Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.242
Filtrar
1.
Biophys Chem ; 314: 107318, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39226875

RESUMO

The Ebola delta peptide is an amphipathic, 40-residue peptide encoded by the Ebola virus, referred to as E40. The membrane-permeabilising activity of the E40 delta peptide has been demonstrated in cells and lipid vesicles suggesting the E40 delta peptide likely acts as a viroporin. The lytic activity of the peptide increases in the presence of anionic lipids and a disulphide bond in the C-terminal part of the peptide. Previous in silico work predicts the peptide to show a partially helical structure, but there is no experimental information on the structure of E40. Here, we use circular dichroism spectroscopy to report the secondary structure propensities of the reduced and oxidised forms of the E40 peptide in water, detergent micelles, and lipid vesicles composed of neutral and anionic lipids (POPC and POPG, respectively). Results indicate that the peptide is predominately a random coil in solution, and the disulphide bond has a small but measurable effect on peptide conformation. Secondary structure analysis shows large uncertainties and dependence on the reference data set and, in our system, cannot be used to accurately determine the secondary structure motifs of the peptide in membrane environments. Nevertheless, the spectra can be used to assess the relative changes in secondary structure propensities of the peptide depending on the solvent environment and disulphide bond. In POPC-POPG vesicles, the peptide transitions from a random coil towards a more structured conformation, which is even more pronounced in negatively charged SDS micelles. In vesicles, the effect depends on the peptide-lipid ratio, likely resulting from vesicle surface saturation. Further experiments with zwitterionic POPC vesicles and DPC micelles show that both curvature and negatively charged lipids can induce a change in conformation, with the two effects being cumulative. Electrostatic screening from Na+ ions reduced this effect. The oxidised form of the peptide shows a slightly lower propensity for secondary structure and retains a more random coil conformation even in the presence of PG-PC vesicles.


Assuntos
Dicroísmo Circular , Ebolavirus , Micelas , Estrutura Secundária de Proteína , Ebolavirus/química , Fosfatidilcolinas/química , Soluções , Fosfatidilgliceróis/química , Peptídeos/química , Água/química , Proteínas Virais/química , Sequência de Aminoácidos
2.
Lancet Glob Health ; 12(10): e1684-e1692, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39222652

RESUMO

BACKGROUND: Uganda has had seven Ebola disease outbreaks, between 2000 and 2022. On Sept 20, 2022, the Ministry of Health declared a Sudan virus disease outbreak in Mubende District, Central Uganda. We describe the epidemiological characteristics and transmission dynamics. METHODS: For this descriptive study, cases were classified as suspected, probable, or confirmed using Ministry of Health case definitions. We investigated all reported cases to obtain data on case-patient demographics, exposures, and signs and symptoms, and identified transmission chains. We conducted a descriptive epidemiological study and also calculated basic reproduction number (Ro) estimates. FINDINGS: Between Aug 8 and Nov 27, 2022, 164 cases (142 confirmed, 22 probable) were identified from nine (6%) of 146 districts. The median age was 29 years (IQR 20-38), 95 (58%) of 164 patients were male, and 77 (47%) patients died. Symptom onsets ranged from Aug 8 to Nov 27, 2022. The case fatality rate was highest in children younger than 10 years (17 [74%] of 23 patients). Fever (135 [84%] of 160 patients), vomiting (93 [58%] patients), weakness (89 [56%] patients), and diarrhoea (81 [51%] patients) were the most common symptoms; bleeding was uncommon (21 [13%] patients). Before outbreak identification, most case-patients (26 [60%] of 43 patients) sought care at private health facilities. The median incubation was 6 days (IQR 5-8), and median time from onset to death was 10 days (7-23). Most early cases represented health-care-associated transmission (43 [26%] of 164 patients); most later cases represented household transmission (109 [66%]). Overall Ro was 1·25. INTERPRETATION: Despite delayed detection, the 2022 Sudan virus disease outbreak was rapidly controlled, possibly thanks to a low Ro. Children (aged <10 years) were at the highest risk of death, highlighting the need for targeted interventions to improve their outcomes during Ebola disease outbreaks. Initial care-seeking occurred at facilities outside the government system, showing a need to ensure that private and public facilities receive training to identify possible Ebola disease cases during an outbreak. Health-care-associated transmission in private health facilities drove the early outbreak, suggesting gaps in infection prevention and control. FUNDING: None.


