Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 662
Filtrar
1.
Nat Commun ; 15(1): 8133, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39285186

RESUMEN

Limited knowledge exists on the quality of polyclonal antibody responses generated following Marburg virus (MARV) infection and its evolution in survivors. In this study, we evaluate MARV proteome-wide antibody repertoire longitudinally in convalescent phase approximately every six months for five years following MARV infection in ten human survivors. Differential kinetics were observed for IgM vs IgG vs IgA epitope diversity, antibody binding, antibody affinity maturation and Fc-receptor interaction to MARV proteins. Durability of MARV-neutralizing antibodies is low in survivors. MARV infection induces a diverse epitope repertoire with predominance against GP, VP40, VP30 and VP24 that persisted up to 5 years post-exposure. However, the IgM and IgA repertoire declines over time. Within MARV-GP, IgG recognize antigenic sites predominantly in the amino-terminus, wing domain and GP2-heptad repeat. Interestingly, MARV infection generates robust durable FcɣRI, FcɣRIIA and FcɣRIIIA IgG-Fc receptor interactions. Immunization with immunodominant MARV epitopes reveals conserved wing region between GP1 and GP2, induces neutralizing antibodies against MARV. These findings demonstrate that MARV infection generates a diverse, long-lasting, non-neutralizing, IgG antibody repertoire that perturbs disease by FcɣR activity. This information, along with discovery of neutralizing immunogen in wing domain, could aid in development of effective therapeutics and vaccines against Marburg virus.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Enfermedad del Virus de Marburg , Marburgvirus , Proteoma , Marburgvirus/inmunología , Humanos , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/virología , Anticuerpos Antivirales/inmunología , Anticuerpos Neutralizantes/inmunología , Proteoma/inmunología , Femenino , Vacunas Virales/inmunología , Inmunoglobulina G/inmunología , Masculino , Epítopos/inmunología , Adulto , Inmunoglobulina M/inmunología , Persona de Mediana Edad , Estudios Longitudinales , Inmunoglobulina A/inmunología , Desarrollo de Vacunas , Proteínas del Envoltorio Viral/inmunología
2.
PLoS One ; 19(9): e0309762, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39236024

RESUMEN

INTRODUCTION: In March 2023, a Marburg Virus Disease (MVD) outbreak was declared in Kagera region, Northwestern Tanzania. This was the first MVD outbreak in the country. We describe the epidemiological characteristics of MVD cases and contacts. METHODS: The Ministry of Health activated an outbreak response team. Outbreak investigation methods were applied to cases identified through MVD standard case definitions and confirmed through reverse-transcriptase polymerase chain reaction (RT PCR). All identified case contacts were added into the contact listing form and followed up in-person daily for any signs or symptoms for 21 days. Data collected from various forms was managed and analyzed using Excel and QGIS software for mapping. RESULTS: A total of nine MVD cases were reported with eight laboratory-confirmed and one probable. Two of the reported cases were frontline healthcare workers and seven were family related members. Cases were children and adults between 1-59 years of age with a median age of 34 years. Six were males. Six cases died equivalent to a case fatality rate (CFR) of 66.7%. A total of 212 individuals were identified as contacts and two (2) became cases. The outbreak was localized in two geo-administrative wards (Maruku and Kanyangereko) of Bukoba District Council. CONCLUSION: Transmission during this outbreak occurred among family members and healthcare workers who provided care to the cases. The delay in detection aggravated the spread and possibly the consequent fatality but once confirmed the swift response stemmed further transmission containing the disease at the epicenter wards. The outbreak lasted for 72 days but as the origin is still unknown, further research is required to explore the source of this outbreak.


