Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 6785, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880247

RESUMO

Marburg and Ebola filoviruses are two of the deadliest infectious agents and several outbreaks have occurred in the last decades. Although several receptors and co-receptors have been reported for Ebola virus, key host factors remain to be elucidated. In this study, using a haploid cell screening platform, we identify the guanine nucleotide exchange factor CCZ1 as a key host factor in the early stage of filovirus replication. The critical role of CCZ1 for filovirus infections is validated in 3D primary human hepatocyte cultures and human blood-vessel organoids, both critical target sites for Ebola and Marburg virus tropism. Mechanistically, CCZ1 controls early to late endosomal trafficking of these viruses. In addition, we report that CCZ1 has a role in the endosomal trafficking of endocytosis-dependent SARS-CoV-2 infections, but not in infections by Lassa virus, which enters endo-lysosomal trafficking at the late endosome stage. Thus, we have identified an essential host pathway for filovirus infections in cell lines and engineered human target tissues. Inhibition of CCZ1 nearly completely abolishes Marburg and Ebola infections. Thus, targeting CCZ1 could potentially serve as a promising drug target for controlling infections caused by various viruses, such as SARS-CoV-2, Marburg, and Ebola.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Doença do Vírus de Marburg , Marburgvirus , Proteínas de Transporte Vesicular , Animais , Humanos , Ebolavirus/metabolismo , Lisossomos , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/metabolismo , Marburgvirus/metabolismo , Proteínas de Transporte Vesicular/metabolismo
2.
Viruses ; 15(8)2023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37632063

RESUMO

The COVID-19 pandemic has not only strained healthcare systems in Africa but has also intensified the impact of emerging and re-emerging diseases. Specifically in Equatorial Guinea, mirroring the situation in other African countries, unique zoonotic outbreaks have occurred during this challenging period. One notable resurgence is Marburg virus disease (MVD), which has further burdened the already fragile healthcare system. The re-emergence of the Marburg virus amid the COVID-19 pandemic is believed to stem from a probable zoonotic spill-over, although the precise transmission routes remain uncertain. Given the gravity of the situation, addressing the existing challenges is paramount. Though the genome sequences from the current outbreak were not available for this study, we analyzed all the available whole genome sequences of this re-emerging pathogen to advocate for a shift towards active surveillance. This is essential to ensure the successful containment of any potential Marburg virus outbreak in Equatorial Guinea and the wider African context. This study, which presents an update on the phylodynamics and the genetic variability of MARV, further confirmed the existence of at least two distinct patterns of viral spread. One pattern demonstrates a slower but continuous and recurring virus circulation, while the other exhibits a faster yet limited and episodic spread. These results highlight the critical need to strengthen genomic surveillance in the region to effectively curb the pathogen's dissemination. Moreover, the study emphasizes the importance of prompt alert management, comprehensive case investigation and analysis, contact tracing, and active case searching. These steps are vital to support the healthcare system's response to this emerging health crisis. By implementing these strategies, we can better arm ourselves against the challenges posed by the resurgence of the Marburg virus and other infectious diseases.


Assuntos
Doença do Vírus de Marburg , Marburgvirus , Animais , Humanos , África/epidemiologia , População Negra , COVID-19/epidemiologia , Marburgvirus/genética , Pandemias , Doença do Vírus de Marburg/epidemiologia , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/virologia , Surtos de Doenças , Guiné Equatorial/epidemiologia , Zoonoses Virais/epidemiologia , Zoonoses Virais/genética , Zoonoses Virais/virologia , Filogenia
3.
Int Immunopharmacol ; 111: 109109, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35926269

RESUMO

Marburgvirus (MARV), a member of the Filovirus family, causes severe hemorrhagic fever in humans. Currently, there are no approved vaccines or post exposure treatment methods available against MARV. With the aim of identifying vaccine candidates against MARV, we employ different sequence-based computational methods to predict the MHC-I and MHC-II T-cell epitopes as well as B-cell epitopes for the complete MARV genome. We analyzed the variations in the predicted epitopes among four MARV variants, the Lake Victoria, Angola, Musoke, and Ravn. We used a consensus approach to identify several epitopes, including novel epitopes, and narrowed down the selection based on different parameters such as antigenicity and IC50 values. The selected epitopes can be used in various vaccine constructs that give effective antibody responses. The MHC-I epitope-allele complexes for GP and NP with favorably low IC50 values were investigated using molecular dynamics computations to determine the molecular details of the epitope-allele complexes. This study provides information for further experimental validation of the potential epitopes and the design and development of MARV vaccines.