Assuntos
Surtos de Doenças , Doença pelo Vírus Ebola , Humanos , Uganda/epidemiologia , Doença pelo Vírus Ebola/epidemiologia , Masculino , Feminino , Adulto , Criança , Adulto Jovem , Sudão/epidemiologia , Adolescente , Pré-Escolar , Ebolavirus , Pessoa de Meia-Idade , Lactente , Estudos Epidemiológicos
3.
J Med Chem ; 67(18): 16381-16402, 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39248591

RESUMO

The endemic nature of the Ebola virus disease in Africa underscores the need for prophylactic and therapeutic drugs that are affordable and easy to administer. Through a phenotypic screening employing viral pseudotypes and our in-house chemical library, we identified a promising hit featuring a thiophene scaffold, exhibiting antiviral activity in the micromolar range. Following up on this thiophene hit, a new series of compounds that retain the five-membered heterocyclic scaffold while modifying several substituents was synthesized. Initial screening using a pseudotype viral system and validation assays employing authentic Ebola virus demonstrated the potential of this new chemical class as viral entry inhibitors. Subsequent investigations elucidated the mechanism of action through site-directed mutagenesis. Furthermore, we conducted studies to assess the pharmacokinetic profile of selected compounds to confirm its pharmacological and therapeutic potential.


Assuntos
Antivirais , Barreira Hematoencefálica , Ebolavirus , Tiofenos , Internalização do Vírus , Tiofenos/química , Tiofenos/farmacocinética , Tiofenos/farmacologia , Tiofenos/síntese química , Ebolavirus/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Humanos , Antivirais/farmacologia , Antivirais/química , Antivirais/farmacocinética , Antivirais/síntese química , Internalização do Vírus/efeitos dos fármacos , Relação Estrutura-Atividade , Animais , Descoberta de Drogas , Administração Oral , Disponibilidade Biológica , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/virologia
4.
Lancet Glob Health ; 12(10): e1730-e1736, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39270687

RESUMO

Ebola virus disease kills more than half of people infected. Since the disease is transmitted via close human contact, identifying individuals at the highest risk of developing the disease is possible on the basis of the type of contact (correlated with viral exposure). Different candidates for post-exposure prophylaxis (PEP; ie, vaccines, antivirals, and monoclonal antibodies) each have their specific benefits and limitations, which we discuss in this Viewpoint. Approved monoclonal antibodies have been found to reduce mortality in people with Ebola virus disease. As monoclonal antibodies act swiftly by directly targeting the virus, they are promising candidates for targeted PEP in contacts at high risk of developing disease. This intervention could save lives, halt viral transmission, and, ultimately, help curtail outbreak propagation. We explore how a strategic integration of monoclonal antibodies and vaccines as PEP could provide both immediate and long-term protection against Ebola virus disease, highlighting ongoing clinical research that aims to refine this approach, and discuss the transformative potential of a successful PEP strategy to help control viral haemorrhagic fever outbreaks.


Assuntos
Surtos de Doenças , Vacinas contra Ebola , Doença pelo Vírus Ebola , Profilaxia Pós-Exposição , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/epidemiologia , Humanos , Profilaxia Pós-Exposição/métodos , Surtos de Doenças/prevenção & controle , Vacinas contra Ebola/uso terapêutico , Vacinas contra Ebola/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Antivirais/uso terapêutico , Ebolavirus
5.
F1000Res ; 13: 642, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39290845

RESUMO

Background: Traditionally in the Democratic Republic of the Congo (DRC), centralised Ebola treatment centres (ETCs) have been set exclusively for Ebola virus disease (EVD) case management during outbreaks. During the 2020 EVD outbreak in DRC's Equateur Province, existing health centres were equipped as decentralised treatment centres (DTC) to improve access for patients with suspected EVD. Between ETCs and DTCs, we compared the time from symptom onset to admission and diagnosis among patients with suspected EVD. Methods: This was a cohort study based on analysis of a line-list containing demographic and clinical information of patients with suspected EVD admitted to any EVD health facility during the outbreak. Results: Of 2359 patients with suspected EVD, 363 (15%) were first admitted to a DTC. Of 1996 EVD-suspected patients initially admitted to an ETC, 72 (4%) were confirmed as EVD-positive. Of 363 EVD-suspected patients initially admitted to a DTC, 6 (2%) were confirmed and managed as EVD-positive in the DTC. Among all EVD-suspected patients, the median (interquartile range) duration between symptom onset and admission was 2 (1-4) days in a DTC compared to 4 (2-7) days in an ETC (p<0.001). Similarly, time from symptom onset to admission was significantly shorter among EVD-suspected patients ultimately diagnosed as EVD-negative. Conclusions: Since <5% of the EVD-suspected patients admitted were eventually diagnosed with EVD, there is a need for better screening to optimise resource utilization and outbreak control. Only one in seven EVD-suspected patients were admitted to a DTC first, as the DTCs were piloted in a limited and phased manner. However, there is a case to be made for considering decentralized care especially in remote and hard-to-reach areas in places like the DRC to facilitate early access to care, contain viral shedding by patients with EVD and ensure no disrupted provision of non-EVD services.