Asunto(s)
Brotes de Enfermedades , Enfermedad del Virus de Marburg , Humanos , Tanzanía/epidemiología , Masculino , Femenino , Adulto , Persona de Mediana Edad , Enfermedad del Virus de Marburg/epidemiología , Enfermedad del Virus de Marburg/transmisión , Enfermedad del Virus de Marburg/virología , Niño , Adolescente , Lactante , Preescolar , Adulto Joven , Marburgvirus/genética , Marburgvirus/aislamiento & purificación , Animales
3.
Viruses ; 16(8)2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39205155

RESUMEN

Filoviruses, like the Marburg (MARV) and Ebola (EBOV) viruses, have caused outbreaks associated with significant hemorrhagic morbidity and high fatality rates. Vaccines offer one of the best countermeasures for fatal infection, but to date only the EBOV vaccine has received FDA licensure. Given the limited cross protection between the EBOV vaccine and Marburg hemorrhagic fever (MHF), we analyzed the protective efficacy of a similar vaccine, rVSV-MARV, in the lethal cynomolgus macaque model. NHPs vaccinated with a single dose (as little as 1.6 × 107 pfu) of rVSV-MARV seroconverted to MARV G-protein prior to challenge on day 42. Vaccinemia was measured in all vaccinated primates, self-resolved by day 14 post vaccination. Importantly, all vaccinated NHPs survived lethal MARV challenge, and showed no significant alterations in key markers of morbid disease, including clinical signs, and certain hematological and clinical chemistry parameters. Further, apart from one primate (from which tissues were not collected and no causal link was established), no pathology associated with Marburg disease was observed in vaccinated animals. Taken together, rVSV-MARV is a safe and efficacious vaccine against MHF in cynomolgus macaques.


Asunto(s)
Macaca fascicularis , Enfermedad del Virus de Marburg , Marburgvirus , Vesiculovirus , Vacunas Virales , Animales , Enfermedad del Virus de Marburg/prevención & control , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/virología , Marburgvirus/inmunología , Marburgvirus/genética , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Vesiculovirus/genética , Vesiculovirus/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/administración & dosificación , Modelos Animales de Enfermedad , Vacunación , Masculino , Femenino , Eficacia de las Vacunas , Vectores Genéticos , Inmunogenicidad Vacunal
4.
Int J Mol Sci ; 25(15)2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39126087

RESUMEN

Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 in Angola. From 2021 to 2023, MARV outbreaks occurred in Guinea, Ghana, New Guinea, and Tanzania, emphasizing the expansion of its endemic area into new geographical regions. There are currently no approved vaccines or therapeutics targeting MARV, but several vaccine candidates have shown promise in preclinical studies. We compared three vaccine platforms simultaneously by vaccinating hamsters with either a single dose of an adenovirus-based (ChAdOx-1 MARV) vaccine, an alphavirus replicon-based RNA (LION-MARV) vaccine, or a recombinant vesicular stomatitis virus-based (VSV-MARV) vaccine, all expressing the MARV glycoprotein as the antigen. Lethal challenge with hamster-adapted MARV 4 weeks after vaccination resulted in uniform protection of the VSV-MARV and LION-MARV groups and 83% of the ChAdOx-1 MARV group. Assessment of the antigen-specific humoral response and its functionality revealed vaccine-platform-dependent differences, particularly in the Fc effector functions.


Asunto(s)
Enfermedad del Virus de Marburg , Marburgvirus , Vacunas Virales , Animales , Cricetinae , Vacunas Virales/inmunología , Marburgvirus/inmunología , Enfermedad del Virus de Marburg/prevención & control , Enfermedad del Virus de Marburg/inmunología , Modelos Animales de Enfermedad , Adenoviridae/genética , Adenoviridae/inmunología , Vesiculovirus/inmunología , Vesiculovirus/genética , Anticuerpos Antivirales/inmunología , Vacunación/métodos
5.
J Virol ; 98(9): e0104724, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39194238

