemiology , Lassa Fever/transmission , Rodentia , Animals , Child , Disease Reservoirs/virology , Guinea/epidemiology , Humans , Lassa Fever/virology , Lassa virus/immunology , Rodent Control , Zoonoses
15.
Lancet Glob Health ; 9(4): e469-e478, 2021 04.
Article in English | MEDLINE | ID: mdl-33740408

ABSTRACT

BACKGROUND: Lassa fever is a viral haemorrhagic fever endemic in parts of west Africa. New treatments are needed to decrease mortality, but pretrial reference data on the disease characteristics are scarce. We aimed to document baseline characteristics and outcomes for patients hospitalised with Lassa fever in Nigeria. METHODS: We did a prospective cohort study (LASCOPE) at the Federal Medical Centre in Owo, Nigeria. All patients admitted with confirmed Lassa fever were invited to participate and asked to give informed consent. Patients of all ages, including newborn infants, were eligible for inclusion, as were pregnant women. All participants received standard supportive care and intravenous ribavirin according to Nigeria Centre for Disease Control guidelines and underwent systematic biological monitoring for 30 days. Patients' characteristics, care received, mortality, and associated factors were recorded using standard WHO forms. We used univariable and multivariable logistic regression models to investigate an association between baseline characteristics and mortality at day 30. FINDINGS: Between April 5, 2018, and March 15, 2020, 534 patients with confirmed Lassa fever were admitted to hospital, of whom 510 (96%) gave consent and were included in the analysis. The cohort included 258 (51%) male patients, 252 (49%) female patients, 426 (84%) adults, and 84 (16%) children (younger than 18 years). The median time between first symptoms and hospital admission was 8 days (IQR 7-13). At baseline, 176 (38%) of 466 patients had a Lassa fever RT-PCR cycle threshold (Ct) lower than 30. From admission to end of follow-up, 120 (25%) of 484 reached a National Early Warning Score (second version; NEWS2) of 7 or higher, 67 (14%) of 495 reached a Kidney Disease-Improving Global Outcome (KDIGO) stage of 2 or higher, and 41 (8%) of 510 underwent dialysis. All patients received ribavirin for a median of 10 days (IQR 9-13). 62 (12%) patients died (57 [13%] adults and five [6%] children). The median time to death was 3 days (1-6). The baseline factors independently associated with mortality were the following: age 45 years or older (adjusted odds ratio 16·30, 95% CI 5·31-50·30), NEWS2 of 7 or higher (4·79, 1·75-13·10), KDIGO grade 2 or higher (7·52, 2·66-21·20), plasma alanine aminotransferase 3 or more times the upper limit of normal (4·96, 1·69-14·60), and Lassa fever RT-PCR Ct value lower than 30 (4·65, 1·50-14·50). INTERPRETATION: Our findings comprehensively document clinical and biological characteristics of patients with Lassa fever and their relationship with mortality, providing prospective estimates that could be useful for designing future therapeutic trials. Such trials comparing new Lassa fever treatments to a standard of care should take no more than 15% as the reference mortality rate and consider adopting a combination of mortality and need for dialysis as the primary endpoint. FUNDING: Institut National de la Santé et de la Recherche Médicale, University of Oxford, EU, UK Department for International Development, Wellcome Trust, French Ministry of Foreign Affairs, Agence Nationale de Recherches sur le SIDA et les hépatites virales, French National Research Institute for Sustainable Development.


Subject(s)
Lassa Fever/mortality , Lassa virus/isolation & purification , Palliative Care , Ribavirin/administration & dosage , Administration, Intravenous , Adolescent , Adult , Age Factors , Child , Child, Preschool , DNA, Viral/isolation & purification , Female , Follow-Up Studies , Hospital Mortality , Hospitalization/statistics & numerical data , Humans , Infant , Infant, Newborn , Kaplan-Meier Estimate , Lassa Fever/diagnosis , Lassa Fever/therapy , Lassa Fever/virology , Lassa virus/genetics , Male , Middle Aged , Nigeria/epidemiology , Pregnancy , Prognosis , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction/statistics & numerical data , Risk Factors , Severity of Illness Index , Time Factors , Treatment Outcome , Young Adult
16.
Viruses ; 13(2)2021 01 30.
Article in English | MEDLINE | ID: mdl-33573250

