Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 6785, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880247

RESUMEN

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.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Enfermedad del Virus de Marburg , Marburgvirus , Proteínas de Transporte Vesicular , Animales , Humanos , Ebolavirus/metabolismo , Lisosomas , Enfermedad del Virus de Marburg/genética , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/metabolismo , Proteínas de Transporte Vesicular/metabolismo
2.
Mol Ther ; 31(1): 269-281, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36114672

RESUMEN

Marburg virus (MARV) infection results in severe viral hemorrhagic fever with mortalities up to 90%, and there is a pressing need for effective therapies. Here, we established a small interfering RNA (siRNA) conjugate platform that enabled successful subcutaneous delivery of siRNAs targeting the MARV nucleoprotein. We identified a hexavalent mannose ligand with high affinity to macrophages and dendritic cells, which are key cellular targets of MARV infection. This ligand enabled successful siRNA conjugate delivery to macrophages both in vitro and in vivo. The delivered hexa-mannose-siRNA conjugates rendered substantial target gene silencing in macrophages when supported by a mannose functionalized endosome release polymer. This hexa-mannose-siRNA conjugate was further evaluated alongside our hepatocyte-targeting GalNAc-siRNA conjugate, to expand targeting of infected liver cells. In MARV-Angola-infected guinea pigs, these platforms offered limited survival benefit when used as individual agents. However, in combination, they achieved up to 100% protection when dosed 24 h post infection. This novel approach, using two different ligands to simultaneously deliver siRNA to multiple cell types relevant to infection, provides a convenient subcutaneous route of administration for treating infection by these dangerous pathogens. The mannose conjugate platform has potential application to other diseases involving macrophages and dendritic cells.


Asunto(s)
Enfermedad del Virus de Marburg , Marburgvirus , Virosis , Animales , Cobayas , ARN Interferente Pequeño/genética , Manosa , Ligandos , ARN Bicatenario , Marburgvirus/genética , Enfermedad del Virus de Marburg/metabolismo , Enfermedad del Virus de Marburg/prevención & control
3.
Front Immunol ; 12: 774026, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777392

RESUMEN

Marburg virus (MARV) is a member of the filovirus family that causes hemorrhagic disease with high case fatality rates. MARV is on the priority list of the World Health Organization for countermeasure development highlighting its potential impact on global public health. We developed a vesicular stomatitis virus (VSV)-based vaccine expressing the MARV glycoprotein (VSV-MARV) and previously demonstrated uniform protection of nonhuman primates (NHPs) with a single dose. Here, we investigated the fast-acting potential of this vaccine by challenging NHPs with MARV 14, 7 or 3 days after a single dose vaccination with VSV-MARV. We found that 100% of the animals survived when vaccinated 7 or 14 days and 75% of the animal survived when vaccinated 3 days prior to lethal MARV challenge. Transcriptional analysis of whole blood samples indicated activation of B cells and antiviral defense after VSV-MARV vaccination. In the day -14 and -7 groups, limited transcriptional changes after challenge were observed with the exception of day 9 post-challenge in the day -7 group where we detected gene expression profiles indicative of a recall response. In the day -3 group, transcriptional analysis of samples from surviving NHPs revealed strong innate immune activation. In contrast, the animal that succumbed to disease in this group lacked signatures of antiviral immunity. In summary, our data demonstrate that the VSV-MARV is a fast-acting vaccine suitable for the use in emergency situations like disease outbreaks in Africa.


Asunto(s)
Enfermedad del Virus de Marburg/prevención & control , Marburgvirus/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Chlorocebus aethiops , Citocinas/sangre , Modelos Animales de Enfermedad , Inmunización , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Activación de Linfocitos , Enfermedad del Virus de Marburg/sangre , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/metabolismo , Vacunación , Células Vero , Vesiculovirus , Carga Viral
4.
J Biol Chem ; 296: 100796, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019871

RESUMEN

Marburg virus (MARV) is a lipid-enveloped virus harboring a negative-sense RNA genome, which has caused sporadic outbreaks of viral hemorrhagic fever in sub-Saharan Africa. MARV assembles and buds from the host cell plasma membrane where MARV matrix protein (mVP40) dimers associate with anionic lipids at the plasma membrane inner leaflet and undergo a dynamic and extensive self-oligomerization into the structural matrix layer. The MARV matrix layer confers the virion filamentous shape and stability but how host lipids modulate mVP40 oligomerization is mostly unknown. Using in vitro and cellular techniques, we present a mVP40 assembly model highlighting two distinct oligomerization interfaces: the (N-terminal domain [NTD] and C-terminal domain [CTD]) in mVP40. Cellular studies of NTD and CTD oligomerization interface mutants demonstrate the importance of each interface in matrix assembly. The assembly steps include protein trafficking to the plasma membrane, homo-multimerization that induced protein enrichment, plasma membrane fluidity changes, and elongations at the plasma membrane. An ascorbate peroxidase derivative (APEX)-transmission electron microscopy method was employed to closely assess the ultrastructural localization and formation of viral particles for wildtype mVP40 and NTD and CTD oligomerization interface mutants. Taken together, these studies present a mechanistic model of mVP40 oligomerization and assembly at the plasma membrane during virion assembly that requires interactions with phosphatidylserine for NTD-NTD interactions and phosphatidylinositol-4,5-bisphosphate for proper CTD-CTD interactions. These findings have broader implications in understanding budding of lipid-enveloped viruses from the host cell plasma membrane and potential strategies to target protein-protein or lipid-protein interactions to inhibit virus budding.


Asunto(s)
Enfermedad del Virus de Marburg/virología , Marburgvirus/fisiología , Lípidos de la Membrana/metabolismo , Proteínas de la Matriz Viral/metabolismo , Virión/metabolismo , Animales , Células COS , Membrana Celular/química , Membrana Celular/metabolismo , Chlorocebus aethiops , Células HEK293 , Humanos , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/química , Lípidos de la Membrana/química , Modelos Moleculares , Multimerización de Proteína , Proteínas de la Matriz Viral/química , Virión/química , Ensamble de Virus
5.
Cell Host Microbe ; 24(3): 405-416.e3, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30173956

RESUMEN

Sexual transmission of filoviruses was first reported in 1968 after an outbreak of Marburg virus (MARV) disease and recently caused flare-ups of Ebola virus disease in the 2013-2016 outbreak. How filoviruses establish testicular persistence and are shed in semen remain unknown. We discovered that persistent MARV infection of seminiferous tubules, an immune-privileged site that harbors sperm production, is a relatively common event in crab-eating macaques that survived infection after antiviral treatment. Persistence triggers severe testicular damage, including spermatogenic cell depletion and inflammatory cell invasion. MARV mainly persists in Sertoli cells, leading to breakdown of the blood-testis barrier formed by inter-Sertoli cell tight junctions. This disruption is accompanied by local infiltration of immunosuppressive CD4+Foxp3+ regulatory T cells. Our study elucidates cellular events associated with testicular persistence that may promote sexual transmission of filoviruses and suggests that targeting immunosuppression may be warranted to clear filovirus persistence in damaged immune-privileged sites.


Asunto(s)
Enfermedad del Virus de Marburg/virología , Marburgvirus/fisiología , Enfermedades de los Primates/virología , Testículo/virología , Animales , Macaca , Masculino , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/metabolismo , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/virología , Sobrevivientes , Linfocitos T Reguladores/inmunología , Uniones Estrechas/metabolismo , Uniones Estrechas/virología
6.
J Infect Dis ; 218(suppl_5): S403-S408, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30165526

RESUMEN

Protein kinase R (PKR) is a key antiviral protein involved in sensing and restricting viral infections. Here we analyzed the ability of Marburg virus (MARV) viral protein 35 (VP35) to inhibit PKR activation in human and bat cells. Similar to the related Ebola and Lloviu viruses, MARV VP35 was able to inhibit PKR activation in 293T cells. In contrast, we found that MARV VP35 did not inhibit human or bat PKR activation in human glioblastoma U-251-MG cells or a Rousettus aegyptiacus cell line. Additional experiments revealed that PACT, a known PKR regulator, was insufficient to rescue the ability of VP35 to inhibit PKR activation in these cells. Taken together, this study indicates that the ability of VP35 to inhibit PKR is cell type specific, potentially explaining discrepancies between the ability of filoviruses to potently block innate immune responses, and the high levels of interferon and interferon-stimulated genes observed in filovirus patients.


Asunto(s)
Marburgvirus/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Línea Celular , Quirópteros , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/inmunología , Proteínas Quinasas/inmunología , Proteínas Virales/inmunología , Proteínas Reguladoras y Accesorias Virales/inmunología
7.
J Clin Invest ; 127(12): 4437-4448, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29106386

RESUMEN

Ebolaviruses and marburgviruses belong to the family Filoviridae and cause high lethality in infected patients. There are currently no licensed filovirus vaccines or antiviral therapies. The development of broad-spectrum therapies against members of the Marburgvirus genus, including Marburg virus (MARV) and Ravn virus (RAVV), is difficult because of substantial sequence variability. RNAi therapeutics offer a potential solution, as identification of conserved target nucleotide sequences may confer activity across marburgvirus variants. Here, we assessed the therapeutic efficacy of lipid nanoparticle (LNP) delivery of a single nucleoprotein-targeting (NP-targeting) siRNA in nonhuman primates at advanced stages of MARV or RAVV disease to mimic cases in which patients begin treatment for fulminant disease. Sixteen rhesus monkeys were lethally infected with MARV or RAVV and treated with NP siRNA-LNP, with MARV-infected animals beginning treatment four or five days after infection and RAVV-infected animals starting treatment three or six days after infection. While all untreated animals succumbed to disease, NP siRNA-LNP treatment conferred 100% survival of RAVV-infected macaques, even when treatment began just 1 day prior to the death of the control animals. In MARV-infected animals, day-4 treatment initiation resulted in 100% survival, and day-5 treatment resulted in 50% survival. These results identify a single siRNA therapeutic that provides broad-spectrum protection against both MARV and RAVV.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Enfermedad del Virus de Marburg/tratamiento farmacológico , Marburgvirus , Nanopartículas/uso terapéutico , ARN Interferente Pequeño/farmacología , Animales , Macaca mulatta , Enfermedad del Virus de Marburg/metabolismo , Enfermedad del Virus de Marburg/patología , Nanopartículas/química , ARN Interferente Pequeño/química
8.
Sci Rep ; 6: 34589, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27713552

RESUMEN

The unprecedented outbreak of Ebola in West Africa resulted in over 28,000 cases and 11,000 deaths, underlining the need for a better understanding of the biology of this highly pathogenic virus to develop specific counter strategies. Two filoviruses, the Ebola and Marburg viruses, result in a severe and often fatal infection in humans. However, bats are natural hosts and survive filovirus infections without obvious symptoms. The molecular basis of this striking difference in the response to filovirus infections is not well understood. We report a systematic overview of differentially expressed genes, activity motifs and pathways in human and bat cells infected with the Ebola and Marburg viruses, and we demonstrate that the replication of filoviruses is more rapid in human cells than in bat cells. We also found that the most strongly regulated genes upon filovirus infection are chemokine ligands and transcription factors. We observed a strong induction of the JAK/STAT pathway, of several genes encoding inhibitors of MAP kinases (DUSP genes) and of PPP1R15A, which is involved in ER stress-induced cell death. We used comparative transcriptomics to provide a data resource that can be used to identify cellular responses that might allow bats to survive filovirus infections.


Asunto(s)
Ebolavirus/metabolismo , Regulación de la Expresión Génica , Fiebre Hemorrágica Ebola/metabolismo , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/metabolismo , Transducción de Señal , Transcripción Genética , Animales , Línea Celular Tumoral , Quirópteros , Humanos
9.
J Infect Dis ; 212 Suppl 2: S258-70, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26092858

RESUMEN

BACKGROUND: Phylogenetic comparisons of known Marburg virus (MARV) strains reveal 2 distinct genetic lineages: Ravn and the Lake Victoria Marburg complex (eg, Musoke, Popp, and Angola strains). Nucleotide variances of >20% between Ravn and other MARV genomes suggest that differing virulence between lineages may accompany this genetic divergence. To date, there exists limited systematic experimental evidence of pathogenic differences between MARV strains. METHODS: Uniformly lethal outbred guinea pig models of MARV-Angola (MARV-Ang) and MARV-Ravn (MARV-Rav) were developed by serial adaptation. Changes in genomic sequence, weight, temperature, histopathologic findings, immunohistochemical findings, hematologic profiles, circulating biochemical enzyme levels, coagulation parameters, viremia levels, cytokine levels, eicanosoid levels, and nitric oxide production were compared between strains. RESULTS: MARV-Rav infection resulted in delayed increases in circulating inflammatory and prothrombotic elements, notably lower viremia levels, less severe histologic alterations, and a delay in mean time to death, compared with MARV-Ang infection. Both strains produced more marked coagulation abnormalities than previously seen in MARV-infected mice or inbred guinea pigs. CONCLUSIONS: Although both strains exhibit great similarity to pathogenic markers of human and nonhuman primate MARV infection, these data highlight several key differences in pathogenicity that may serve to guide the choice of strain and model used for development of vaccines or therapeutics for Marburg hemorrhagic fever.


Asunto(s)
Enfermedad del Virus de Marburg/virología , Marburgvirus/patogenicidad , Virulencia/genética , Angola , Animales , Coagulación Sanguínea/fisiología , Citocinas/metabolismo , Femenino , Ligamiento Genético/genética , Variación Genética/genética , Cobayas , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/genética , Óxido Nítrico/metabolismo , Viremia/metabolismo
10.
J Infect Dis ; 212 Suppl 2: S219-25, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25926685

RESUMEN

Marburg viruses (MARVs) cause highly lethal infections in humans and nonhuman primates. Mice are not generally susceptible to MARV infection; however, if the strain is first adapted to mice through serial passaging, it becomes able to cause disease in this animal. A previous study correlated changes accrued during mouse adaptation in the VP40 gene of a MARV strain known as Ravn virus (RAVV) with an increased capacity to inhibit interferon (IFN) signaling in mouse cell lines. The MARV strain Ci67, which belongs to a different phylogenetic clade than RAVV, has also been adapted to mice and in the process the Ci67 VP40 acquired a different collection of genetic changes than did RAVV VP40. Here, we demonstrate that the mouse-adapted Ci67 VP40 more potently antagonizes IFN-α/ß-induced STAT1 and STAT2 tyrosine phosphorylation, gene expression, and antiviral activity in both mouse and human cell lines, compared with the parental Ci67 VP40. Ci67 VP40 is also demonstrated to target the activation of kinase Jak1. A single change at VP40 residue 79 was found to be sufficient for the increased VP40 IFN antagonism. These data argue that VP40 IFN-antagonist activity plays a key role in MARV pathogenesis in mice.


Asunto(s)
Aminoácidos/metabolismo , Marburgvirus/metabolismo , Marburgvirus/patogenicidad , Proteínas de la Matriz Viral/metabolismo , Animales , Línea Celular , Células HEK293 , Humanos , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Janus Quinasa 1/metabolismo , Enfermedad del Virus de Marburg/metabolismo , Enfermedad del Virus de Marburg/virología , Ratones , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo
11.
J Infect Dis ; 212 Suppl 2: S154-9, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25926686

RESUMEN

BACKGROUND: Marburg virus (MARV) is an emerging zoonotic pathogen that causes hemorrhagic fever. MARV VP24 (mVP24) protein interacts with the host cell protein Kelch-like-ECH-associated protein 1 (Keap1). Keap1 interacts with and promotes the degradation of IκB kinase ß (IKKß), a component of the IκB kinase (IKK) complex that regulates nuclear factor-κB (NF-κB) activity. We studied whether mVP24 could relieve Keap1 repression of the NF-κB pathway. METHODS: Coimmunoprecipitation assays were used to examine the interaction between Keap1 and IKKß in the presence of wild-type mVP24 and mutants of mVP24 defective for binding to Keap1. Western blotting was used to determine levels of IKKß expression in the presence of Keap1 and mVP24. NF-κB promoter-luciferase assays were used to determine the effect of mVP24 on Keap1-induced repression of activity. RESULTS: Expression of wild-type mVP24 disrupted the interaction of IKKß and Keap1, whereas weakly interacting and noninteracting mVP24 mutants did not disrupt the interaction between Keap1 and IKKß. The interaction of mVP24 with Keap1 enhanced IKKß levels in the presence of Keap1. The interaction of mVP24 with Keap1 also relieved Keap1 repression of NF-κB reporter activity. CONCLUSIONS: mVP24 can relieve Keap1 repression of the NF-κB pathway through its interaction with Keap1.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Marburgvirus/metabolismo , FN-kappa B/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Células HEK293 , Humanos , Quinasa I-kappa B/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Enfermedad del Virus de Marburg/metabolismo , Enfermedad del Virus de Marburg/virología , Dominios y Motivos de Interacción de Proteínas/fisiología , Transducción de Señal/fisiología
12.
Cell Rep ; 6(6): 1017-1025, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24630991

RESUMEN

Kelch-like ECH-associated protein 1 (Keap1) is a ubiquitin E3 ligase specificity factor that targets transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) for ubiquitination and degradation. Disrupting Keap1-Nrf2 interaction stabilizes Nrf2, resulting in Nrf2 nuclear accumulation, binding to antioxidant response elements (AREs), and transcription of cytoprotective genes. Marburg virus (MARV) is a zoonotic pathogen that likely uses bats as reservoir hosts. We demonstrate that MARV protein VP24 (mVP24) binds the Kelch domain of either human or bat Keap1. This binding is of high affinity and 1:1 stoichiometry and activates Nrf2. Modeling based on the Zaire ebolavirus (EBOV) VP24 (eVP24) structure identified in mVP24 an acidic loop (K-loop) critical for Keap1 interaction. Transfer of the K-loop to eVP24, which otherwise does not bind Keap1, confers Keap1 binding and Nrf2 activation, and infection by MARV, but not EBOV, activates ARE gene expression. Therefore, MARV targets Keap1 to activate Nrf2-induced cytoprotective responses during infection.


Asunto(s)
Antioxidantes/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Marburgvirus/metabolismo , Proteínas Virales/metabolismo , Animales , Citoprotección , Expresión Génica , Células HEK293 , Humanos , Proteína 1 Asociada A ECH Tipo Kelch , Enfermedad del Virus de Marburg/metabolismo , Enfermedad del Virus de Marburg/virología , Marburgvirus/genética , Modelos Moleculares , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/fisiología , Unión Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato , Transfección , Proteínas Virales/biosíntesis , Proteínas Virales/genética
13.
J Infect Dis ; 209(4): 562-70, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23990568

RESUMEN

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.


Asunto(s)
Lípidos/administración & dosificación , Enfermedad del Virus de Marburg/tratamiento farmacológico , Enfermedad del Virus de Marburg/prevención & control , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/análisis , Citocinas/sangre , Portadores de Fármacos/química , Femenino , Genes Virales , Cobayas , Lípidos/química , Hígado/química , Enfermedad del Virus de Marburg/genética , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/efectos de los fármacos , Marburgvirus/genética , Ratones , Ratones Endogámicos ICR , ARN Interferente Pequeño/química , Proteínas de Unión al ARN , Análisis de Supervivencia , Carga Viral
14.
Viruses ; 4(10): 2400-16, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23202470

RESUMEN

Baboons are susceptible to natural Ebola virus (EBOV) infection and share 96% genetic homology with humans. Despite these characteristics, baboons have rarely been utilized as experimental models of human EBOV infection to evaluate the efficacy of prophylactics and therapeutics in the United States. This review will summarize what is known about the pathogenesis of EBOV infection in baboons compared to EBOV infection in humans and other Old World nonhuman primates. In addition, we will discuss how closely the baboon model recapitulates human EBOV infection. We will also review some of the housing requirements and behavioral attributes of baboons compared to other Old World nonhuman primates. Due to the lack of data available on the pathogenesis of Marburg virus (MARV) infection in baboons, discussion of the pathogenesis of MARV infection in baboons will be limited.