RESUMEN

Little is known regarding the molecular mechanisms that highly pathogenic Marburg virus (MARV) utilizes to transcribe and replicate its genome. Previous studies assumed that dephosphorylation of the filoviral transcription factor VP30 supports transcription, while phosphorylated VP30 reduces transcription. Here, we focused on the role of the host protein phosphatase 2A (PP2A) for VP30 dephosphorylation and promotion of viral transcription. We could show that MARV NP interacts with the subunit B56 of PP2A, as previously shown for the Ebola virus, and that this interaction is important for MARV transcription activity. Inhibition of the interaction between PP2A and NP either by mutating the B56 binding motif encoded on NP, or the use of a PP2A inhibitor, induced VP30 hyperphosphorylation, and as a consequence a decrease of MARV transcription as well as viral growth. These results suggest that NP plays a key role in the dephosphorylation of VP30 by recruiting PP2A. Generation of recombinant (rec) MARV lacking the PP2A-B56 interaction motif on NP was not possible suggesting an essential role of PP2A-mediated VP30 dephosphorylation for the MARV replication cycle. Likewise, we were not able to generate recMARV containing VP30 phosphomimetic mutants indicating that dynamic cycles of VP30 de- and rephosphorylation are a prerequisite for an efficient viral life cycle. As the specific binding motifs of PP2A-B56 and VP30 within NP are highly conserved among the filoviral family, our data suggest a conserved mechanism for filovirus VP30 dephosphorylation by PP2A, revealing the host factor PP2A as a promising target for pan-filoviral therapies. IMPORTANCE: Our study elucidates the crucial role of host protein phosphatase 2A (PP2A) in Marburg virus (MARV) transcription. The regulatory subunit B56 of PP2A facilitates VP30 dephosphorylation, and hence transcription activation, via binding to NP. Our results, together with previous data, reveal a conserved mechanism of filovirus VP30 dephosphorylation by host factor PP2A at the NP interface and provide novel insights into potential pan-filovirus therapies.


Asunto(s)
Marburgvirus , Proteína Fosfatasa 2 , Transcripción Genética , Marburgvirus/fisiología , Marburgvirus/genética , Marburgvirus/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/genética , Humanos , Fosforilación , Replicación Viral , Células HEK293 , Animales , Interacciones Huésped-Patógeno , Proteínas Virales/metabolismo , Proteínas Virales/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Enfermedad del Virus de Marburg/virología , Enfermedad del Virus de Marburg/metabolismo , Unión Proteica , Línea Celular
6.
Viruses ; 16(8)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39205171

RESUMEN

Egyptian rousette bats (ERBs) are implicated as reservoir hosts for Marburg virus (MARV), but natural mechanisms involved in maintenance of MARV in ERB populations remain undefined. A number of hematophagous ectoparasites, including fleas, parasitize bats. Subcutaneous (SC) inoculation of ERBs with MARV consistently results in viremia, suggesting that infectious MARV could be ingested by blood-sucking ectoparasites during feeding. In our study, MARV RNA was detected in fleas that took a blood meal during feeding on viremic bats on days 3, 7, and 11 after SC inoculation. Virus concentration in individual ectoparasites was consistent with detectable levels of viremia in the blood of infected host bats. There was neither seroconversion nor viremia in control bats kept in close contact with MARV-infected bats infested with fleas for up to 40 days post-exposure. In fleas inoculated intracoelomically, MARV was detected up to 14 days after intracoelomic (IC) inoculation, but the virus concentration was lower than that delivered in the inoculum. All bats that had been infested with inoculated, viremic fleas remained virologically and serologically negative up to 38 days after infestation. Of 493 fleas collected from a wild ERB colony in Matlapitsi Cave, South Africa, where the enzootic transmission of MARV occurs, all tested negative for MARV RNA. While our findings seem to demonstrate that bat fleas lack vectorial capacity to transmit MARV biologically, their role in mechanical transmission should not be discounted. Regular blood-feeds, intra- and interhost mobility, direct feeding on blood vessels resulting in venous damage, and roosting behaviour of ERBs provide a potential physical bridge for MARV dissemination in densely populated cave-dwelling bats by fleas. The virus transfer might take place through inoculation of skin, mucosal membranes, and wounds when contaminated fleas are squashed during auto- and allogrooming, eating, biting, or fighting.


