Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Publication year range
1.
Lancet ; 401(10377): 673-687, 2023 02 25.
Article in English | MEDLINE | ID: mdl-36682374

ABSTRACT

The COVID-19 pandemic has exposed faults in the way we assess preparedness and response capacities for public health emergencies. Existing frameworks are limited in scope, and do not sufficiently consider complex social, economic, political, regulatory, and ecological factors. One Health, through its focus on the links among humans, animals, and ecosystems, is a valuable approach through which existing assessment frameworks can be analysed and new ways forward proposed. Although in the past few years advances have been made in assessment tools such as the International Health Regulations Joint External Evaluation, a rapid and radical increase in ambition is required. To sufficiently account for the range of complex systems in which health emergencies occur, assessments should consider how problems are defined across stakeholders and the wider sociopolitical environments in which structures and institutions operate. Current frameworks do little to consider anthropogenic factors in disease emergence or address the full array of health security hazards across the social-ecological system. A complex and interdependent set of challenges threaten human, animal, and ecosystem health, and we cannot afford to overlook important contextual factors, or the determinants of these shared threats. Health security assessment frameworks should therefore ensure that the process undertaken to prioritise and build capacity adheres to core One Health principles and that interventions and outcomes are assessed in terms of added value, trade-offs, and cobenefits across human, animal, and environmental health systems.


Subject(s)
COVID-19 , One Health , Animals , Humans , Global Health , Ecosystem , Emergencies , Pandemics
2.
Nature ; 544(7650): 309-315, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28405027

ABSTRACT

The 2013-2016 West African epidemic caused by the Ebola virus was of unprecedented magnitude, duration and impact. Here we reconstruct the dispersal, proliferation and decline of Ebola virus throughout the region by analysing 1,610 Ebola virus genomes, which represent over 5% of the known cases. We test the association of geography, climate and demography with viral movement among administrative regions, inferring a classic 'gravity' model, with intense dispersal between larger and closer populations. Despite attenuation of international dispersal after border closures, cross-border transmission had already sown the seeds for an international epidemic, rendering these measures ineffective at curbing the epidemic. We address why the epidemic did not spread into neighbouring countries, showing that these countries were susceptible to substantial outbreaks but at lower risk of introductions. Finally, we reveal that this large epidemic was a heterogeneous and spatially dissociated collection of transmission clusters of varying size, duration and connectivity. These insights will help to inform interventions in future epidemics.


Subject(s)
Ebolavirus/genetics , Ebolavirus/physiology , Genome, Viral/genetics , Hemorrhagic Fever, Ebola/transmission , Hemorrhagic Fever, Ebola/virology , Climate , Disease Outbreaks/statistics & numerical data , Ebolavirus/isolation & purification , Geography , Hemorrhagic Fever, Ebola/epidemiology , Humans , Internationality , Linear Models , Molecular Epidemiology , Phylogeny , Travel/legislation & jurisprudence , Travel/statistics & numerical data
3.
J Public Health (Oxf) ; 45(Suppl 1): i63-i70, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38127562

ABSTRACT

BACKGROUND: For effective Public Health measures, factors that influence Knowledge, Attitudes and Practices (KAP) need to be understood. In this paper, we document the relationship between levels of education on the KAP towards COVID-19 among the population of Edo State, Nigeria. METHODS: A cross-sectional KAP questionnaire was delivered across 13 communities. The study population was dichotomized into those who had or had not completed secondary education. Chi-square tests were conducted to determine statistical significance. RESULTS: Of the 538 participants, 30% had completed secondary education. Those with secondary education were significantly more likely to recognize symptoms of COVID-19 (P < 0.001). For attitudes, only one option showed a statistically significant association with 78.4% of those who did not complete secondary education having a high confidence in the church compared with 66% of those who had (P = 0.022). For practices, those who have completed secondary education practiced all of the supplied measures more than those who had not, except for taking herbal supplements. DISCUSSION: Higher levels of education impacts on knowledge and practices but has less impact on attitudes regarding how well certain individuals/organizations can handle the pandemic. Findings highlighted the importance of tailoring public health communication and strategies to local populations to improve the efficacy.