Assuntos
Doença do Vírus de Marburg , Marburgvirus , Vacinas Virais , Alelos , Animais , Epitopos de Linfócito B/genética , Epitopos de Linfócito T/genética , Humanos , Doença do Vírus de Marburg/genética , Marburgvirus/genética
4.
Front Immunol ; 12: 703986, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484200

RESUMO

Ebola (EBOV), Marburg (MARV) and Sudan (SUDV) viruses are the three filoviruses which have caused the most fatalities in humans. Transmission from animals into the human population typically causes outbreaks of limited scale in endemic regions. In contrast, the 2013-16 outbreak in several West African countries claimed more than 11,000 lives revealing the true epidemic potential of filoviruses. This is further emphasized by the difficulty seen with controlling the 2018-2020 outbreak of EBOV in the Democratic Republic of Congo (DRC), despite the availability of two emergency use-approved vaccines and several experimental therapeutics targeting EBOV. Moreover, there are currently no vaccine options to protect against the other epidemic filoviruses. Protection of a monovalent EBOV vaccine against other filoviruses has never been demonstrated in primate challenge studies substantiating a significant void in capability should a MARV or SUDV outbreak of similar magnitude occur. Herein we show progress on developing vaccines based on recombinant filovirus glycoproteins (GP) from EBOV, MARV and SUDV produced using the Drosophila S2 platform. The highly purified recombinant subunit vaccines formulated with CoVaccine HT™ adjuvant have not caused any safety concerns (no adverse reactions or clinical chemistry abnormalities) in preclinical testing. Candidate formulations elicit potent immune responses in mice, guinea pigs and non-human primates (NHPs) and consistently produce high antigen-specific IgG titers. Three doses of an EBOV candidate vaccine elicit full protection against lethal EBOV infection in the cynomolgus challenge model while one of four animals infected after only two doses showed delayed onset of Ebola Virus Disease (EVD) and eventually succumbed to infection while the other three animals survived challenge. The monovalent MARV or SUDV vaccine candidates completely protected cynomolgus macaques from infection with lethal doses of MARV or SUDV. It was further demonstrated that combinations of MARV or SUDV with the EBOV vaccine can be formulated yielding bivalent vaccines retaining full efficacy. The recombinant subunit vaccine platform should therefore allow the development of a safe and efficacious multivalent vaccine candidate for protection against Ebola, Marburg and Sudan Virus Disease.


Assuntos
Vacinas contra Ebola/farmacologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Animais , Vacinas contra Ebola/genética , Vacinas contra Ebola/imunologia , Ebolavirus/genética , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/genética , Doença pelo Vírus Ebola/imunologia , Humanos , Macaca fascicularis , Doença do Vírus de Marburg/epidemiologia , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/imunologia , Marburgvirus/genética , Vacinas Sintéticas
5.
Viruses ; 13(2)2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673144

RESUMO

Filoviruses Ebola (EBOV) and Marburg (MARV) are devastating high-priority pathogens capable of causing explosive outbreaks with high human mortality rates. The matrix proteins of EBOV and MARV, as well as eVP40 and mVP40, respectively, are the key viral proteins that drive virus assembly and egress and can bud independently from cells in the form of virus-like particles (VLPs). The matrix proteins utilize proline-rich Late (L) domain motifs (e.g., PPxY) to hijack specific host proteins that contain WW domains, such as the HECT family E3 ligases, to facilitate the last step of virus-cell separation. We identified E3 ubiquitin ligase Smad Ubiquitin Regulatory Factor 2 (SMURF2) as a novel interactor with VP40 that positively regulates VP40 VLP release. Our results show that eVP40 and mVP40 interact with the three WW domains of SMURF2 via their PPxY motifs. We provide evidence that the eVP40-SMURF2 interaction is functional as the expression of SMURF2 positively regulates VLP egress, while siRNA knockdown of endogenous SMURF2 decreases VLP budding compared to controls. In sum, our identification of novel interactor SMURF2 adds to the growing list of identified host proteins that can regulate PPxY-mediated egress of VP40 VLPs. A more comprehensive understanding of the modular interplay between filovirus VP40 and host proteins may lead to the development of new therapies to combat these deadly infections.