Asunto(s)
Quirópteros , Enfermedad del Virus de Marburg , Marburgvirus , Siphonaptera , Animales , Quirópteros/virología , Marburgvirus/genética , Marburgvirus/fisiología , Siphonaptera/virología , Enfermedad del Virus de Marburg/virología , Enfermedad del Virus de Marburg/transmisión , Reservorios de Enfermedades/virología , Viremia , Infestaciones por Pulgas/veterinaria , Infestaciones por Pulgas/transmisión , Infestaciones por Pulgas/virología , ARN Viral/genética , Egipto
7.
Health Secur ; 22(S1): S45-S49, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39037030

RESUMEN

Developing and sustaining relationships and networks before an emergency occurs is crucial. The Biocontainment Unit Leadership Workgroup is a consortium of the 13 Regional Emerging Special Pathogen Treatment Centers in the United States. Established in 2017, the volunteer-based workgroup is composed of operational leaders dedicated to maintaining readiness for special pathogen care. Monthly meetings focus on addressing operational challenges, sharing best practices, and brainstorming solutions to common problems. Task forces are leveraged to tackle more complex issues that are identified as priorities. In 2022, members of the workgroup were harnessed for response efforts related to mpox, Sudan ebolavirus, and Marburg virus disease. The weekly Outbreak Readiness call is a shared effort between the Biocontainment Unit Leadership Workgroup and the Special Pathogens Research Network of the National Emerging Special Pathogens Training and Education Center. Call participants included leaders of the Regional Emerging Special Pathogen Treatment Centers and federal partners who shared weekly updates on operational readiness of units, case counts, laboratory capacity, available medical countermeasures, and other pertinent information. The routine exchange of real-time information enabled learning and collegial sharing of experiences, highlighted the experience of the network to federal partners, and provided situational awareness of special pathogen outbreaks across the country. The consortium enabled this rapid convening of partners to meet an urgent need for special pathogen response. The weekly Outbreak Readiness call is a communication model and scalable framework that serves both domestic preparedness efforts and international efforts should the need for a collaborative global response arise. In this case study, we describe the framework and experience of this partnership, along with the structure of rapid deployment for group convening.


Asunto(s)
Brotes de Enfermedades , Fiebre Hemorrágica Ebola , Liderazgo , Humanos , Brotes de Enfermedades/prevención & control , Fiebre Hemorrágica Ebola/prevención & control , Fiebre Hemorrágica Ebola/epidemiología , Estados Unidos , Contención de Riesgos Biológicos/métodos , Enfermedad del Virus de Marburg/prevención & control
8.
BMC Biotechnol ; 24(1): 45, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970027

RESUMEN

Marburg virus (MARV) is a highly contagious and virulent agent belonging to Filoviridae family. MARV causes severe hemorrhagic fever in humans and non-human primates. Owing to its highly virulent nature, preventive approaches are promising for its control. There is currently no approved drug or vaccine against MARV, and management mainly involves supportive care to treat symptoms and prevent complications. Our aim was to design a novel multi-epitope vaccine (MEV) against MARV using immunoinformatics studies. In this study, various proteins (VP35, VP40 and glycoprotein precursor) were used and potential epitopes were selected. CTL and HTL epitopes covered 79.44% and 70.55% of the global population, respectively. The designed MEV construct was stable and expressed in Escherichia coli (E. coli) host. The physicochemical properties were also acceptable. MARV MEV candidate could predict comprehensive immune responses such as those of humoral and cellular in silico. Additionally, efficient interaction to toll-like receptor 3 (TLR3) and its agonist (ß-defensin) was predicted. There is a need for validation of these results using further in vitro and in vivo studies.


Asunto(s)
Biología Computacional , Enfermedad del Virus de Marburg , Marburgvirus , Vacunas Virales , Marburgvirus/inmunología , Enfermedad del Virus de Marburg/prevención & control , Enfermedad del Virus de Marburg/inmunología , Vacunas Virales/inmunología , Biología Computacional/métodos , Animales , Humanos , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/genética , Epítopos/inmunología , Epítopos/genética , Epítopos/química , Escherichia coli/genética , Escherichia coli/metabolismo , Inmunoinformática
10.
J Virol ; 98(7): e0015524, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38832790