Subject(s)
COVID-19 , Humans , COVID-19/epidemiology , Nigeria/epidemiology , Health Knowledge, Attitudes, Practice , Cross-Sectional Studies , Educational Status , Surveys and Questionnaires
4.
Dev World Bioeth ; 23(3): 242-251, 2023 09.
Article in English | MEDLINE | ID: mdl-35944158

ABSTRACT

The COVID-19 pandemic has reinforced the critical role of ethics and community engagement in designing and conducting clinical research during infectious disease outbreaks where no vaccine or treatment already exists. In reviewing current practices across Africa, we distinguish between three distinct roles for community engagement in clinical research that are often conflated: 1) the importance of community engagement for identifying and honouring cultural sensitivities; 2) the importance of recognising the socio-political context in which the research is proposed; and 3) the importance of understanding what is in the interest of communities recruited to research according to their own views and values. By making these distinctions, we show that current practice of clinical research could draw on anthropology in ways which are sometimes unnecessary to solicit local cultural values, overlook the importance of socio-political contexts and wider societal structures within which it works, potentially serving to reinforce unjust political or social regimes, and threaten to cast doubt on the trustworthiness of the research. We argue that more discerning anthropological engagement as well as wider collaboration with other social scientists and those working in the humanities is urgently needed to improve the ethics of current biomedical and pharmaceutical research practice in Africa.


Subject(s)
COVID-19 , Pandemics , Humans , Africa , Anthropology , Disease Outbreaks , Pandemics/prevention & control , Clinical Trials as Topic
5.
J Clin Microbiol ; 60(6): e0007522, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35574677

ABSTRACT

Sensitive and specific serological tests are mandatory for epidemiological studies evaluating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) prevalence as well as coronavirus disease 2019 (COVID-19) morbidity and mortality rates. The accuracy of results is challenged by antibody waning after convalescence and by cross-reactivity induced by previous infections with other pathogens. By employing a patented platform technology based on capturing antigen-antibody complexes with a solid-phase-bound Fcγ receptor (FcγR) and truncated nucleocapsid protein as the antigen, two SARS-CoV-2 IgG enzyme-linked immunosorbent assays (ELISAs), featuring different serum and antigen dilutions, were developed. Validation was performed using a serum panel comprising 213 longitudinal samples from 35 COVID-19 patients and a negative-control panel consisting of 790 pre-COVID-19 samples from different regions of the world. While both assays show similar diagnostic sensitivities in the early convalescent phase, ELISA 2 (featuring a higher serum concentration) enables SARS-CoV-2 IgG antibody detection for a significantly longer time postinfection (≥15 months). Correspondingly, analytical sensitivity referenced to indirect immunofluorescence testing (IIFT) is significantly higher for ELISA 2 in samples with a titer of ≤1:640; for high-titer samples, a prozone effect is observed for ELISA 2. The specificities of both ELISAs were excellent not only for pre-COVID-19 serum samples from Europe, Asia, and South America but also for several challenging African sample panels. The SARS-CoV-2 IgG FcγR ELISAs, methodically combining antigen-antibody binding in solution and isotype-specific detection of immune complexes, are valuable tools for seroprevalence studies requiring the (long-term) detection of anti-SARS-CoV-2 IgG antibodies in populations with a challenging immunological background and/or in which spike-protein-based vaccine programs have been rolled out.