Assuntos
Surtos de Doenças , Doença pelo Vírus Ebola , Humanos , República Democrática do Congo/epidemiologia , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/terapia , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Adolescente , Adulto Jovem , Estudos de Coortes , Criança , Atenção à Saúde/organização & administração , Ebolavirus
6.
Nat Commun ; 15(1): 7666, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227399

RESUMO

Recent Ebola outbreaks underscore the importance of continuous prevention and disease control efforts. Authorized vaccines include Merck's Ervebo (rVSV-ZEBOV) and Johnson & Johnson's two-dose combination (Ad26.ZEBOV/MVA-BN-Filo). Here, in a five-year follow-up of the PREVAC randomized trial (NCT02876328), we report the results of the immunology ancillary study of the trial. The primary endpoint is to evaluate long-term memory T-cell responses induced by three vaccine regimens: Ad26-MVA, rVSV, and rVSV-booster. Polyfunctional EBOV-specific CD4+ T-cell responses increase after Ad26 priming and are further boosted by MVA, whereas minimal responses are observed in the rVSV groups, declining after one year. In-vitro expansion for eight days show sustained EBOV-specific T-cell responses for up to 60 months post-prime vaccination with both Ad26-MVA and rVSV, with no decline. Cytokine production analysis identify shared biomarkers between the Ad26-MVA and rVSV groups. In secondary endpoint, we observed an elevation of pro-inflammatory cytokines at Day 7 in the rVSV group. Finally, we establish a correlation between EBOV-specific T-cell responses and anti-EBOV IgG responses. Our findings can guide booster vaccination recommendations and help identify populations likely to benefit from revaccination.


Assuntos
Linfócitos T CD4-Positivos , Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Imunidade Celular , Humanos , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/imunologia , Vacinas contra Ebola/imunologia , Vacinas contra Ebola/administração & dosagem , Ebolavirus/imunologia , Linfócitos T CD4-Positivos/imunologia , Feminino , Masculino , Adulto , Anticorpos Antivirais/imunologia , Vacinação , Citocinas/metabolismo , Citocinas/imunologia , Seguimentos , Pessoa de Meia-Idade , Células T de Memória/imunologia , Imunização Secundária , Adulto Jovem , Memória Imunológica/imunologia
7.
J Infect ; 89(4): 106237, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39121969

RESUMO

BACKGROUND: Unlike adults, children experienced stronger and longer vector replication in plasma and shedding in saliva following rVSVΔG-ZEBOV-GP vaccination. The resulting risks of immunosuppression or immune hyperactivation leading to increased Adverse Events (AEs) and altered antibody responses are concerns that have been addressed in the present manuscript. METHODS: Children aged 1-12 years living in Gabon received either rVSVΔG-ZEBOV-GP (ERVEBO®) vaccine or the varicella-zoster virus (VZV) vaccine (VZV). The concentration of rVSVΔG vector in blood and saliva, the occurrence of AEs up to day 28; the anti-rVSVΔG-ZEBOV-GP and anti-VZV IgG antibody titres, neutralising and avidity functions of anti-rVSVΔG-ZEBOV-GP by day 365; were assessed in serum. (PACTR202005733552021) FINDINGS: In the rVSVΔG-ZEBOV-GP group, 70% and 7% of children had >0 copies/ml of rVSVΔG respectively in plasma by day 3 and in saliva by day 14 after vaccination, with no detection on day 28. Significantly higher but transient AEs occurred in the rVSVΔG-ZEBOV-GP group. Both vaccines induced seroconversion on day 28 and sustainable IgG antibody titres by day 365. Avidity and neutralisation functions of the anti-rVSVΔG-ZEBOV-GP antibodies peaked at day 28 and were maintained by day 365. INTERPRETATION: The replication and shedding do not affect the favourable risk-benefit balance of the rVSVΔG-ZEBOV-GP in children.


Assuntos
Anticorpos Antivirais , Vacinas contra Ebola , Humanos , Gabão , Pré-Escolar , Anticorpos Antivirais/sangue , Masculino , Feminino , Criança , Lactente , Vacinas contra Ebola/imunologia , Vacinas contra Ebola/efeitos adversos , Vacinas contra Ebola/administração & dosagem , Saliva/imunologia , Saliva/virologia , Ebolavirus/imunologia , Ebolavirus/genética , Imunoglobulina G/sangue , Doença pelo Vírus Ebola/prevenção & controle , Replicação Viral , Imunogenicidade da Vacina , Anticorpos Neutralizantes/sangue , Vacinação , Eliminação de Partículas Virais
8.
J Virol ; 98(9): e0068524, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39162435