Asunto(s)
Modelos Animales de Enfermedad , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/patología , Enfermedad del Virus de Marburg/patología , Papio/virología , Animales , Secuencia de Bases , Factores de Coagulación Sanguínea/metabolismo , Fibrina/metabolismo , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/virología , Humanos , Enfermedades Linfáticas/patología , Enfermedades Linfáticas/virología , Enfermedad del Virus de Marburg/metabolismo , Enfermedad del Virus de Marburg/virología , Marburgvirus/patogenicidad , Necrosis/patología , Necrosis/virología , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Trombocitopenia/patología , Trombocitopenia/virología
15.
Nature ; 477(7364): 340-3, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21866103

RESUMEN

Infections by the Ebola and Marburg filoviruses cause a rapidly fatal haemorrhagic fever in humans for which no approved antivirals are available. Filovirus entry is mediated by the viral spike glycoprotein (GP), which attaches viral particles to the cell surface, delivers them to endosomes and catalyses fusion between viral and endosomal membranes. Additional host factors in the endosomal compartment are probably required for viral membrane fusion; however, despite considerable efforts, these critical host factors have defied molecular identification. Here we describe a genome-wide haploid genetic screen in human cells to identify host factors required for Ebola virus entry. Our screen uncovered 67 mutations disrupting all six members of the homotypic fusion and vacuole protein-sorting (HOPS) multisubunit tethering complex, which is involved in the fusion of endosomes to lysosomes, and 39 independent mutations that disrupt the endo/lysosomal cholesterol transporter protein Niemann-Pick C1 (NPC1). Cells defective for the HOPS complex or NPC1 function, including primary fibroblasts derived from human Niemann-Pick type C1 disease patients, are resistant to infection by Ebola virus and Marburg virus, but remain fully susceptible to a suite of unrelated viruses. We show that membrane fusion mediated by filovirus glycoproteins and viral escape from the vesicular compartment require the NPC1 protein, independent of its known function in cholesterol transport. Our findings uncover unique features of the entry pathway used by filoviruses and indicate potential antiviral strategies to combat these deadly agents.


Asunto(s)
Proteínas Portadoras/metabolismo , Colesterol/metabolismo , Ebolavirus/fisiología , Glicoproteínas de Membrana/metabolismo , Internalización del Virus , Animales , Transporte Biológico , Proteínas Portadoras/genética , Línea Celular , Endosomas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/virología , Genoma Humano/genética , Glicoproteínas/metabolismo , Haploidia , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lisosomas/metabolismo , Enfermedad del Virus de Marburg/tratamiento farmacológico , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/fisiología , Fusión de Membrana/genética , Fusión de Membrana/fisiología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Complejos Multiproteicos/química , Complejos Multiproteicos/deficiencia , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación/genética , Proteína Niemann-Pick C1 , Enfermedades de Niemann-Pick/patología , Enfermedades de Niemann-Pick/virología , Receptores Virales/metabolismo , Proteínas de Transporte Vesicular , Proteínas Virales de Fusión/metabolismo
16.
J Pharm Sci ; 100(12): 5156-73, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21858822

RESUMEN

The filoviruses, Ebola virus and Marburg virus, cause severe hemorrhagic fever with up to 90% human mortality. Virus-like particles of EBOV (eVLPs) and MARV (mVLPs) are attractive vaccine candidates. For the development of stable vaccines, the conformational stability of these two enveloped VLPs produced in insect cells was characterized by various spectroscopic techniques over a wide pH and temperature range. Temperature-induced aggregation of the VLPs at various pH values was monitored by light scattering. Temperature/pH empirical phase diagrams (EPDs) of the two VLPs were constructed to summarize the large volume of data generated. The EPDs show that both VLPs lose their conformational integrity above about 50°C-60°C, depending on solution pH. The VLPs were maximally thermal stable in solution at pH 7-8, with a significant reduction in stability at pH 5 and 6. They were much less stable in solution at pH 3-4 due to increased susceptibility of the VLPs to aggregation. The characterization data and conformational stability profiles from these studies provide a basis for selection of optimized solution conditions for further vaccine formulation and long-term stability studies of eVLPs and mVLPs.


Asunto(s)
Ebolavirus/metabolismo , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/metabolismo , Virión/química , Animales , Anticuerpos Antivirales , Baculoviridae/genética , Baculoviridae/metabolismo , Línea Celular , Vacunas contra el Virus del Ébola/análisis , Vacunas contra el Virus del Ébola/química , Vacunas contra el Virus del Ébola/metabolismo , Ebolavirus/química , Ebolavirus/genética , Ebolavirus/inmunología , Vectores Genéticos , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Insectos , Enfermedad del Virus de Marburg/inmunología , Marburgvirus/química , Marburgvirus/genética , Marburgvirus/inmunología , Conformación Molecular , Temperatura , Vacunas de Partículas Similares a Virus/análisis , Vacunas de Partículas Similares a Virus/química , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/metabolismo
17.
PLoS Pathog ; 6(1): e1000721, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20084112

RESUMEN

Previous studies have demonstrated that Marburg viruses (MARV) and Ebola viruses (EBOV) inhibit interferon (IFN)-alpha/beta signaling but utilize different mechanisms. EBOV inhibits IFN signaling via its VP24 protein which blocks the nuclear accumulation of tyrosine phosphorylated STAT1. In contrast, MARV infection inhibits IFNalpha/beta induced tyrosine phosphorylation of STAT1 and STAT2. MARV infection is now demonstrated to inhibit not only IFNalpha/beta but also IFNgamma-induced STAT phosphorylation and to inhibit the IFNalpha/beta and IFNgamma-induced tyrosine phosphorylation of upstream Janus (Jak) family kinases. Surprisingly, the MARV matrix protein VP40, not the MARV VP24 protein, has been identified to antagonize Jak and STAT tyrosine phosphorylation, to inhibit IFNalpha/beta or IFNgamma-induced gene expression and to inhibit the induction of an antiviral state by IFNalpha/beta. Global loss of STAT and Jak tyrosine phosphorylation in response to both IFNalpha/beta and IFNgamma is reminiscent of the phenotype seen in Jak1-null cells. Consistent with this model, MARV infection and MARV VP40 expression also inhibit the Jak1-dependent, IL-6-induced tyrosine phosphorylation of STAT1 and STAT3. Finally, expression of MARV VP40 is able to prevent the tyrosine phosphorylation of Jak1, STAT1, STAT2 or STAT3 which occurs following over-expression of the Jak1 kinase. In contrast, MARV VP40 does not detectably inhibit the tyrosine phosphorylation of STAT2 or Tyk2 when Tyk2 is over-expressed. Mutation of the VP40 late domain, essential for efficient VP40 budding, has no detectable impact on inhibition of IFN signaling. This study shows that MARV inhibits IFN signaling by a mechanism different from that employed by the related EBOV. It identifies a novel function for the MARV VP40 protein and suggests that MARV may globally inhibit Jak1-dependent cytokine signaling.


Asunto(s)
Evasión Inmune/inmunología , Interferones/inmunología , Marburgvirus/inmunología , Transducción de Señal/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Western Blotting , Línea Celular , Citocinas/inmunología , Ebolavirus/inmunología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Fiebre Hemorrágica Ebola/inmunología , Humanos , Janus Quinasa 1/inmunología , Janus Quinasa 1/metabolismo , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/metabolismo , Reacción en Cadena de la Polimerasa , Transfección
18.
J Virol ; 80(19): 9659-66, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16973570

RESUMEN

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.