Subject(s)
COVID-19 , Receptors, IgG , Antibodies, Viral , COVID-19/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Humans , Immunoglobulin G , Nucleocapsid Proteins , SARS-CoV-2 , Sensitivity and Specificity , Seroepidemiologic Studies , Spike Glycoprotein, Coronavirus
6.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32817220

ABSTRACT

Lassa fever (LF) is a zoonotic viral hemorrhagic fever caused by Lassa virus (LASV), which is endemic to West African countries. Previous studies have suggested an important role for T-cell-mediated immunopathology in LF pathogenesis, but the mechanisms by which T cells influence disease severity and outcome are not well understood. Here, we present a multiparametric analysis of clinical immunology data collected during the 2017-2018 Lassa fever outbreak in Nigeria. During the acute phase of LF, we observed robust activation of the polyclonal T-cell repertoire, which included LASV-specific and antigenically unrelated T cells. However, severe and fatal LF cases were characterized by poor LASV-specific effector T-cell responses. Severe LF was also characterized by the presence of circulating T cells with homing capacity to inflamed tissues, including the gut mucosa. These findings in LF patients were recapitulated in a mouse model of LASV infection, in which mucosal exposure resulted in remarkably high lethality compared to skin exposure. Taken together, our findings indicate that poor LASV-specific T-cell responses and activation of nonspecific T cells with homing capacity to inflamed tissues are associated with severe LF.IMPORTANCE Lassa fever may cause severe disease in humans, in particular in areas of endemicity like Sierra Leone and Nigeria. Despite its public health importance, the pathophysiology of Lassa fever in humans is poorly understood. Here, we present clinical immunology data obtained in the field during the 2018 Lassa fever outbreak in Nigeria indicating that severe Lassa fever is associated with activation of T cells antigenically unrelated to Lassa virus and poor Lassa virus-specific effector T-cell responses. Mechanistically, we show that these bystander T cells express defined tissue homing signatures that suggest their recruitment to inflamed tissues and a putative role of these T cells in immunopathology. These findings open a window of opportunity to consider T-cell targeting as a potential postexposure therapeutic strategy against severe Lassa fever, a hypothesis that could be tested in relevant animal models, such as nonhuman primates.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Outbreaks , Intestinal Mucosa/immunology , Lassa Fever/immunology , Lassa virus/pathogenicity , Lymphocyte Activation , Adolescent , Adult , Aged , Animals , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Child , Child, Preschool , Female , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Infant , Infant, Newborn , Integrin beta1/genetics , Integrin beta1/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lassa Fever/genetics , Lassa Fever/mortality , Lassa Fever/virology , Lassa virus/growth & development , Lassa virus/immunology , Lysosomal-Associated Membrane Protein 1/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Male , Mice , Middle Aged , Nigeria/epidemiology , Retrospective Studies , Severity of Illness Index , Skin/immunology , Skin/pathology , Skin/virology , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
Trop Med Int Health ; 26(6): 621-631, 2021 06.
Article in English | MEDLINE | ID: mdl-33666297

ABSTRACT

OBJECTIVES: Specific serological tests are mandatory for reliable SARS-CoV-2 diagnostics and seroprevalence studies. Here, we assess the specificities of four commercially available SARS-CoV-2 IgG ELISAs in serum/plasma panels originating from Africa, South America, and Europe. METHODS: 882 serum/plasma samples collected from symptom-free donors before the COVID-19 pandemic in three African countries (Ghana, Madagascar, Nigeria), Colombia, and Germany were analysed with three nucleocapsid-based ELISAs (Euroimmun Anti-SARS-CoV-2-NCP IgG, EDI™ Novel Coronavirus COVID-19 IgG, Mikrogen recomWell SARS-CoV-2 IgG), one spike/S1-based ELISA (Euroimmun Anti-SARS-CoV-2 IgG), and in-house common cold CoV ELISAs. RESULTS: High specificity was confirmed for all SARS-CoV-2 IgG ELISAs for Madagascan (93.4-99.4%), Colombian (97.8-100.0%), and German (95.9-100.0%) samples. In contrast, specificity was much lower for the Ghanaian and Nigerian serum panels (Ghana: NCP-based assays 77.7-89.7%, spike/S1-based assay 94.3%; Nigeria: NCP-based assays 39.3-82.7%, spike/S1-based assay 90.7%). 15 of 600 African sera were concordantly classified as positive in both the NCP-based and the spike/S1-based Euroimmun ELISA, but did not inhibit spike/ACE2 binding in a surrogate virus neutralisation test. IgG antibodies elicited by previous infections with common cold CoVs were found in all sample panels, including those from Madagascar, Colombia, and Germany and thus do not inevitably hamper assay specificity. Nevertheless, high levels of IgG antibodies interacting with OC43 NCP were found in all 15 SARS-CoV-2 NCP/spike/S1 ELISA positive sera. CONCLUSIONS: Depending on the chosen antigen and assay protocol, SARS-CoV-2 IgG ELISA specificity may be significantly reduced in certain populations probably due to interference of immune responses to endemic pathogens like other viruses or parasites.