RESUMO

MIL77-3 is one component of antibody cocktail that is produced in our lab and represents an effective regimen for animals suffering from Zaire Ebolavirus (EBOV) infection. MIL77-3 is engineered to increase its affinity for the FcγRIIIa (CD16a) by deleting the fucose in the framework region. The potential effects of this modification on host immune responses, however, remain largely unknown. Herein, we demonstrated that MIL77-3 recognized secreted glycoproptein (sGP), produced by EBOV, and formed the immunocomplex to potently augment antibody-dependent cytotoxicity of human peripheral blood-derived natural killer cells (pNKs), including CD56dim and CD56bright subpopulations, in contrast to the counterparts (Mab114, rEBOV548, fucosylated MIL77-3). Intriguingly, this effect was not observed when NK92-CD16a cell line was utilized and restored by the addition of beads-coupled or membrane-anchored sGP in combination with MIL77-3. Furthermore, sGP bound to unrecognized receptors on T cells contaminated in pNKs rather than NK92-CD16a cells. Administration of beads-coupled sGP/MIL77-3 complex in mice elicited NK activation. Overall, this work reveals an immune-stimulating function of sGP/MIL77-3 complex by triggering cytotoxic activity of NK cells, highlighting the necessity to evaluate the potential impact of MIL77-3 on host immune reaction in clinical trials. IMPORTANCE: Zaire Ebolavirus (EBOV) is highly lethal and causes sporadic outbreaks. The passive administration of monoclonal antibodies (mAbs) represents a promising treatment regimen against EBOV. Mounting evidence has shown that the efficacy of a subset of therapeutic mAbs in vivo is intimately associated with its capacity to trigger NK activity, supporting glycomodification of Fc region of anti-EBOV mAbs as a putative strategy to enhance Fc-mediated immune effector function as well as protection in vivo. Our work here uncovers the potential harmful influence of this modification on host immune responses, especially for mAbs with cross-reactivity to secreted glycoproptein (sGP) (e.g., MIL77-3), and highlights it is necessary to evaluate the NK-stimulating activity of a fucosylated mAb engaged with sGP when a new candidate is developed.


Assuntos
Anticorpos Antivirais , Citotoxicidade Celular Dependente de Anticorpos , Ebolavirus , Doença pelo Vírus Ebola , Células Matadoras Naturais , Receptores de IgG , Células Matadoras Naturais/imunologia , Humanos , Animais , Ebolavirus/imunologia , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Camundongos , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Anticorpos Antivirais/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Fucose , Linhagem Celular
9.
Emerg Microbes Infect ; 13(1): 2392651, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39155772

RESUMO

Ebola disease is a lethal viral hemorrhagic fever caused by ebolaviruses within the Filoviridae family with mortality rates of up to 90%. Monoclonal antibody (mAb) based therapies have shown great potential for the treatment of EVD. However, the potential emerging ebolavirus isolates and the negative effect of decoy protein on the therapeutic efficacy of antibodies highlight the necessity of developing novel antibodies to counter the threat of Ebola. Here, 11 fully human mAbs were isolated from transgenic mice immunized with GP protein and recombinant vesicular stomatitis virus-bearing GP (rVSV-EBOV GP). These mAbs were divided into five groups according to their germline genes and exhibited differential binding activities and neutralization capabilities. In particular, mAbs 8G6, 2A4, and 5H4 were cross-reactive and bound at least three ebolavirus glycoproteins. mAb 4C1 not only exhibited neutralizing activity but no cross-reaction with sGP. mAb 7D8 exhibited the strongest neutralizing capacity. Further analysis on the critical residues for the bindings of 4C1 and 8G6 to GPs was conducted using antibodies complementarity-determining regions (CDRs) alanine scanning. It has been shown that light chain CDR3 played a crucial role in binding and neutralization and that any mutation in CDRs could not improve the binding of 4C1 to sGP. Importantly, mAbs 7D8, 8G6, and 4C1 provided complete protections against EBOV infection in a hamster lethal challenge model when administered 12 h post-infection. These results support mAbs 7D8, 8G6, and 4C1 as potent antibody candidates for further investigations and pave the way for further developments of therapies and vaccines.


Assuntos
Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Modelos Animais de Doenças , Ebolavirus , Doença pelo Vírus Ebola , Animais , Ebolavirus/imunologia , Ebolavirus/genética , Anticorpos Monoclonais/imunologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/virologia , Anticorpos Antivirais/imunologia , Cricetinae , Camundongos , Anticorpos Neutralizantes/imunologia , Humanos , Camundongos Transgênicos , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Reações Cruzadas
10.
Nat Commun ; 15(1): 6953, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39138205

RESUMO

Filovirus-host interactions play important roles in all stages of the virus lifecycle. Here, we identify LATS1/2 kinases and YAP, key components of the Hippo pathway, as critical regulators of EBOV transcription and egress. Specifically, we find that when YAP is phosphorylated by LATS1/2, it localizes to the cytoplasm (Hippo "ON") where it sequesters VP40 to prevent egress. In contrast, when the Hippo pathway is "OFF", unphosphorylated YAP translocates to the nucleus where it transcriptionally activates host genes and promotes viral egress. Our data reveal that LATS2 indirectly modulates filoviral VP40-mediated egress through phosphorylation of AMOTp130, a positive regulator of viral egress, but more surprisingly that LATS1/2 kinases directly modulate EBOV transcription by phosphorylating VP30, an essential regulator of viral transcription. In sum, our findings highlight the potential to exploit the Hippo pathway/filovirus axis for the development of host-oriented countermeasures targeting EBOV and related filoviruses.


Assuntos
Ebolavirus , Via de Sinalização Hippo , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Fatores de Transcrição , Transcrição Gênica , Liberação de Vírus , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Fosforilação , Ebolavirus/fisiologia , Ebolavirus/genética , Ebolavirus/metabolismo , Células HEK293 , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Sinalização YAP/metabolismo , Proteínas da Matriz Viral/metabolismo , Proteínas da Matriz Viral/genética , Doença pelo Vírus Ebola/virologia , Doença pelo Vírus Ebola/metabolismo , Interações Hospedeiro-Patógeno , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética
11.
PLoS Pathog ; 20(8): e1012444, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39173055

RESUMO

The Ebola virus (EBOV) has emerged as a significant global health concern, notably during the 2013-2016 outbreak in West Africa. Despite the clinical approval of two EBOV antibody drugs, there is an urgent need for more diverse and effective antiviral drugs, along with comprehensive understanding of viral-host interactions. In this study, we harnessed a biologically contained EBOVΔVP30-EGFP cell culture model which could recapitulate the entire viral life cycle, to conduct a genome-wide CRISPR/Cas9 screen. Through this, we identified PIK3C3 (phosphatidylinositide 3-kinase) and SLC39A9 (zinc transporter) as crucial host factors for EBOV infection. Genetic depletion of SLC39A9 and PIK3C3 lead to reduction of EBOV entry, but not impact viral genome replication, suggesting that SLC39A9 and PIK3C3 act as entry factors, facilitating viral entry into host cells. Moreover, PIK3C3 kinase activity is indispensable for the internalization of EBOV virions, presumably through the regulation of endocytic and autophagic membrane traffic, which has been previously recognized as essential for EBOV internalization. Notably, our study demonstrated that PIK3C3 kinase inhibitor could effectively block EBOV infection, underscoring PIK3C3 as a promising drug target. Furthermore, biochemical analysis showed that recombinant SLC39A9 protein could directly bind viral GP protein, which further promotes the interaction of viral GP protein with cellular receptor NPC1. These findings suggests that SLC39A9 plays dual roles in EBOV entry. Initially, it serves as an attachment factor during the early entry phase by engaging with the viral GP protein. Subsequently, SLC39A9 functions an adaptor protein, facilitating the interaction between virions and the NPC1 receptor during the late entry phase, prior to cathepsin cleavage on the viral GP. In summary, this study offers novel insights into virus-host interactions, contributing valuable information for the development of new therapies against EBOV infection.


Assuntos
Sistemas CRISPR-Cas , Ebolavirus , Doença pelo Vírus Ebola , Internalização do Vírus , Animais , Humanos , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Classe III de Fosfatidilinositol 3-Quinases/genética , Ebolavirus/genética , Ebolavirus/fisiologia , Ebolavirus/metabolismo , Células HEK293 , Doença pelo Vírus Ebola/virologia , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/genética , Replicação Viral
12.
J Med Chem ; 67(16): 13737-13764, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39169825

RESUMO

Since the largest and most fatal Ebola virus epidemic during 2014-2016, there have been several consecutive filoviral outbreaks in recent years, including those in 2021, 2022, and 2023. Ongoing outbreak prevalence and limited FDA-approved filoviral therapeutics emphasize the need for novel small molecule treatments. Here, we showcase the structure-activity relationship development of N-substituted pyrrole-based heterocycles and their potent, submicromolar entry inhibition against diverse filoviruses in a target-based pseudovirus assay. Inhibitor antiviral activity was validated using replication-competent Ebola, Sudan, and Marburg viruses. Mutational analysis was used to map the targeted region within the Ebola virus glycoprotein. Antiviral counter-screen and phospholipidosis assays were performed to demonstrate the reduced off-target activity of these filoviral entry inhibitors. Favorable antiviral potency, selectivity, and drug-like properties of the N-substituted pyrrole-based heterocycles support their potential as broad-spectrum antifiloviral treatments.