RESUMEN

Marburg virus infection in humans is associated with case fatality rates that can reach up to 90%, but to date, there are no approved vaccines or monoclonal antibody (mAb) countermeasures. Here, we immunized Rhesus macaques with multivalent combinations of filovirus glycoprotein (GP) antigens belonging to Marburg, Sudan, and Ebola viruses to generate monospecific and cross-reactive antibody responses against them. From the animal that developed the highest titers of Marburg virus GP-specific neutralizing antibodies, we sorted single memory B cells using a heterologous Ravn virus GP probe and cloned and characterized a panel of 34 mAbs belonging to 28 unique lineages. Antibody specificities were assessed by overlapping pepscan and binding competition analyses, revealing that roughly a third of the lineages mapped to the conserved receptor binding region, including potent neutralizing lineages that were confirmed by negative stain electron microscopy to target this region. Additional lineages targeted a protective region on GP2, while others were found to possess cross-filovirus reactivity. Our study advances the understanding of orthomarburgvirus glycoprotein antigenicity and furthers efforts to develop candidate antibody countermeasures against these lethal viruses. IMPORTANCE: Marburg viruses were the first filoviruses characterized to emerge in humans in 1967 and cause severe hemorrhagic fever with average case fatality rates of ~50%. Although mAb countermeasures have been approved for clinical use against the related Ebola viruses, there are currently no approved countermeasures against Marburg viruses. We successfully isolated a panel of orthomarburgvirus GP-specific mAbs from a macaque immunized with a multivalent combination of filovirus antigens. Our analyses revealed that roughly half of the antibodies in the panel mapped to regions on the glycoprotein shown to protect from infection, including the host cell receptor binding domain and a protective region on the membrane-anchoring subunit. Other antibodies in the panel exhibited broad filovirus GP recognition. Our study describes the discovery of a diverse panel of cross-reactive macaque antibodies targeting orthomarburgvirus and other filovirus GPs and provides candidate immunotherapeutics for further study and development.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Reacciones Cruzadas , Macaca mulatta , Enfermedad del Virus de Marburg , Marburgvirus , Animales , Marburgvirus/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Monoclonales/inmunología , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/prevención & control , Reacciones Cruzadas/inmunología , Glicoproteínas/inmunología , Proteínas del Envoltorio Viral/inmunología , Inmunización , Humanos , Ebolavirus/inmunología , Antígenos Virales/inmunología
11.
PLoS Pathog ; 20(6): e1012262, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38924060

RESUMEN

Viral haemorrhagic fevers (VHF) pose a significant threat to human health. In recent years, VHF outbreaks caused by Ebola, Marburg and Lassa viruses have caused substantial morbidity and mortality in West and Central Africa. In 2022, an Ebola disease outbreak in Uganda caused by Sudan virus resulted in 164 cases with 55 deaths. In 2023, a Marburg disease outbreak was confirmed in Equatorial Guinea and Tanzania resulting in over 49 confirmed or suspected cases; 41 of which were fatal. There are no clearly defined correlates of protection against these VHF, impeding targeted vaccine development. Any vaccine developed should therefore induce strong and preferably long-lasting humoral and cellular immunity against these viruses. Ideally this immunity should also cross-protect against viral variants, which are known to circulate in animal reservoirs and cause human disease. We have utilized two viral vectored vaccine platforms, an adenovirus (ChAdOx1) and Modified Vaccinia Ankara (MVA), to develop a multi-pathogen vaccine regime against three filoviruses (Ebola virus, Sudan virus, Marburg virus) and an arenavirus (Lassa virus). These platform technologies have consistently demonstrated the capability to induce robust cellular and humoral antigen-specific immunity in humans, most recently in the rollout of the licensed ChAdOx1-nCoV19/AZD1222. Here, we show that our multi-pathogen vaccines elicit strong cellular and humoral immunity, induce a diverse range of chemokines and cytokines, and most importantly, confers protection after lethal Ebola virus, Sudan virus and Marburg virus challenges in a small animal model.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Fiebre de Lassa , Virus Lassa , Enfermedad del Virus de Marburg , Marburgvirus , Animales , Ratones , Ebolavirus/inmunología , Virus Lassa/inmunología , Marburgvirus/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Fiebre Hemorrágica Ebola/inmunología , Fiebre de Lassa/inmunología , Fiebre de Lassa/prevención & control , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/prevención & control , Vacunas Virales/inmunología , Humanos , Vacunación , Femenino , Anticuerpos Antivirales/inmunología , Inmunogenicidad Vacunal , Vacunas contra el Virus del Ébola/inmunología
12.
BMC Infect Dis ; 24(1): 628, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38914946