Subject(s)
Antibodies, Viral/blood , COVID-19/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Immunoglobulin G/blood , Adolescent , Adult , COVID-19/virology , Child , Child, Preschool , Colombia , Coronavirus Nucleocapsid Proteins/immunology , Female , Germany , Ghana , Humans , Madagascar , Male , Middle Aged , Nigeria , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Sensitivity and Specificity , Spike Glycoprotein, Coronavirus/immunology , Young Adult
8.
Nature ; 524(7563): 97-101, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26083749

ABSTRACT

West Africa is currently witnessing the most extensive Ebola virus (EBOV) outbreak so far recorded. Until now, there have been 27,013 reported cases and 11,134 deaths. The origin of the virus is thought to have been a zoonotic transmission from a bat to a two-year-old boy in December 2013 (ref. 2). From this index case the virus was spread by human-to-human contact throughout Guinea, Sierra Leone and Liberia. However, the origin of the particular virus in each country and time of transmission is not known and currently relies on epidemiological analysis, which may be unreliable owing to the difficulties of obtaining patient information. Here we trace the genetic evolution of EBOV in the current outbreak that has resulted in multiple lineages. Deep sequencing of 179 patient samples processed by the European Mobile Laboratory, the first diagnostics unit to be deployed to the epicentre of the outbreak in Guinea, reveals an epidemiological and evolutionary history of the epidemic from March 2014 to January 2015. Analysis of EBOV genome evolution has also benefited from a similar sequencing effort of patient samples from Sierra Leone. Our results confirm that the EBOV from Guinea moved into Sierra Leone, most likely in April or early May. The viruses of the Guinea/Sierra Leone lineage mixed around June/July 2014. Viral sequences covering August, September and October 2014 indicate that this lineage evolved independently within Guinea. These data can be used in conjunction with epidemiological information to test retrospectively the effectiveness of control measures, and provides an unprecedented window into the evolution of an ongoing viral haemorrhagic fever outbreak.


Subject(s)
Disease Outbreaks/statistics & numerical data , Ebolavirus/genetics , Evolution, Molecular , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Phylogeny , Spatio-Temporal Analysis , Amino Acid Substitution/genetics , Ebolavirus/isolation & purification , Female , Guinea/epidemiology , Hemorrhagic Fever, Ebola/transmission , High-Throughput Nucleotide Sequencing , Humans , Liberia/epidemiology , Male , Mali/epidemiology , Molecular Sequence Data , Sierra Leone/epidemiology
9.
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
10.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31413134