Asunto(s)
Enfermedad del Virus de Marburg/inmunología , Marburgvirus/clasificación , Marburgvirus/inmunología , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos/inmunología , Reacciones Cruzadas/inmunología , Macaca fascicularis , Enfermedad del Virus de Marburg/genética , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/genética , Marburgvirus/metabolismo , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Viremia/sangre , Viremia/inmunología , Viremia/virología
19.
J Virol ; 80(14): 7235-44, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16809329

RESUMEN

Marburg virus (MARV) and Ebola virus (EBOV), members of the viral family Filoviridae, cause fatal hemorrhagic fevers in humans and nonhuman primates. High viral burden is coincident with inadequate adaptive immune responses and robust inflammatory responses, and virus-mediated dysregulation of early host defenses has been proposed. Recently, a novel class of innate receptors called the triggering receptors expressed in myeloid cells (TREM) has been discovered and shown to play an important role in innate inflammatory responses and sepsis. Here, we report that MARV and EBOV activate TREM-1 on human neutrophils, resulting in DAP12 phosphorylation, TREM-1 shedding, mobilization of intracellular calcium, secretion of proinflammatory cytokines, and phenotypic changes. A peptide specific to TREM-1 diminished the release of tumor necrosis factor alpha by filovirus-activated human neutrophils in vitro, and a soluble recombinant TREM-1 competitively inhibited the loss of cell surface TREM-1 that otherwise occurred on neutrophils exposed to filoviruses. These data imply direct activation of TREM-1 by filoviruses and also indicate that neutrophils may play a prominent role in the immune and inflammatory responses to filovirus infections.


Asunto(s)
Ebolavirus/inmunología , Marburgvirus/inmunología , Glicoproteínas de Membrana/inmunología , Activación Neutrófila/inmunología , Neutrófilos/metabolismo , Neutrófilos/virología , Receptores Inmunológicos/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Chlorocebus aethiops , Ebolavirus/metabolismo , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/mortalidad , Humanos , Inmunidad Innata/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/mortalidad , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/metabolismo , Enfermedad del Virus de Marburg/mortalidad , Marburgvirus/metabolismo , Glicoproteínas de Membrana/biosíntesis , Proteínas de la Membrana , Fosforilación , Procesamiento Proteico-Postraduccional/inmunología , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/metabolismo , Sepsis/inmunología , Sepsis/metabolismo , Sepsis/mortalidad , Receptor Activador Expresado en Células Mieloides 1 , Células Vero
20.
Biochemistry ; 38(19): 6157-63, 1999 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-10320343

RESUMEN

Myelin basic protein (MBP) exists in a population of isoforms and isomers. The 18.5 kDa MBP-C1, the main human adult isoform, has 170 residues and is relatively unmodified, whereas the same isoform can be citrullinated on six arginine residues to create the MBP-C8 (MBP Cit6) isomer. MBP Cit6 dominates in MS brain, accounting for 45% rather than 25% of the population of MBP isomers. In the fulminant form of MS, known as Marburg's Disease, 18 of the 19 arginines in MBP are citrullinated (MBP Cit18). Citrullination of MBP could lead to instability of myelin or limited remyelination. In this investigation, the susceptibilities to degradation by cathepsin D of MBP Cit6 and MBP-C1, both from normal and MS brain tissue, and Marburg MBP Cit18 were compared. The pattern of digestion was similar, and no differences of corresponding isomers in normal and MS brain were noted. However, normal MBP Cit6 was degraded 10-fold more rapidly than MBP-C1, and MBP Cit18 was degraded even more rapidly. MBP peptide 45-89 was preserved regardless of isomer type or source. Its generation was directly related to the citrulline content of the MBP substrate being 4 times faster in normal MBP Cit6 and 35 times faster in Marburg MBP Cit18 than in normal MBP-C1. Peptide 45-89 from a citrullinated MBP exhibited more deamidation, and, regardless of source, showed an alpha-helix structure in a lipid mimetic environment. We postulate that the generation of MBP peptides, including those that are dominant and encephalitogenic, is directly related to deimination of arginine to citrulline in MBP.


Asunto(s)
Citrulina/metabolismo , Epítopos Inmunodominantes/metabolismo , Proteína Básica de Mielina/metabolismo , Encéfalo/metabolismo , Catepsina D/metabolismo , Humanos , Enfermedad del Virus de Marburg/metabolismo , Proteína Básica de Mielina/inmunología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...