ABSTRACT

Defective interfering particles (DIPs) are naturally occurring products during virus replication in infected cells. DIPs contain defective viral genomes (DVGs) and interfere with replication and propagation of their corresponding standard viral genomes by competing for viral and cellular resources, as well as promoting innate immune antiviral responses. Consequently, for many different viruses, including mammarenaviruses, DIPs play key roles in the outcome of infection. Due to their ability to broadly interfere with viral replication, DIPs are attractive tools for the development of a new generation of biologics to target genetically diverse and rapidly evolving viruses. Here, we provide evidence that in cells infected with the Lassa fever (LF) vaccine candidate ML29, a reassortant that carries the nucleoprotein (NP) and glycoprotein (GP) dominant antigens of the pathogenic Lassa virus (LASV) together with the L polymerase and Z matrix protein of the non-pathogenic genetically related Mopeia virus (MOPV), L-derived truncated RNA species are readily detected following infection at low multiplicity of infection (MOI) or in persistently-infected cells originally infected at high MOI. In the present study, we show that expression of green fluorescent protein (GFP) driven by a tri-segmented form of the mammarenavirus lymphocytic choriomeningitis virus (r3LCMV-GFP/GFP) was strongly inhibited in ML29-persistently infected cells, and that the magnitude of GFP suppression was dependent on the passage history of the ML29-persistently infected cells. In addition, we found that DIP-enriched ML29 was highly attenuated in immunocompetent CBA/J mice and in Hartley guinea pigs. Likewise, STAT-1-/- mice, a validated small animal model for human LF associated hearing loss sequelae, infected with DIP-enriched ML29 did not exhibit any hearing abnormalities throughout the observation period (62 days).


Subject(s)
Lassa Fever/prevention & control , Lassa virus/immunology , Viral Vaccines/immunology , Animals , Female , Genome, Viral , Guinea Pigs , Humans , Lassa Fever/genetics , Lassa Fever/immunology , Lassa Fever/virology , Lassa virus/genetics , Lassa virus/physiology , Mice , Mice, Inbred CBA , RNA, Viral/genetics , RNA, Viral/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virus Replication
17.
PLoS Comput Biol ; 17(1): e1008535, 2021 01.
Article in English | MEDLINE | ID: mdl-33411731

ABSTRACT

Lassa fever is an haemorrhagic fever caused by Lassa virus (LASV). There is no vaccine approved against LASV and the only recommended antiviral treatment relies on ribavirin, despite limited evidence of efficacy. Recently, the nucleotide analogue favipiravir showed a high antiviral efficacy, with 100% survival obtained in an otherwise fully lethal non-human primate (NHP) model of Lassa fever. However the mechanism of action of the drug is not known and the absence of pharmacokinetic data limits the translation of these results to the human setting. Here we aimed to better understand the antiviral effect of favipiravir by developping the first mathematical model recapitulating Lassa viral dynamics and treatment. We analyzed the viral dynamics in 24 NHPs left untreated or treated with ribavirin or favipiravir, and we put the results in perspective with those obtained with the same drugs in the context of Ebola infection. Our model estimates favipiravir EC50 in vivo to 2.89 µg.mL-1, which is much lower than what was found against Ebola virus. The main mechanism of action of favipiravir was to decrease virus infectivity, with an efficacy of 91% at the highest dose. Based on our knowledge acquired on the drug pharmacokinetics in humans, our model predicts that favipiravir doses larger than 1200 mg twice a day should have the capability to strongly reduce the production infectious virus and provide a milestone towards a future use in humans.


Subject(s)
Amides , Antiviral Agents , Lassa Fever/virology , Lassa virus , Pyrazines , Ribavirin , Amides/pharmacokinetics , Amides/pharmacology , Amides/therapeutic use , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Female , Host-Pathogen Interactions/drug effects , Lassa Fever/drug therapy , Lassa virus/drug effects , Lassa virus/pathogenicity , Lassa virus/physiology , Macaca fascicularis , Models, Biological , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Pyrazines/therapeutic use , Ribavirin/pharmacokinetics , Ribavirin/pharmacology , Ribavirin/therapeutic use , Viral Load/drug effects
18.
Commun Biol ; 4(1): 27, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33398113

ABSTRACT

Lassa virus (LASV) is endemic in West Africa and induces a viral hemorrhagic fever (VHF) with up to 30% lethality among clinical cases. The mechanisms involved in control of Lassa fever or, in contrast, the ensuing catastrophic illness and death are poorly understood. We used the cynomolgus monkey model to reproduce the human disease with asymptomatic to mild or fatal disease. After initial replication at the inoculation site, LASV reached the secondary lymphoid organs. LASV did not spread further in nonfatal disease and was rapidly controlled by balanced innate and T-cell responses. Systemic viral dissemination occurred during severe disease. Massive replication, a cytokine/chemokine storm, defective T-cell responses, and multiorgan failure were observed. Clinical, biological, immunological, and transcriptomic parameters resembled those observed during septic-shock syndrome, suggesting that similar pathogenesis is induced during Lassa fever. The outcome appears to be determined early, as differentially expressed genes in PBMCs were associated with fatal and non-fatal Lassa fever outcome very early after infection. These results provide a full characterization and important insights into Lassa fever pathogenesis and could help to develop early diagnostic tools.