ABSTRACT

Lassa virus is genetically diverse with several lineages circulating in West Africa. This study aimed at describing the sequence variability of Lassa virus across Nigeria and inferring its spatiotemporal evolution. We sequenced and isolated 77 Lassa virus strains from 16 Nigerian states. The final data set, including previous works, comprised metadata and sequences of 219 unique strains sampled between 1969 and 2018 in 22 states. Most of this data originated from Lassa fever patients diagnosed at Irrua Specialist Teaching Hospital, Edo State, Nigeria. The majority of sequences clustered with the main Nigerian lineages II and III, while a few sequences formed a new cluster related to Lassa virus strains from Hylomyscus pamfi Within lineages II and III, seven and five sublineages, respectively, were distinguishable. Phylogeographic analysis suggests an origin of lineage II in the southeastern part of the country around Ebonyi State and a main vector of dispersal toward the west across the Niger River, through Anambra, Kogi, Delta, and Edo into Ondo State. The frontline of virus dispersal appears to be in Ondo. Minor vectors are directed northeast toward Taraba and Adamawa and south toward Imo and Rivers. Lineage III might have spread from northern Plateau State into Kaduna, Nasarawa, Federal Capital Territory, and Bauchi. One sublineage moved south and crossed the Benue River into Benue State. This study provides a geographic mapping of lineages and phylogenetic clusters in Nigeria at a higher resolution. In addition, we estimated the direction and time frame of virus dispersal in the country.IMPORTANCE Lassa virus is the causative agent of Lassa fever, a viral hemorrhagic fever with a case fatality rate of approximately 30% in Africa. Previous studies disclosed a geographical pattern in the distribution of Lassa virus strains and a westward movement of the virus across West Africa during evolution. Our study provides a deeper understanding of the geography of genetic lineages and sublineages of the virus in Nigeria. In addition, we modeled how the virus spread in the country. This knowledge allows us to predict into which geographical areas the virus might spread in the future and prioritize areas for Lassa fever surveillance. Our study not only aimed to generate Lassa virus sequences from across Nigeria but also to isolate and conserve the respective viruses for future research. Both isolates and sequences are important for the development and evaluation of medical countermeasures to treat and prevent Lassa fever, such as diagnostics, therapeutics, and vaccines.


Subject(s)
Lassa Fever/virology , Lassa virus/classification , Animals , Evolution, Molecular , Genetic Variation , Humans , Lassa Fever/epidemiology , Lassa Fever/transmission , Lassa virus/genetics , Murinae/virology , Nigeria/epidemiology , Phylogeny , Phylogeography
12.
Trop Med Int Health ; 20(11): 1424-1430, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26171669

ABSTRACT

OBJECTIVE: To describe our experiences in the management of a case of Lassa fever (LF) and follow-up of nosocomial primary contacts during the 2014 Ebola outbreak in West Africa. METHODS: Clinical management of the index case and infection control/surveillance activities for primary contacts are described. Laboratory confirmation was by Lassa virus-specific reverse-transcriptase PCR. RESULTS: A 28-year-old man with a 10-day history of febrile illness was referred to a major tertiary hospital in south-east Nigeria from a city that previously experienced a LF outbreak and was recently affected by Ebola. On observation of haemorrhagic features, clinicians were at a crossroads. Diagnosis of LF was confirmed at a National Reference Centre. The patient died despite initiation of ribavirin therapy. Response activities identified 121 primary contacts comprising 78 (64.5%) hospital staff/interns, 19 (15.7%) medical students, 18 (14.9%) inpatients and 6 (5.0%) relatives. Their mean age was 32.8 ± 6.6 years, and 65.3% were women. Twenty (16.5%) had high-risk exposure and were offered ribavirin as post-exposure prophylaxis. No secondary case of LF occurred. Fatigue (43.8%) and dizziness (31.3%) were the commonest side effects of ribavirin. CONCLUSIONS: Response activities contained nosocomial spread of LF, but challenges were experienced including lack of a purpose-built isolation facility, absence of local Lassa virus laboratory capacity, failure to use appropriate protective equipment and stigmatisation of contacts. A key lesson is that the weak health systems of Africa should be comprehensively strengthened; otherwise, we might win the Ebola battle but lose the one against less virulent infections for which effective treatment exists.