RESUMEN

Marburg viral disease (MVD) is a highly infectious disease with a case fatality rate of up to 90%, particularly impacting resource-limited countries where implementing Infection Prevention and Control (IPC) measures is challenging. This paper shares the experience of how Tanzania has improved its capacity to prevent and control highly infectious diseases, and how this capacity was utilized during the outbreak of the MVD disease that occurred for the first time in the country in 2023.In 2016 and the subsequent years, Tanzania conducted self and external assessments that revealed limited IPC capacity in responding to highly infectious diseases. To address these gaps, initiatives were undertaken, including the enhancement of IPC readiness through the development and dissemination of guidelines, assessments of healthcare facilities, supportive supervision and mentorship, procurement of supplies, and the renovation or construction of environments to bolster IPC implementation.The official confirmation and declaration of MVD on March 21, 2023, came after five patients had already died of the disease. MVD primarily spreads through contact and presents with severe symptoms, which make patient care and prevention challenging, especially in resource-limited settings. However, with the use of a trained workforce; IPC rapid needs assessment was conducted, identifying specific gaps. Based on the results; mentorship programs were carried out, specific policies and guidelines were developed, security measures were enhanced, all burial activities in the area were supervised, and both patients and staff were monitored across all facilities. By the end of the outbreak response on June 1, 2023, a total of 212 contacts had been identified, with the addition of only three deaths. Invasive procedures like dialysis and Manual Vacuum Aspiration prevented some deaths in infected patients, procedures previously discouraged.In summary, this experience underscores the critical importance of strict adherence to IPC practices in controlling highly infectious diseases. Recommendations for low-income countries include motivating healthcare providers and improving working conditions to enhance commitment in challenging environments. This report offers valuable insights and practical interventions for preparing for and addressing highly infectious disease outbreaks through implementation of IPC measures.


Asunto(s)
Brotes de Enfermedades , Enfermedad del Virus de Marburg , Tanzanía/epidemiología , Humanos , Brotes de Enfermedades/prevención & control , Enfermedad del Virus de Marburg/epidemiología , Enfermedad del Virus de Marburg/prevención & control , Control de Infecciones/métodos , Animales , Países en Desarrollo
13.
Rev Med Suisse ; 20(872): 881-885, 2024 May 01.
Artículo en Francés | MEDLINE | ID: mdl-38693801

RESUMEN

Marburg virus disease (MVD) is a dreadful but exceptional disease. Formerly mainly identified in Uganda, Angola and the Democratic Republic of Congo, it has recently appeared in the Republic of Guinea, Ghana, Equatorial Guinea and Tanzania, adding West Africa to the affected regions. Humans become infected through exposure to bats Roussettus aegyptiacus or during unprotected care of infected people. Five cases are linked to travellers, the last one dates to 2008 and involved a visit to caves colonized by bats. At present, there is no specific treatment or vaccine. Despite its rarity, adventurous travelers should be aware of the risks of exposure and avoid entering places inhabited by bats.


La maladie à virus Marburg est une maladie redoutable mais exceptionnelle. Autrefois identifiée en Ouganda, Angola et République démocratique du Congo, elle a récemment fait son apparition en République de Guinée, au Ghana, en Guinée équatoriale et en Tanzanie, ajoutant l'Afrique de l'Ouest aux régions touchées. Les humains s'infectent lors d'une exposition avec les chauves-souris roussettes d'Égypte ou lors de la prise en charge sans protection de personnes infectées. Cinq cas sont liés à des voyageurs, le dernier remonte à 2008 et était associé à la visite de grottes colonisées par des roussettes d'Égypte. Actuellement, il n'existe aucun traitement spécifique ni vaccin. Malgré sa rareté, les voyageurs aventureux doivent être informés des risques d'exposition et éviter de pénétrer dans des lieux habités par des chauves-souris.