Assuntos
Antivirais , Ebolavirus , Pirróis , Internalização do Vírus , Pirróis/farmacologia , Pirróis/química , Pirróis/síntese química , Antivirais/farmacologia , Antivirais/química , Antivirais/síntese química , Humanos , Relação Estrutura-Atividade , Ebolavirus/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Compostos Heterocíclicos/química , Compostos Heterocíclicos/síntese química , Filoviridae/efeitos dos fármacos , Marburgvirus/efeitos dos fármacos
13.
Health Secur ; 22(S1): S86-S96, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39137059

RESUMO

This case study describes a feasibility assessment of a novel isolation care tent used in health facilities in Uganda during the 2022 Sudan ebolavirus outbreak. The Isolation System for Treatment and Agile Response to High-Risk Infections Model 1B (ISTARI 1B) is a single-occupancy, portable, negative-pressure isolation tent designed for the safe delivery of standard care to patients with a communicable disease, including Ebola disease (Sudan). At the request of the Uganda Ministry of Health, the Makerere University Infectious Diseases Institute and University of Nebraska Medical Center partnered to evaluate 7 health facilities across 4 districts in Uganda for infrastructure, case management, and infection prevention and control (IPC) capacity relevant to isolation care and ISTARI 1B use. A 3-day workshop was held with IPC leaders to provide familiarization and hands-on experience with the ISTARI 1B, delineate appropriate use scenarios in Ugandan healthcare settings, contextualize ISTARI 1B use in case management and IPC workflows, develop a framework for site assessment and implementation readiness, and consider ongoing monitoring, assessment, and intervention tools. Workshop participants performed a comprehensive site assessment and mock deployment of the ISTARI 1B. In this case study, we describe lessons learned from health facility assessments and workshop outcomes and offer recommendations to support successful ISTARI 1B implementation. Use scenarios and implementation strategies were identified across facility levels, including tools for site assessment, training, risk communication, and ongoing quality and safety monitoring.


Assuntos
Surtos de Doenças , Estudos de Viabilidade , Doença pelo Vírus Ebola , Uganda/epidemiologia , Humanos , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/epidemiologia , Surtos de Doenças/prevenção & controle , Isolamento de Pacientes/métodos , Ebolavirus , Sudão/epidemiologia , Instalações de Saúde , Controle de Infecções/métodos , Controle de Infecções/organização & administração
14.
Viruses ; 16(8)2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39205153

RESUMO

Filoviruses are negative-sense single-stranded RNA viruses often associated with severe and highly lethal hemorrhagic fever in humans and nonhuman primates, with case fatality rates as high as 90%. Of the known filoviruses, Ebola virus (EBOV), the prototype of the genus Orthoebolavirus, has been a major public health concern as it frequently causes outbreaks and was associated with an unprecedented outbreak in several Western African countries in 2013-2016, affecting 28,610 people, 11,308 of whom died. Thereafter, filovirus research mostly focused on EBOV, paying less attention to other equally deadly orthoebolaviruses (Sudan, Bundibugyo, and Taï Forest viruses) and orthomarburgviruses (Marburg and Ravn viruses). Some of these filoviruses have emerged in nonendemic areas, as exemplified by four Marburg disease outbreaks recorded in Guinea, Ghana, Tanzania, and Equatorial Guinea between 2021 and 2023. Similarly, the Sudan virus has reemerged in Uganda 10 years after the last recorded outbreak. Moreover, several novel bat-derived filoviruses have been discovered in the last 15 years (Lloviu virus, Bombali virus, Menglà virus, and Dehong virus), most of which are poorly characterized but may display a wide host range. These novel viruses have the potential to cause outbreaks in humans. Several gaps are yet to be addressed regarding known and emerging filoviruses. These gaps include the virus ecology and pathogenicity, mechanisms of zoonotic transmission, host range and susceptibility, and the development of specific medical countermeasures. In this review, we summarize the current knowledge on non-Ebola filoviruses (Bombali virus, Bundibugyo virus, Reston virus, Sudan virus, Tai Forest virus, Marburg virus, Ravn virus, Lloviu virus, Menglà virus, and Dehong virus) and suggest some strategies to accelerate specific countermeasure development.


Assuntos
Surtos de Doenças , Infecções por Filoviridae , Filoviridae , Saúde Global , Humanos , Animais , Filoviridae/patogenicidade , Infecções por Filoviridae/epidemiologia , Infecções por Filoviridae/virologia , Ebolavirus/fisiologia , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/virologia , Doença pelo Vírus Ebola/transmissão , Zoonoses/epidemiologia , Zoonoses/virologia
15.
Viruses ; 16(8)2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39205201

RESUMO

Bombali virus (BOMV) is a novel Orthoebolavirus that has been detected in free-tailed bats in Sierra Leone, Guinea, Kenya, and Mozambique. We screened our collection of 349 free-tailed bat lungs collected in Côte d'Ivoire and Tanzania for BOMV RNA and tested 228 bat blood samples for BOMV antibodies. We did not detect BOMV-specific antibodies but found BOMV RNA in a Mops condylurus bat from Tanzania, marking the first detection of an ebolavirus in this country. Our findings further expand the geographic range of BOMV and support M. condylurus' role as a natural BOMV host.


Assuntos
Quirópteros , Animais , Quirópteros/virologia , Tanzânia , Anticorpos Antivirais/sangue , Filogenia , RNA Viral/genética , Côte d'Ivoire , Ebolavirus/isolamento & purificação , Ebolavirus/genética , Ebolavirus/imunologia , Pulmão/virologia
16.
Viruses ; 16(7)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-39066227

RESUMO

Although next-generation sequencing (NGS) has been instrumental in determining the genomic sequences of emerging RNA viruses, de novo sequence determination often lacks sufficient coverage of the 5' and 3' ends of the viral genomes. Since the genome ends of RNA viruses contain the transcription and genome replication promoters that are essential for viral propagation, a lack of terminal sequence information hinders the efforts to study the replication and transcription mechanisms of emerging and re-emerging viruses. To circumvent this, we have developed a novel method termed ViBE-Seq (Viral Bona Fide End Sequencing) for the high-resolution sequencing of filoviral genome ends using a simple yet robust protocol with high fidelity. This technique allows for sequence determination of the 5' end of viral RNA genomes and mRNAs with as little as 50 ng of total RNA. Using the Ebola virus and Marburg virus as prototypes for highly pathogenic, re-emerging viruses, we show that ViBE-Seq is a reliable technique for rapid and accurate 5' end sequencing of filovirus RNA sourced from virions, infected cells, and tissue obtained from infected animals. We also show that ViBE-Seq can be used to determine whether distinct reverse transcriptases have terminal deoxynucleotidyl transferase activity. Overall, ViBE-Seq will facilitate the access to complete sequences of emerging viruses.


Assuntos
Ebolavirus , Filoviridae , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , RNA Viral , Análise de Sequência de RNA , RNA Viral/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Ebolavirus/genética , Análise de Sequência de RNA/métodos , Filoviridae/genética , Marburgvirus/genética , Humanos , Animais
17.
Viruses ; 16(7)2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-39066263

RESUMO

Favipiravir is a ribonucleoside analogue that has been explored as a therapeutic for the treatment of Ebola Virus Disease (EVD). Promising data from rodent models has informed nonhuman primate trials, as well as evaluation in patients during the 2013-2016 West African EVD outbreak of favipiravir treatment. However, mixed results from these studies hindered regulatory approval of favipiravir for the indication of EVD. This study examined the influence of route of administration, duration of treatment, and treatment schedule of favipiravir in immune competent mouse and guinea pig models using rodent-adapted Zaire ebolavirus (EBOV). A dose of 300 mg/kg/day of favipiravir with an 8-day treatment was found to be fully effective at preventing lethal EVD-like disease in BALB/c mice regardless of route of administration (oral, intraperitoneal, or subcutaneous) or whether it was provided as a once-daily dose or a twice-daily split dose. Preclinical data generated in guinea pigs demonstrates that an 8-day treatment of 300 mg/kg/day of favipiravir reduces mortality following EBOV challenge regardless of route of treatment or duration of treatments for 8, 11, or 15 days. This work supports the future translational development of favipiravir as an EVD therapeutic.


Assuntos
Amidas , Antivirais , Modelos Animais de Doenças , Ebolavirus , Doença pelo Vírus Ebola , Camundongos Endogâmicos BALB C , Pirazinas , Animais , Amidas/uso terapêutico , Amidas/administração & dosagem , Amidas/farmacologia , Cobaias , Pirazinas/administração & dosagem , Pirazinas/uso terapêutico , Doença pelo Vírus Ebola/tratamento farmacológico , Camundongos , Ebolavirus/efeitos dos fármacos , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Feminino , Vias de Administração de Medicamentos , Esquema de Medicação
18.
PLoS One ; 19(7): e0307579, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39052567

RESUMO

VP30 and VP40 proteins of Ebola and Marburg viruses have been recognized as potential targets for antiviral drug development due to their essential roles in the viral lifecycle. Targeting these proteins could disrupt key stages of the viral replication process, inhibiting the viruses' ability to propagate and cause disease. The current study aims to perform molecular docking and virtual screening on deep-sea fungal metabolites targeting Marburg virus VP40 Dimer, matrix protein VP40 from Ebola virus Sudan, Ebola VP35 Interferon Inhibitory Domain, and VP35 from Marburg virus. The top ten compounds for each protein target were chosen using the glide score. All the compounds obtained indicate a positive binding interaction. Furthermore, AdmetSAR was utilized to investigate the pharmacokinetics of the inhibitors chosen. Gliotoxin was used as a ligand with Marburg virus VP40 Dimer, Austinol with matrix protein VP40 from Ebola virus Sudan, Ozazino-cyclo-(2,3-dihydroxyl-trp-tyr) with Ebola VP35 Interferon Inhibitory Domain, and Dehydroaustinol with VP35 from Marburg virus. MD modeling and MMPBSA studies were used to provide a better understanding of binding behaviors. Pre-clinical experiments can assist validate our in-silico studies and assess whether the molecule can be employed as an anti-viral drug.