14.
Nat Microbiol ; 9(3): 751-762, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38326571

ABSTRACT

Infection with Lassa virus (LASV) can cause Lassa fever, a haemorrhagic illness with an estimated fatality rate of 29.7%, but causes no or mild symptoms in many individuals. Here, to investigate whether human genetic variation underlies the heterogeneity of LASV infection, we carried out genome-wide association studies (GWAS) as well as seroprevalence surveys, human leukocyte antigen typing and high-throughput variant functional characterization assays. We analysed Lassa fever susceptibility and fatal outcomes in 533 cases of Lassa fever and 1,986 population controls recruited over a 7 year period in Nigeria and Sierra Leone. We detected genome-wide significant variant associations with Lassa fever fatal outcomes near GRM7 and LIF in the Nigerian cohort. We also show that a haplotype bearing signatures of positive selection and overlapping LARGE1, a required LASV entry factor, is associated with decreased risk of Lassa fever in the Nigerian cohort but not in the Sierra Leone cohort. Overall, we identified variants and genes that may impact the risk of severe Lassa fever, demonstrating how GWAS can provide insight into viral pathogenesis.


Subject(s)
Lassa Fever , Humans , Lassa Fever/genetics , Lassa Fever/diagnosis , Lassa Fever/epidemiology , Genome-Wide Association Study , Seroepidemiologic Studies , Lassa virus/genetics , Fever , Human Genetics
15.
Emerg Microbes Infect ; 12(1): 2219350, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37288752

ABSTRACT

We phylogenetically compared sequences of the zoonotic Lassa virus (LASV) obtained from Mastomys rodents in seven localities across the highly endemic Edo and Ondo States within Nigeria. Sequencing 1641 nt from the S segment of the virus genome, we resolved clades within lineage II that were either limited to Ebudin and Okhuesan in Edo state (2g-beta) or along Owo-Okeluse-Ifon in Ondo state (2g-gamma). We also found clades within Ekpoma, a relatively large cosmopolitan town in Edo state, that extended into other localities within Edo (2g-alpha) and Ondo (2g-delta). LASV variants from M. natalensis within Ebudin and Ekpoma in Edo State (dated approximately 1961) were more ancient compared to those from Ondo state (approximately 1977), suggesting a broadly east-west virus migration across south-western Nigeria; a pattern not always consistent with LASV sequences derived from humans in the same localities. Additionally, in Ebudin and Ekpoma, LASV sequences between M. natalensis and M. erythroleucus were interspersed on the phylogenetic tree, but those from M. erythroleucus were estimated to emerge more recently (approximately 2005). Overall, our results show that LASV amplification in certain localities (reaching a prevalence as high as 76% in Okeluse), anthropogenically-aided spread of rodent-borne variants amidst the larger towns (involving communal accommodation such as student hostels), and virus-exchange between syntopic M. natalensis and M. erythroleucus rodents (as the latter, a savanna species, encroaches southward into the degraded forest) pose perpetual zoonotic hazard across the Edo-Ondo Lassa fever belt, threatening to accelerate the dissemination of the virus into non endemic areas.


Subject(s)
Lassa Fever , Lassa virus , Humans , Mice , Animals , Lassa virus/genetics , Nigeria/epidemiology , Phylogeny , Lassa Fever/epidemiology , Lassa Fever/veterinary , Murinae
16.
PLOS Glob Public Health ; 3(11): e0002159, 2023.
Article in English | MEDLINE | ID: mdl-37939051

ABSTRACT

Lassa fever (LF) is a potentially lethal viral haemorrhagic infection of humans caused by Lassa mammarenavirus (LASV). It is an important endemic zoonotic disease in West Africa with growing evidence for increasing frequency and sizes of outbreaks. Phylogeographic and molecular epidemiology methods have projected expansion of the Lassa fever endemic zone in the context of future global change. The Natal multimammate mouse (Mastomys natalensis) is the predominant LASV reservoir, with few studies investigating the role of other animal species. To explore host sequencing biases, all LASV nucleotide sequences and associated metadata available on GenBank (n = 2,298) were retrieved. Most data originated from Nigeria (54%), Guinea (20%) and Sierra Leone (14%). Data from non-human hosts (n = 703) were limited and only 69 sequences encompassed complete genes. We found a strong positive correlation between the number of confirmed human cases and sequences at the country level (r = 0.93 (95% Confidence Interval = 0.71-0.98), p < 0.001) but no correlation exists between confirmed cases and the number of available rodent sequences (r = -0.019 (95% C.I. -0.71-0.69), p = 0.96). Spatial modelling of sequencing effort highlighted current biases in locations of available sequences, with increased sequencing effort observed in Southern Guinea and Southern Nigeria. Phylogenetic analyses showed geographic clustering of LASV lineages, suggestive of isolated events of human-to-rodent transmission and the emergence of currently circulating strains of LASV from the year 1498 in Nigeria. Overall, the current study highlights significant geographic limitations in LASV surveillance, particularly, in non-human hosts. Further investigation of the non-human reservoir of LASV, alongside expanded surveillance, are required for precise characterisation of the emergence and dispersal of LASV. Accurate surveillance of LASV circulation in non-human hosts is vital to guide early detection and initiation of public health interventions for future Lassa fever outbreaks.

17.
J Infect ; 87(1): 27-33, 2023 07.
Article in English | MEDLINE | ID: mdl-37075910

ABSTRACT

BACKGROUND: Managing Lassa fever (LF) patients is challenging because of the complexity of this life-threatening infectious disease, the necessary isolation measures, and the limited resources in countries where it is endemic. Point-of-care ultrasonography (POCUS) is a promising low-cost imaging technique that may help in guiding the management of patients. METHODS: We conducted this observational study at the Irrua Specialist Teaching Hospital in Nigeria. We developed a POCUS protocol, trained local physicians who applied the protocol to LF patients and recorded and interpreted the clips. These were then independently re-evaluated by an external expert, and associations with clinical, laboratory and virological data were analyzed. FINDINGS: We developed the POCUS protocol based on existing literature and expert opinion and trained two clinicians, who then used POCUS to examine 46 patients. We observed at least one pathological finding in 29 (63%) patients. Ascites was found in 14 (30%), pericardial effusion in 10 (22%), pleural effusion in 5 (11%), and polyserositis in 7 (15%) patients, respectively. Eight patients (17%) showed hyperechoic kidneys. Seven patients succumbed to the disease while 39 patients survived, resulting in a fatality rate of 15%. Pleural effusions and hyper-echoic kidneys were associated with increased mortality. INTERPRETATION: In acute LF, a newly established POCUS protocol readily identified a high prevalence of clinically relevant pathological findings. The assessment by POCUS required minimal resources and training; the detected pathologies such as pleural effusions and kidney injury may help to guide the clinical management of the most at-risk LF patients.


Subject(s)
Lassa Fever , Physicians , Pleural Effusion , Humans , Lassa Fever/diagnostic imaging , Point-of-Care Systems , Ultrasonography/methods
18.
PLoS One ; 18(3): e0283643, 2023.
Article in English | MEDLINE | ID: mdl-36996258

ABSTRACT

BACKGROUND: Lassa fever (LF), a haemorrhagic illness caused by the Lassa fever virus (LASV), is endemic in West Africa and causes 5000 fatalities every year. The true prevalence and incidence rates of LF are unknown as infections are often asymptomatic, clinical presentations are varied, and surveillance systems are not robust. The aim of the Enable Lassa research programme is to estimate the incidences of LASV infection and LF disease in five West African countries. The core protocol described here harmonises key study components, such as eligibility criteria, case definitions, outcome measures, and laboratory tests, which will maximise the comparability of data for between-country analyses. METHOD: We are conducting a prospective cohort study in Benin, Guinea, Liberia, Nigeria (three sites), and Sierra Leone from 2020 to 2023, with 24 months of follow-up. Each site will assess the incidence of LASV infection, LF disease, or both. When both incidences are assessed the LASV cohort (nmin = 1000 per site) will be drawn from the LF cohort (nmin = 5000 per site). During recruitment participants will complete questionnaires on household composition, socioeconomic status, demographic characteristics, and LF history, and blood samples will be collected to determine IgG LASV serostatus. LF disease cohort participants will be contacted biweekly to identify acute febrile cases, from whom blood samples will be drawn to test for active LASV infection using RT-PCR. Symptom and treatment data will be abstracted from medical records of LF cases. LF survivors will be followed up after four months to assess sequelae, specifically sensorineural hearing loss. LASV infection cohort participants will be asked for a blood sample every six months to assess LASV serostatus (IgG and IgM). DISCUSSION: Data on LASV infection and LF disease incidence in West Africa from this research programme will determine the feasibility of future Phase IIb or III clinical trials for LF vaccine candidates.