Subject(s)
Disease Models, Animal , Lassa Fever/immunology , Lassa Fever/virology , Macaca fascicularis , Adaptive Immunity , Animals , Biomarkers/metabolism , Female , Immunity, Innate , Lassa Fever/blood , Lassa Fever/pathology , Lung/pathology , Lymphoid Tissue/pathology , Male , Transcriptome
19.
Curr Comput Aided Drug Des ; 17(1): 32-37, 2021.
Article in English | MEDLINE | ID: mdl-31648648

ABSTRACT

BACKGROUND: Lassa Virus (LV) infection is an endemic disease from West Africa posing threat to the entire world. A thorough understanding of the mechanistic workings of the genome products of LV may be a key to develop drug candidates for the treatment of LV infection. METHODS: Molecular dynamic simulation has been used to provide insight into the mechanistic basis for total loss of ssRNA interaction in Nucleoprotein (NP) K309A, partial loss in S247A, and no loss in S237A by following the hydrogen bond interaction, water influx into the ssRNA pocket and glycosidic torsion angle (χ) of the ssRNA. RESULTS: The results revealed that K309A mutation is associated with a complete loss of saltbridge interaction between lysine e-amino and U4-O2P phosphodiester linkage but not in S237A where S247-OG atom played a redundant role. S247A is also associated with partial loss of hydrogen bond between OG atom of S247 and C5-O2P phosphodiester bond as T178-OG1 group seems to have a seemingly redundant interaction with C5-O2P. While S247A is only associated with the alteration of χ rotation in U6/C7, both K309A and S247 but not S237A is associated with increased water influx into the ssRNA binding pocket. CONCLUSION: K309A mutation may result in non-viable Lassa viron as a loss of ssRNA interaction may negatively affect genome biochemistry, semi-viable Lassa viron in S247A mutation may be due to the loss of 3D arrangement of ssRNA due to splayed out nucleotides.


Subject(s)
Lassa Fever/virology , Lassa virus/genetics , Nucleoproteins/genetics , RNA, Viral/genetics , Genome, Viral/genetics , Hydrogen Bonding , Molecular Dynamics Simulation , Mutation , Water/chemistry
20.
Am J Trop Med Hyg ; 103(5): 2085-2090, 2020 11.
Article in English | MEDLINE | ID: mdl-32959767

ABSTRACT

Diseases caused by many highly pathogenic viruses, including Ebola virus (EBOV) and Lassa virus (LASV), present with nonspecific signs and symptoms that overlap with common tropical diseases such as malaria. Initial diagnostic tests performed on patients under investigation for viral hemorrhagic fevers routinely include analysis of peripheral blood smears to detect and quantify Plasmodium species. In light of recent and ongoing Ebola virus disease and Lassa fever epidemics, clinical laboratories around the world require protocols for dealing with highly infectious specimens from patients with suspected or confirmed high-consequence diseases. Few validated protocols for safe analysis of peripheral blood smears are available, revealing a need for further research. In this study, we evaluated the performance of two plastic microscope slide types that offer safe alternatives to glass slides, determined the temporal parameters required to inactivate EBOV and LASV in thin blood smears by methanol fixation, and assessed the virucidal activity of Giemsa stain. Both types of plastic microscope slides performed optimally; there were no significant differences in blood cell morphology or tinctorial properties nor were differences noted in Plasmodium ovale morphology or staining, when compared with glass slides. For both EBOV and LASV, viable viruses were not detected in thin blood smears following fixation in absolute methanol for at least 2 minutes. By contrast, viable EBOV and LASV were recovered from all Giemsa-stained thick blood smears.


Subject(s)
Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/virology , Lassa Fever/virology , Lassa virus/drug effects , Methanol/pharmacology , Virus Inactivation/drug effects , Azure Stains , Specimen Handling
SELECTION OF CITATIONS
SEARCH DETAIL
...