Assuntos
Ebolavirus/fisiologia , Doença pelo Vírus Ebola/enzimologia , Doença do Vírus de Marburg/enzimologia , Marburgvirus/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas da Matriz Viral/metabolismo , Liberação de Vírus , Motivos de Aminoácidos , Animais , Ebolavirus/química , Ebolavirus/genética , Doença pelo Vírus Ebola/genética , Doença pelo Vírus Ebola/virologia , Humanos , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/virologia , Marburgvirus/química , Marburgvirus/genética , Ligação Proteica , Ubiquitina-Proteína Ligases/genética , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética , Vírion/genética , Vírion/fisiologia , Montagem de Vírus
6.
Proc Natl Acad Sci U S A ; 117(49): 31142-31148, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33229516

RESUMO

Marburg virus (MARV) disease is lethal, with fatality rates up to 90%. Neutralizing antibodies (Abs) are promising drug candidates to prevent or treat the disease. Current efforts are focused in part on vaccine development to induce such MARV-neutralizing Abs. We analyzed the antibody repertoire from healthy unexposed and previously MARV-infected individuals to assess if naïve repertoires contain suitable precursor antibodies that could become neutralizing with a limited set of somatic mutations. We computationally searched the human Ab variable gene repertoire for predicted structural homologs of the neutralizing Ab MR78 that is specific to the receptor binding site (RBS) of MARV glycoprotein (GP). Eight Ab heavy-chain complementarity determining region 3 (HCDR3) loops from MARV-naïve individuals and one from a previously MARV-infected individual were selected for testing as HCDR3 loop chimeras on the MR78 Ab framework. Three of these chimerized antibodies bound to MARV GP. We then tested a full-length native Ab heavy chain encoding the same 17-residue-long HCDR3 loop that bound to the MARV GP the best among the chimeric Abs tested. Despite only 57% amino acid sequence identity, the Ab from a MARV-naïve donor recognized MARV GP and possessed neutralizing activity against the virus. Crystallization of both chimeric and full-length native heavy chain-containing Abs provided structural insights into the mechanism of binding for these types of Abs. Our work suggests that the MARV GP RBS is a promising candidate for epitope-focused vaccine design to induce neutralizing Abs against MARV.


Assuntos
Anticorpos Antivirais/genética , Regiões Determinantes de Complementaridade/genética , Doença do Vírus de Marburg/imunologia , Marburgvirus/imunologia , Animais , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Regiões Determinantes de Complementaridade/imunologia , Epitopos/genética , Epitopos/imunologia , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Doença do Vírus de Marburg/tratamento farmacológico , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/virologia , Marburgvirus/patogenicidade , Mutação/genética , Mutação/imunologia , Proteínas do Envelope Viral , Vacinas Virais/genética , Vacinas Virais/imunologia
7.
Sci Rep ; 10(1): 3071, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32080323

RESUMO

Postexposure immunization can prevent disease and reduce transmission following pathogen exposure. The rapid immunostimulatory properties of recombinant vesicular stomatitis virus (rVSV)-based vaccines make them suitable postexposure treatments against the filoviruses Ebola virus and Marburg virus (MARV); however, the mechanisms that drive this protection are undefined. Previously, we reported 60-75% survival of rhesus macaques treated with rVSV vectors expressing MARV glycoprotein (GP) 20-30 minutes after a low dose exposure to the most pathogenic variant of MARV, Angola. Survival in this model was linked to production of GP-specific antibodies and lower viral load. To confirm these results and potentially identify novel correlates of postexposure protection, we performed a similar experiment, but analyzed plasma cytokine levels, frequencies of immune cell subsets, and the transcriptional response to infection in peripheral blood. In surviving macaques (80-89%), we observed induction of genes mapping to antiviral and interferon-related pathways early after treatment and a higher percentage of T helper 1 (Th1) and NK cells. In contrast, the response of non-surviving macaques was characterized by hypercytokinemia; a T helper 2 signature; recruitment of low HLA-DR expressing monocytes and regulatory T-cells; and transcription of immune checkpoint (e.g., PD-1, LAG3) genes. These results suggest dysregulated immunoregulation is associated with poor prognosis, whereas early innate signaling and Th1-skewed immunity are important for survival.


Assuntos
Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Marburgvirus/imunologia , Profilaxia Pós-Exposição , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Citocinas/sangue , Citotoxicidade Imunológica , Relação Dose-Resposta Imunológica , Regulação para Baixo/genética , Feminino , Inflamação/sangue , Inflamação/imunologia , Interferons/genética , Interferons/metabolismo , Células Matadoras Naturais/imunologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Masculino , Doença do Vírus de Marburg/sangue , Doença do Vírus de Marburg/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recombinação Genética/genética , Linfócitos T Auxiliares-Indutores/imunologia , Células Th1/imunologia , Células Th2/imunologia , Transcriptoma/genética , Regulação para Cima/genética , Vesiculovirus/genética , Carga Viral/imunologia
8.
Viruses ; 10(11)2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30400182

RESUMO

The Egyptian rousette bat (ERB) is the only known Marburg virus (MARV) reservoir host. ERBs develop a productive MARV infection with low viremia and shedding but no overt disease, suggesting this virus is efficiently controlled by ERB antiviral responses. This dynamic would contrast with humans, where MARV-mediated interferon (IFN) antagonism early in infection is thought to contribute to the severe, often fatal disease. The newly-annotated ERB genome and transcriptome have now enabled us to use a custom-designed NanoString nCounter ERB CodeSet in conjunction with RNA-seq to investigate responses in a MARV-infected ERB cell line. Both transcriptomic platforms correlated well and showed that MARV inhibited the antiviral program in ERB cells, while an IFN antagonism-impaired MARV was less efficient at suppressing the response gene induction, phenotypes previously reported for primate cells. Interestingly, and despite the expansion of IFN loci in the ERB genome, neither MARV showed specific induction of almost any IFN gene. However, we detected an upregulation of putative, unannotated ERB antiviral paralogs, as well as an elevated basal expression in uninfected ERB cells of key antiviral genes.


Assuntos
Quirópteros/genética , Quirópteros/virologia , Resistência à Doença/genética , Interações Hospedeiro-Patógeno/genética , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/virologia , Marburgvirus/fisiologia , Transcriptoma , Animais , Linhagem Celular , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Imunidade Inata/genética , Interferons/farmacologia
9.
Sci Rep ; 7(1): 3390, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28611428

RESUMO

Marburg virus (MARV) has caused outbreaks of filoviral hemorrhagic fever since its discovery in 1967. The largest and deadliest outbreak occurred in Angola in 2005, with 252 cases and 227 deaths. In 2014, we developed a mouse-adapted MARV, Angola variant through serial passaging in mice. The mouse-adapted MARV exhibits many of the hallmarks of MARV disease in humans. By applying deep-sequencing to every passage of the virus, we are able to study virus evolution in this host with surprising precision. We show that two regions go through substantial changes: the intergenic region between NP and VP35, as well as the first 100 amino acids of the VP40 protein. Our results also reveal that there were profound changes during the production of the final virus stock in cell culture. Overall, our results show that a handful of regions carry most of the mutations acquired during the adaptation of the virus to a new host and that many mutations become fixed very early during the adaptation process.


Assuntos
Adaptação Biológica/genética , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Doença do Vírus de Marburg/patologia , Doença do Vírus de Marburg/virologia , Marburgvirus/genética , Proteínas Virais/genética , Animais , Células Cultivadas , Doença do Vírus de Marburg/genética , Marburgvirus/crescimento & desenvolvimento , Marburgvirus/isolamento & purificação , Camundongos , RNA Viral/genética , Inoculações Seriadas , Carga Viral
10.
J Infect Dis ; 212 Suppl 2: S404-9, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26022440

RESUMO

Stat1(-/-) mice lack a response to interferon α, ß, and γ, allowing for replication of nonadapted wild-type (wt) Ebolavirus and Marburgvirus. We sought to establish a mouse model for efficacy testing of live attenuated recombinant vesicular stomatitis virus (rVSV)-based filovirus vaccine vectors using wt Ebolavirus and Marburgvirus challenge strains. While infection of immunocompetent mice with different rVSV-based filovirus vectors did not cause disease, infection of Stat1(-/-) mice with the same vectors resulted in systemic infection and lethal outcome for the majority of tested rVSVs. Despite differences in viral loads, organ tropism was remarkably similar between rVSV filovirus vaccine vectors and rVSVwt, with the exception of the brain. In conclusion, Stat1(-/-) mice are not an appropriate immunocompromised mouse model for efficacy testing of live attenuated, replication-competent rVSV vaccine vectors.


Assuntos
Filoviridae/imunologia , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética , Vacinas Atenuadas/imunologia , Estomatite Vesicular/imunologia , Vacinas Virais/imunologia , Animais , Chlorocebus aethiops , Modelos Animais de Doenças , Ebolavirus/imunologia , Infecções por Filoviridae/genética , Infecções por Filoviridae/imunologia , Infecções por Filoviridae/virologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Doença pelo Vírus Ebola/genética , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Marburgvirus/imunologia , Camundongos , Fator de Transcrição STAT1/imunologia , Células Vero , Carga Viral/imunologia , Replicação Viral/genética , Replicação Viral/imunologia
11.
J Infect Dis ; 209(4): 562-70, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23990568

RESUMO

BACKGROUND: Marburg virus (MARV) infection causes severe morbidity and mortality in humans and nonhuman primates. Currently, there are no licensed therapeutics available for treating MARV infection. Here, we present the in vitro development and in vivo evaluation of lipid-encapsulated small interfering RNA (siRNA) as a potential therapeutic for the treatment of MARV infection. METHODS: The activity of anti-MARV siRNAs was assessed using dual luciferase reporter assays followed by in vitro testing against live virus. Lead candidates were tested in lethal guinea pig models of 3 different MARV strains (Angola, Ci67, Ravn). RESULTS: Treatment resulted in 60%-100% survival of guinea pigs infected with MARV. Although treatment with siRNA targeting other MARV messenger RNA (mRNA) had a beneficial effect, targeting the MARV NP mRNA resulted in the highest survival rates. NP-718m siRNA in lipid nanoparticles provided 100% protection against MARV strains Angola and Ci67, and 60% against Ravn. A cocktail containing NP-718m and NP-143m provided 100% protection against MARV Ravn. CONCLUSIONS: These data show protective efficacy against the most pathogenic Angola strain of MARV. Further development of the lipid nanoparticle technology has the potential to yield effective treatments for MARV infection.


Assuntos
Lipídeos/administração & dosagem , Doença do Vírus de Marburg/tratamento farmacológico , Doença do Vírus de Marburg/prevenção & controle , Nanopartículas/administração & dosagem , Nanopartículas/química , RNA Interferente Pequeno/administração & dosagem , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/análise , Citocinas/sangue , Portadores de Fármacos/química , Feminino , Genes Virais , Cobaias , Lipídeos/química , Fígado/química , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/metabolismo , Marburgvirus/efeitos dos fármacos , Marburgvirus/genética , Camundongos , Camundongos Endogâmicos ICR , RNA Interferente Pequeno/química , Proteínas de Ligação a RNA , Análise de Sobrevida , Carga Viral
12.
Forensic Sci Int ; 233(1-3): 338-47, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24314539

RESUMO

Marburgvirus is one of the most important hemorrhagic fever viruses with extremely high infectivity and fatality rate (~90%). It is transmitted easily in human populations through a respiratory route and therefore considered as a major biothreat agent. Although detection assays have been developed, no assay is available for forensic analysis. Here we report development of forensic assays for Marburgvirus. We performed detailed phylogenetic analysis of strains and isolates from all known Marburg virus outbreaks as well as from several laboratory strains and identified canonical SNPs for all major clades (outbreaks) and strains. TaqMan-MGB allelic discrimination assays targeting these SNPs were designed and experimentally screened against synthetic RNA templates and genomic RNAs. A total of 45 assays were validated to provide 100% coverage of the clades (outbreaks) and 91% at the strain level (21 out of the 23 targeted Marburgvirus strains) with built-in redundancy for increased robustness. Using these validated assays, we were able to provide accurate forensic analysis on 3 "unknown" Marburgviruses. These high-resolution forensic assays allow rapid and accurate genotyping of Marburgviruses for forensic investigations.


Assuntos
Marburgvirus/genética , Polimorfismo de Nucleotídeo Único , Animais , Primers do DNA , Genoma Viral , Genótipo , Humanos , Doença do Vírus de Marburg/epidemiologia , Doença do Vírus de Marburg/genética , Filogenia , Reação em Cadeia da Polimerase , RNA Viral/análise , Análise de Sequência
13.
J Virol ; 87(5): 2608-16, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23255795

RESUMO

Viruses in the Ebolavirus and Marburgvirus genera (family Filoviridae) have been associated with large outbreaks of hemorrhagic fever in human and nonhuman primates. The first documented cases occurred in primates over 45 years ago, but the amount of virus genetic diversity detected within bat populations, which have recently been identified as potential reservoir hosts, suggests that the filoviruses are much older. Here, detailed Bayesian coalescent phylogenetic analyses are performed on 97 whole-genome sequences, 55 of which are newly reported, to comprehensively examine molecular evolutionary rates and estimate dates of common ancestry for viruses within the family Filoviridae. Molecular evolutionary rates for viruses belonging to different species range from 0.46 × 10(-4) nucleotide substitutions/site/year for Sudan ebolavirus to 8.21 × 10(-4) nucleotide substitutions/site/year for Reston ebolavirus. Most recent common ancestry can be traced back only within the last 50 years for Reston ebolavirus and Zaire ebolavirus species and suggests that viruses within these species may have undergone recent genetic bottlenecks. Viruses within Marburg marburgvirus and Sudan ebolavirus species can be traced back further and share most recent common ancestors approximately 700 and 850 years before the present, respectively. Examination of the whole family suggests that members of the Filoviridae, including the recently described Lloviu virus, shared a most recent common ancestor approximately 10,000 years ago. These data will be valuable for understanding the evolution of filoviruses in the context of natural history as new reservoir hosts are identified and, further, for determining mechanisms of emergence, pathogenicity, and the ongoing threat to public health.


Assuntos
Ebolavirus/genética , Genoma Viral , Doença pelo Vírus Ebola/genética , Doença do Vírus de Marburg/genética , Marburgvirus/genética , Substituição de Aminoácidos , Animais , Sequência de Bases , Quirópteros/virologia , Ebolavirus/classificação , Evolução Molecular , Variação Genética , Doença pelo Vírus Ebola/epidemiologia , Humanos , Doença do Vírus de Marburg/epidemiologia , Marburgvirus/classificação , Dados de Sequência Molecular , Filogenia , Primatas/virologia , Análise de Sequência de DNA , Proteínas Virais/química , Proteínas Virais/genética
14.
J Virol ; 86(6): 3284-92, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22238307

RESUMO

Filoviruses are enveloped viruses that cause sporadic outbreaks of severe hemorrhagic fever [CDC, MMWR Morb. Mortal. Wkly. Rep. 50:73-77, 2001; Colebunders and Borchert, J. Infect. 40:16-20, 2000; Colebunders et al., J. Infect. Dis. 196(Suppl. 2):S148-S153, 2007; Geisbert and Jahrling, Nat. Med. 10:S110-S121, 2004]. Previous studies revealed that endosomal cysteine proteases are host factors for ebolavirus Zaire (Chandran et al., Science 308:1643-1645, 2005; Schornberg et al., J. Virol. 80:4174-4178, 2006). In this report, we show that infection mediated by glycoproteins from other phylogenetically diverse filoviruses are also dependent on these proteases and provide additional evidence indicating that they cleave GP1 and expose the binding domain for the critical host factor Niemann-Pick C1. Using selective inhibitors and knockout-derived cell lines, we show that the ebolaviruses Zaire and Cote d'Ivoire are strongly dependent on cathepsin B, while the ebolaviruses Sudan and Reston and Marburg virus are not. Taking advantage of previous studies of cathepsin B inhibitor-resistant viruses (Wong et al., J. Virol. 84:163-175, 2010), we found that virus-specific differences in the requirement for cathepsin B are correlated with sequence polymorphisms at residues 47 in GP1 and 584 in GP2. We applied these findings to the analysis of additional ebolavirus isolates and correctly predicted that the newly identified ebolavirus species Bundibugyo, containing D47 and I584, is cathepsin B dependent and that ebolavirus Zaire-1995, the single known isolate of ebolavirus Zaire that lacks D47, is not. We also obtained evidence for virus-specific differences in the role of cathepsin L, including cooperation with cathepsin B. These studies strongly suggest that the use of endosomal cysteine proteases as host factors for entry is a general property of members of the family Filoviridae.


Assuntos
Cisteína Proteases/metabolismo , Ebolavirus/fisiologia , Endossomos/enzimologia , Doença pelo Vírus Ebola/enzimologia , Doença do Vírus de Marburg/enzimologia , Marburgvirus/fisiologia , Internalização do Vírus , Animais , Linhagem Celular , Cisteína Proteases/genética , Ebolavirus/genética , Endossomos/genética , Doença pelo Vírus Ebola/genética , Doença pelo Vírus Ebola/virologia , Humanos , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/virologia , Marburgvirus/genética , Especificidade da Espécie
15.
Nat Med ; 16(9): 991-4, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20729866

RESUMO

Currently, no vaccines or therapeutics are licensed to counter Ebola or Marburg viruses, highly pathogenic filoviruses that are causative agents of viral hemorrhagic fever. Here we show that administration of positively charged phosphorodiamidate morpholino oligomers (PMOplus), delivered by various dosing strategies initiated 30-60 min after infection, protects>60% of rhesus monkeys against lethal Zaire Ebola virus (ZEBOV) and 100% of cynomolgus monkeys against Lake Victoria Marburg virus (MARV) infection. PMOplus may be useful for treating these and other highly pathogenic viruses in humans.


Assuntos
Infecções por Filoviridae/genética , Infecções por Filoviridae/prevenção & controle , Vacinas Sintéticas/uso terapêutico , Vacinas Virais/uso terapêutico , Animais , Ebolavirus/isolamento & purificação , Infecções por Filoviridae/imunologia , Infecções por Filoviridae/mortalidade , Doença pelo Vírus Ebola/genética , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Macaca mulatta/imunologia , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/isolamento & purificação , Primatas , Segurança
16.
J Virol ; 80(19): 9659-66, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973570

RESUMO

Marburg virus (MARV) has been associated with sporadic episodes of hemorrhagic fever, including a recent highly publicized outbreak in Angola that produced severe disease and significant mortality in infected patients. MARV is also considered to have potential as a biological weapon. Recently, we reported the development of a promising attenuated, replication-competent vaccine against MARV based on recombinant vesicular stomatitis virus (VSV) expressing the glycoprotein of the Musoke strain of MARV (VSVDeltaG/MARVGP-Musoke). We used this vaccine to demonstrate complete protection of cynomolgus monkeys against a homologous MARV challenge. While these results are highly encouraging, an effective vaccine would need to confer protection against all relevant strains of MARV. Here, we evaluated the protective efficacy of the VSVDeltaG/MARVGP-Musoke vaccine against two heterologous MARV strains, the seemingly more pathogenic Angola strain and the more distantly related Ravn strain. In this study, seven cynomolgus monkeys were vaccinated with the VSVDeltaG/MARVGP-Musoke vector. Three of these animals were challenged with the Angola strain, three with the Ravn strain, and a single animal with the Musoke strain of MARV. Two animals served as controls and were each injected with a nonspecific VSV vector; these controls were challenged with the Angola and Ravn strains, respectively. Both controls succumbed to challenge by day 8. However, none of the specifically vaccinated animals showed any evidence of illness either from the vaccination or from the MARV challenges and all of these animals survived. These data suggest that the VSVDeltaG/MARVGP-Musoke vaccine should be sufficient to protect against all known MARV strains.


Assuntos
Doença do Vírus de Marburg/imunologia , Marburgvirus/classificação , Marburgvirus/imunologia , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos/imunologia , Reações Cruzadas/imunologia , Macaca fascicularis , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/metabolismo , Marburgvirus/genética , Marburgvirus/metabolismo , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Viremia/sangue , Viremia/imunologia , Viremia/virologia
17.
J Virol ; 80(13): 6497-516, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16775337

RESUMO

In March 2005, the Centers for Disease Control and Prevention (CDC) investigated a large hemorrhagic fever (HF) outbreak in Uige Province in northern Angola, West Africa. In total, 15 initial specimens were sent to CDC, Atlanta, Ga., for testing for viruses associated with viral HFs known to be present in West Africa, including ebolavirus. Marburgvirus was also included despite the fact that the origins of all earlier outbreaks were linked directly to East Africa. Surprisingly, marburgvirus was confirmed (12 of 15 specimens) as the cause of the outbreak. The outbreak likely began in October 2004 and ended in July 2005, and it included 252 cases and 227 (90%) fatalities (report from the Ministry of Health, Republic of Angola, 2005), making it the largest Marburg HF outbreak on record. A real-time quantitative reverse transcription-PCR assay utilized and adapted during the outbreak proved to be highly sensitive and sufficiently robust for field use. Partial marburgvirus RNA sequence analysis revealed up to 21% nucleotide divergence among the previously characterized East African strains, with the most distinct being Ravn from Kenya (1987). The Angolan strain was less different ( approximately 7%) from the main group of East African marburgviruses than one might expect given the large geographic separation. To more precisely analyze the virus genetic differences between outbreaks and among viruses within the Angola outbreak itself, a total of 16 complete virus genomes were determined, including those of the virus isolates Ravn (Kenya, 1987) and 05DRC, 07DRC, and 09DRC (Democratic Republic of Congo, 1998) and the reference Angolan virus isolate (Ang1379v). In addition, complete genome sequences were obtained from RNAs extracted from 10 clinical specimens reflecting various stages of the disease and locations within the Angolan outbreak. While the marburgviruses exhibit high overall genetic diversity (up to 22%), only 6.8% nucleotide difference was found between the West African Angolan viruses and the majority of East African viruses, suggesting that the virus reservoir species in these regions are not substantially distinct. Remarkably few nucleotide differences were found among the Angolan clinical specimens (0 to 0.07%), consistent with an outbreak scenario in which a single (or rare) introduction of virus from the reservoir species into the human population was followed by person-to-person transmission with little accumulation of mutations. This is in contrast to the 1998 to 2000 marburgvirus outbreak, where evidence of several virus genetic lineages (with up to 21% divergence) and multiple virus introductions into the human population was found.


Assuntos
Surtos de Doenças , Genoma Viral/genética , Doença do Vírus de Marburg/genética , Doença do Vírus de Marburg/mortalidade , Marburgvirus/genética , Mutação , Angola/epidemiologia , Sequência de Bases , Surtos de Doenças/história , Feminino , História do Século XXI , Humanos , Quênia/epidemiologia , Masculino , Doença do Vírus de Marburg/história , Doença do Vírus de Marburg/transmissão , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de RNA , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...