Assuntos
Antivirais , Ebolavirus , Marburgvirus , Simulação de Acoplamento Molecular , Ebolavirus/efeitos dos fármacos , Ebolavirus/metabolismo , Marburgvirus/efeitos dos fármacos , Marburgvirus/metabolismo , Antivirais/farmacologia , Antivirais/química , Proteínas da Matriz Viral/metabolismo , Proteínas da Matriz Viral/antagonistas & inibidores , Proteínas da Matriz Viral/química , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/virologia , Humanos , Simulação por Computador , Simulação de Dinâmica Molecular , Proteínas Virais Reguladoras e Acessórias
19.
J Math Biol ; 89(2): 25, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38963509

RESUMO

The Ebola virus disease (EVD) has been endemic since 1976, and the case fatality rate is extremely high. EVD is spread by infected animals, symptomatic individuals, dead bodies, and contaminated environment. In this paper, we formulate an EVD model with four transmission modes and a time delay describing the incubation period. Through dynamical analysis, we verify the importance of blocking the infection source of infected animals. We get the basic reproduction number without considering the infection source of infected animals. And, it is proven that the model has a globally attractive disease-free equilibrium when the basic reproduction number is less than unity; the disease eventually becomes endemic when the basic reproduction number is greater than unity. Taking the EVD epidemic in Sierra Leone in 2014-2016 as an example, we complete the data fitting by combining the effect of the media to obtain the unknown parameters, the basic reproduction number and its time-varying reproduction number. It is shown by parameter sensitivity analysis that the contact rate and the removal rate of infected group have the greatest influence on the prevalence of the disease. And, the disease-controlling thresholds of these two parameters are obtained. In addition, according to the existing vaccination strategy, only the inoculation ratio in high-risk areas is greater than 0.4, the effective reproduction number can be less than unity. And, the earlier the vaccination time, the greater the inoculation ratio, and the faster the disease can be controlled.


Assuntos
Número Básico de Reprodução , Ebolavirus , Doença pelo Vírus Ebola , Conceitos Matemáticos , Modelos Biológicos , Doença pelo Vírus Ebola/transmissão , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/epidemiologia , Número Básico de Reprodução/estatística & dados numéricos , Humanos , Animais , Serra Leoa/epidemiologia , Ebolavirus/patogenicidade , Ebolavirus/fisiologia , Epidemias/estatística & dados numéricos , Epidemias/prevenção & controle , Simulação por Computador , Modelos Epidemiológicos , Surtos de Doenças/prevenção & controle , Surtos de Doenças/estatística & dados numéricos
20.
Virus Res ; 347: 199430, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38964470

RESUMO

A multistep priming process involving furin and endosomal cathepsin B and L (CatB/L) has been described for the Orthoebolavirus zairense (EBOV) glycoprotein GP. Inhibition or knockdown of either furin or endosomal cathepsins, however, did not prevent virus multiplication in cell cultures. Moreover, an EBOV mutant lacking the furin cleavage motif (RRTRR→AGTAA) was able to replicate and cause fatal disease in nonhuman primates, indicating that furin cleavage may be dispensable for virus infectivity. Here, by using protease inhibitors and EBOV GP-carrying recombinant vesicular stomatitis virus (VSV) and transcription and replication-competent virus-like particles (trVLPs) we found that processing of EBOV GP is mediated by different proteases in different cell lines depending on the protease repertoire available. Endosomal cathepsins were essential for EBOV GP entry in Huh-7 but not in Vero cells, in which trypsin-like proteases and stably expressed trypsin-like transmembrane serine protease 2 (TMPRSS2) supported wild-type EBOV GP and EBOV GP_AGTAA mutant entry. Furthermore, we show that the EBOV GP_AGTAA mutant is cleaved into fusion-competent GP2 by TMPRSS2 and by CatL at a so far unknown site. Fluorescence microscopy co-localization studies indicate that EBOV GP cleavage by TMPRSS2 may occur in the TGN prior to virus release or in the late endosome at the stage of virus entry into a new cell. Our data show that EBOV GP must be proteolytically activated to support virus entry but has even greater flexibility in terms of proteases and the precise cleavage site than previously assumed.


Assuntos
Catepsina L , Ebolavirus , Furina , Serina Endopeptidases , Proteínas do Envelope Viral , Internalização do Vírus , Catepsina L/metabolismo , Catepsina L/genética , Furina/metabolismo , Furina/genética , Ebolavirus/genética , Ebolavirus/fisiologia , Ebolavirus/metabolismo , Animais , Humanos , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Chlorocebus aethiops , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Proteólise , Células Vero , Linhagem Celular , Endossomos/metabolismo , Endossomos/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...