Asunto(s)
Marburgvirus , Viaje , Femenino , Humanos , Masculino , Enfermedad del Virus de Marburg/epidemiología , Enfermedad del Virus de Marburg/transmisión , Enfermedad del Virus de Marburg/virología , Marburgvirus/aislamiento & purificación , Zoonosis Virales/epidemiología , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Quirópteros/virología
14.
Public Health ; 230: 128-137, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38537496

RESUMEN

OBJECTIVES: Marburg virus, previously referred to as Marburg hemorrhagic fever, is a highly severe and frequently fatal illness that affects humans. This study aimed to develop and validate a French questionnaire to assess knowledge, attitude, and practice toward Marburg virus disease (FKAP-MVD). STUDY DESIGN: An anonymous online survey was used, which was distributed through various platforms and emails. Data were collected from Burkina Faso, Guinea, the Democratic Republic of Congo, and Senegal. METHODS: To conduct the study, an anonymous online survey was used, which was distributed through various platforms such as Facebook, Twitter, WhatsApp, and emails. The survey was uploaded onto a Google form to facilitate data collection. Data were collected from Burkina Faso, Guinea, the Democratic Republic of Congo, and Senegal. RESULTS: Of the total sample of 510 participants, 60.0% were male, their mean age was 28.41 ± 6.32 years, 38.0% were married, 86.6% resided in urban areas and 64.1% had a university education. The questionnaire had good internal consistency; Cronbach's alpha was 0.87. The correlation between knowledge and attitude was 0.002, the correlation between knowledge and practice was 0.204, and the correlation between practice and attitude was relatively weak and negative at -0.060. This indicates the divergent validity of the questionnaire. The KMO value of 0.91 indicates a high level of adequacy, suggesting that the data are suitable for factor analysis. The Bartlett test of Sphericity yielded an approximate χ2 value of 4016.890 with 300 degrees of freedom and a P-value of 0.0001. The confirmatory factor analysis revealed 25 questions in three domains. The normed chi-square value is 1.224. The goodness of Fit Index (GFI) is 0.902, the Comparative Fit Index (CFI) is 0.982, the Root Mean Square Error of Approximation (RMSEA) is 0.033, and the Root Mean Square Residual (RMR) is 0.062. These values indicate a good fit of the model to the data. CONCLUSIONS: In general, the developed questionnaire has significant potential to inform public health initiatives and interventions related to MVD.


Asunto(s)
Enfermedad del Virus de Marburg , Animales , Humanos , Masculino , Adulto Joven , Adulto , Femenino , Conocimientos, Actitudes y Práctica en Salud , Salud Pública , Encuestas y Cuestionarios , Reproducibilidad de los Resultados , África del Sur del Sahara , Psicometría
15.
Nat Commun ; 15(1): 1826, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38418477

RESUMEN

Bats are increasingly recognized as reservoirs of emerging zoonotic pathogens. Egyptian rousette bats (ERBs) are the known reservoir of Marburg virus (MARV), a filovirus that causes deadly Marburg virus disease (MVD) in humans. However, ERBs harbor MARV asymptomatically, likely due to a coadapted and specific host immunity-pathogen relationship. Recently, we measured transcriptional responses in MARV-infected ERB whole tissues, showing that these bats possess a disease tolerant strategy that limits pro-inflammatory gene induction, presumably averting MVD-linked immunopathology. However, the host resistant strategy by which ERBs actively limit MARV burden remains elusive, which we hypothesize requires localized inflammatory responses unresolvable at bulk-tissue scale. Here, we use dexamethasone to attenuate ERB pro-inflammatory responses and assess MARV replication, shedding and disease. We show that MARV-infected ERBs naturally mount coordinated pro-inflammatory responses at liver foci of infection, comprised of recruited mononuclear phagocytes and T cells, the latter of which proliferate with likely MARV-specificity. When pro-inflammatory responses are diminished, ERBs display heightened MARV replication, oral/rectal shedding and severe MVD-like liver pathology, demonstrating that ERBs balance immunoprotective tolerance with discreet MARV-resistant pro-inflammatory responses. These data further suggest that natural ERB immunomodulatory stressors like food scarcity and habitat disruption may potentiate viral shedding, transmission and therefore outbreak risk.