Subject(s)
Lassa Fever , Humans , Cohort Studies , Immunoglobulin G , Incidence , Lassa Fever/epidemiology , Lassa Fever/diagnosis , Lassa virus , Liberia , Prospective Studies , Multicenter Studies as Topic
19.
Trop Med Int Health ; 17(8): 1001-4, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22594713

ABSTRACT

OBJECTIVES: To estimate the burden of Lassa fever in northern and central Edo, a state in south Nigeria where Lassa fever has been reported. METHODS: Blood samples were obtained from 60 patients hospitalised at the Irrua Specialist Teaching Hospital (ISTH), Irrua, with a clinical suspicion of Lassa fever and from 451 febrile outpatients seen at the ISTH and hospitals in Ekpoma, Iruekpen, Uromi, Auchi and Igarra. All samples were tested retrospectively by Lassa virus-specific RT-PCR. Outpatients were additionally screened for Lassa virus-specific antibodies by indirect immunofluorescent antibody assay. RESULTS: Lassa virus was detected in 25 of 60 (42%) patients with a clinical suspicion of Lassa fever. The disease affected persons of all age groups and with various occupations, including healthcare workers. The clinical picture was dominated by gastrointestinal symptoms. The case fatality rate was 29%. Lassa virus was detected in 2 of 451 (0.44%) febrile outpatients, and 8 (1.8%) were positive for Lassa virus-specific IgG. CONCLUSIONS: Lassa fever contributes to hospital mortality in Edo State. The low prevalence of the disease among outpatients and the low seroprevalence may indicate that the population-level incidence is not high. Surveillance for Lassa fever should focus on the hospitalised patient.


Subject(s)
Hospitals, Teaching/statistics & numerical data , Lassa Fever/epidemiology , Adolescent , Adult , Antibodies, Viral , Child , Child, Preschool , Female , Fluorescent Antibody Technique, Indirect , Humans , Incidence , Infant , Lassa Fever/genetics , Lassa Fever/mortality , Male , Middle Aged , Nigeria/epidemiology , Prevalence , RNA, Viral/analysis , Retrospective Studies , Socioeconomic Factors , Young Adult
20.
Int J Infect Dis ; 122: 107-111, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35640830

ABSTRACT

We explain research gaps on Monkeypox (MPX) virus epidemiology in endemic countries and present hypotheses for the recent increase of MPX cases in West Africa as a possible explanation for the current epidemic in Europe, America, and Australia. The detection of >400 MPX cases in less than a month in May 2022, across many countries underscores the epidemic potential of MPX in humans and demonstrates several important research gaps. First, the true burden of MPX in West and Central Africa is poorly understood, although it is critical for prevention and control of future outbreaks. Second, the diversity and extent of the animal reservoir remain unknown. We hypothesize that the synanthropic rodent population has increased in recent years in Africa leading to more human-rodent interactions and thus increased transmission of MPXV. We further hypothesise that nearly 45 years after the end of routine smallpox vaccination, the larger and more interconnected immune-naïve population has crossed a threshold resulting in more sustainable human-to-human transmission of MPXV. The current epidemic in the Western World is possibly a consequence of increased local transmission of MPXV in Africa. A new estimation of the basic and effective reproduction rate (R0 and Re) in different populations is required. National, regional, and international collaborations are needed to address research gaps related to MPX outbreaks.


Subject(s)
Mpox (monkeypox) , Africa, Central , Animals , Cost of Illness , Disease Outbreaks , Humans , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/prevention & control , Monkeypox virus , Rodentia
SELECTION OF CITATIONS
SEARCH DETAIL