Asunto(s)
Quirópteros , Filoviridae , Enfermedad del Virus de Marburg , Marburgvirus , Animales , Humanos , Marburgvirus/genética , Inmunidad
17.
AMA J Ethics ; 26(2): E109-115, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38306200

RESUMEN

Marburg virus, the first filovirus discovered and a close cousin to the Ebola virus, is carried by the Egyptian rousette bat, a common cave-dwelling fruit bat endemic to sub-Saharan Africa whose populations can exceed 50 000 individuals. Community outbreaks of Marburg virus can result in high morbidity rates. In eastern Africa, favorite habitats of these bats include rural subterranean gold mines-sometimes worked illegally-that create environments conducive to zoonotic virus transmission. This commentary on a case describes how outbreaks of Marburg virus disease among people exposed to sub-Saharan African caves and mines containing these bats cause tensions among miners, companies, public health officials, and conservationists.


Asunto(s)
Quirópteros , Enfermedad del Virus de Marburg , Marburgvirus , Animales , Humanos , Salud Pública , Enfermedad del Virus de Marburg/epidemiología , Brotes de Enfermedades
18.
Mar Drugs ; 22(1)2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38248659

RESUMEN

The Marburg virus (MBV), a deadly pathogen, poses a serious threat to world health due to the lack of effective treatments, calling for an immediate search for targeted and efficient treatments. In this study, we focused on compounds originating from marine fungi in order to identify possible inhibitory compounds against the Marburg virus (MBV) VP35-RNA binding domain (VP35-RBD) using a computational approach. We started with a virtual screening procedure using the Lipinski filter as a guide. Based on their docking scores, 42 potential candidates were found. Four of these compounds-CMNPD17596, CMNPD22144, CMNPD25994, and CMNPD17598-as well as myricetin, the control compound, were chosen for re-docking analysis. Re-docking revealed that these particular compounds had a higher affinity for MBV VP35-RBD in comparison to the control. Analyzing the chemical interactions revealed unique binding properties for every compound, identified by a range of Pi-cation interactions and hydrogen bond types. We were able to learn more about the dynamic behaviors and stability of the protein-ligand complexes through a 200-nanosecond molecular dynamics simulation, as demonstrated by the compounds' consistent RMSD and RMSF values. The multidimensional nature of the data was clarified by the application of principal component analysis, which suggested stable conformations in the complexes with little modification. Further insight into the energy profiles and stability states of these complexes was also obtained by an examination of the free energy landscape. Our findings underscore the effectiveness of computational strategies in identifying and analyzing potential inhibitors for MBV VP35-RBD, offering promising paths for further experimental investigations and possible therapeutic development against the MBV.


Asunto(s)
Enfermedad del Virus de Marburg , Animales , Motivos de Unión al ARN , Hongos , Enlace de Hidrógeno , Simulación de Dinámica Molecular
19.
Ann Glob Health ; 90(1): 5, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38273871

RESUMEN

The co-existence of deadly viral pandemics can be considered a nightmare for public health authorities. The surge of a Marburg virus disease (MVD) outbreak in Africa at a time when the coronavirus-19 (COVID-19) pandemic is partially controlled with its limited resources is an urgent call for concern. Over the past decades, several bouts of MVD outbreaks have occurred in Africa with an alarming case fatality rate. Despite this, little has been done to end its recurrence, and affected countries essentially depend on preventative rather than curative measures of management. The recent outbreak of MVD declared by the health officials of Equatorial Guinea, causing several deaths in the context of the COVID-19 pandemic, signals the need for speed in the establishment and the implementation of appropriate health policies and health system strategies to contain, destroy, and prevent the spread of this deadly virus to other neighboring countries.


Asunto(s)
Infecciones por Coronavirus , Enfermedad del Virus de Marburg , Marburgvirus , Animales , Humanos , Guinea Ecuatorial , Pandemias/prevención & control , Brotes de Enfermedades/prevención & control , Enfermedad del Virus de Marburg/epidemiología , Enfermedad del Virus de Marburg/prevención & control , Infecciones por Coronavirus/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA