Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 366
Filtrar
1.
Vaccines (Basel) ; 12(9)2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39340043

RESUMO

Marburg hemorrhagic fever (MHF) is a fatal infectious disease caused by Marburg virus (MARV) infection, and MARV has been identified as a priority pathogen for vaccine development by the WHO. The glycoprotein (GP) of MARV mediates viral adhesion and invasion of host cells and therefore can be used as an effective target for vaccine development. Moreover, DNA vaccines have unique advantages, such as simple construction processes, low production costs, and few adverse reactions, but their immunogenicity may decrease due to the poor absorption rate of plasmids. Lysosome-associated membrane protein 1 (LAMP1) can direct antigens to lysosomes and endosomes and has great potential for improving the immunogenicity of nucleic acid vaccines. Therefore, we constructed a DNA vaccine based on a codon-optimized MARV GP (ID MF939097.1) fused with LAMP1 and explored the effect of a LAMP targeting strategy on improving the immunogenicity of the MARV DNA vaccine. ELISA, ELISpot, and flow cytometry revealed that the introduction of LAMP1 into the MARV DNA candidate vaccine improved the humoral and cellular immune response, enhanced the secretion of cytokines, and established long-term immune protection. Transcriptome analysis revealed that the LAMP targeting strategy significantly enriched antigen processing and presentation-related pathways, especially the MHC class II-related pathway, in the candidate vaccine. Our study broadens the strategic vision for enhanced DNA vaccine design and provides a promising candidate vaccine for MHF prevention.

2.
J Cell Mol Med ; 28(18): e70116, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39340487

RESUMO

Global impact of viral diseases specially Monkeypox (mpox) and Marburg virus, emphasizing the urgent need for effective drug interventions. Oxymatrine is an alkaloid which has been selected and modified using various functional groups to enhance its efficacy. The modifications were evaluated using various computatioanal analysis such as pass prediction, molecular docking, ADMET, and molecular dynamic simulation. Mpox and Marburg virus were chosen as target diseases based on their maximum pass prediction spectrum against viral disease. After that, molecular docking, dynamic simulation, DFT, calculation and ADMET prediction were determined. The main objective of this study was to enhance the efficacy of oxymatrine derivatives through functional group modifications and computational analyses to develop effective drug candidates against mpox and Marburg viruses. The calculated binding affinities indicated strong interactions against both mpox virus and Marburg virus. After that, the molecular dynamic simulation was conducted at 100 ns, which confirmed the stability of the binding interactions between the modified oxymatrine derivatives and target proteins. Then, the modified oxymatrine derivatives conducted theoretical ADMET profiling, which demonstrated their potential for effective drug development. Moreover, HOMO-LUMO calculation was performed to understand the chemical reactivity and physicochemical properties of compounds. This computational analysis indicated that modified oxymatrine derivatives for the treatment of mpox and Marburg virus suggested effective drug candidates based on their binding affinity, drug-like properties, stability and chemical reactivity. However, further experimental validation is necessary to confirm their clinical value and efficacy as therapeutic candidates.


Assuntos
Alcaloides , Antivirais , Desenho de Fármacos , Marburgvirus , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Quinolizinas , Alcaloides/química , Alcaloides/farmacologia , Quinolizinas/química , Quinolizinas/farmacologia , Antivirais/farmacologia , Antivirais/química , Marburgvirus/efeitos dos fármacos , Animais , Humanos , Matrinas
3.
Int J Mol Sci ; 25(18)2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39337389

RESUMO

The life cycle of Ebola and Marburg viruses includes a step of the virion envelope fusion with the cell membrane. Here, we analyzed whether the fusion of liposome membranes under the action of fragments of fusion peptides of Ebola and Marburg viruses depends on the composition of lipid vesicles. A fluorescence assay and electron microscopy were used to quantify the fusogenic activity of the virus fusion peptides and to identify the lipid determinants affecting membrane merging. Differential scanning calorimetry of lipid phase transitions revealed alterations in the physical properties of the lipid matrix produced by virus fusion peptides. Additionally, we found that plant polyphenols, quercetin, and myricetin inhibited vesicle fusion induced by the Marburg virus fusion peptide.


Assuntos
Ebolavirus , Flavonoides , Marburgvirus , Ebolavirus/efeitos dos fármacos , Marburgvirus/efeitos dos fármacos , Marburgvirus/química , Flavonoides/química , Flavonoides/farmacologia , Fusão de Membrana/efeitos dos fármacos , Lipossomos/química , Quercetina/química , Quercetina/farmacologia , Internalização do Vírus/efeitos dos fármacos , Doença pelo Vírus Ebola/virologia , Polifenóis/química , Polifenóis/farmacologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Humanos , Membrana Celular/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia
4.
Viruses ; 16(9)2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39339831

RESUMO

High-consequence pathogens such as the Ebola, Marburg, and Lassa viruses are handled in maximum-containment biosafety level 4 (BSL-4) laboratories. Genetic material is often isolated from such viruses and subsequently removed from BSL-4 laboratories for a multitude of downstream analyses using readily accessible technologies and equipment available at lower-biosafety level laboratories. However, it is essential to ensure that these materials are free of viable viruses before removal from BSL-4 laboratories to guarantee sample safety. This study details the in-house procedure used for validating the inactivation of Ebola, Marburg, and Lassa virus cultures after incubation with AVL lysis buffer (Qiagen) and ethanol. This study's findings show that no viable virus was detectable when high-titer cultures of Ebola, Marburg, and Lassa viruses were incubated with AVL lysis buffer for 10 min, followed by an equal volume of 95% ethanol for 3 min, using a method with a sensitivity of ≤0.8 log10 TCID50 as the limit of detection.


Assuntos
Ebolavirus , Etanol , Vírus Lassa , Marburgvirus , Inativação de Vírus , Vírus Lassa/efeitos dos fármacos , Marburgvirus/efeitos dos fármacos , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Etanol/farmacologia , Inativação de Vírus/efeitos dos fármacos , Animais , Humanos , Contenção de Riscos Biológicos/métodos , Febre Lassa/virologia , Cultura de Vírus/métodos , Chlorocebus aethiops , Células Vero
5.
Sci One Health ; 3: 100076, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39309209

RESUMO

The Marburg virus (MARV), belonging to the Filoviridae family, poses a significant global health threat, emphasizing the urgency to develop Marburg virus-like particle (VLP) vaccines for outbreak mitigation. The virus's menacing traits accentuate the need for such vaccines, which can be addressed by VLPs that mimic its structure safely, potentially overcoming past limitations. Early Marburg vaccine endeavors and their challenges are examined in the historical perspectives section, followed by an exploration of VLPs as transformative tools, capable of eliciting immune responses without conventional risks. Noteworthy milestones and achievements in Marburg VLP vaccine development, seen through preclinical and clinical trials, indicate potential cross-protection. Ongoing challenges, encompassing durability, strain diversity, and equitable distribution, are addressed, with proposed innovations like novel adjuvant, mRNA technology, and structure-based design poised to enhance Marburg VLP vaccines. This review highlights the transformative potential of Marburg VLPs in countering the virus, showcasing global collaboration, regulatory roles, and health equity for a safer future through the harmonious interplay of science, regulation, and global efforts.

6.
J Virol ; 98(9): e0104724, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39194238

RESUMO

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


Assuntos
Marburgvirus , Proteína Fosfatase 2 , Transcrição Gênica , Marburgvirus/fisiologia , Marburgvirus/genética , Marburgvirus/metabolismo , Proteína Fosfatase 2/metabolismo , Proteína Fosfatase 2/genética , Humanos , Fosforilação , Replicação Viral , Células HEK293 , Animais , Interações Hospedeiro-Patógeno , Proteínas Virais/metabolismo , Proteínas Virais/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Doença do Vírus de Marburg/virologia , Doença do Vírus de Marburg/metabolismo , Ligação Proteica , Linhagem Celular
7.
Viruses ; 16(8)2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39205155

RESUMO

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


Assuntos
Macaca fascicularis , Doença do Vírus de Marburg , Marburgvirus , Vesiculovirus , Vacinas Virais , Animais , Doença do Vírus de Marburg/prevenção & controle , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Marburgvirus/imunologia , Marburgvirus/genética , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vesiculovirus/genética , Vesiculovirus/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Modelos Animais de Doenças , Vacinação , Masculino , Feminino , Eficácia de Vacinas , Vetores Genéticos , Imunogenicidade da Vacina
8.
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
9.
Viruses ; 16(8)2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39205171

RESUMO

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


Assuntos
Quirópteros , Doença do Vírus de Marburg , Marburgvirus , Sifonápteros , Animais , Quirópteros/virologia , Marburgvirus/genética , Marburgvirus/fisiologia , Sifonápteros/virologia , Doença do Vírus de Marburg/virologia , Doença do Vírus de Marburg/transmissão , Reservatórios de Doenças/virologia , Viremia , Infestações por Pulgas/veterinária , Infestações por Pulgas/transmissão , Infestações por Pulgas/virologia , RNA Viral/genética , Egito
10.
Health Sci Rep ; 7(8): e2270, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39100711

RESUMO

Background and Aim: Marburg virus (MARV) is a highly virulent virus of animal origin and the cause of a lethal infection (known as Marburg virus disease [MVD]) with a case-fatality ratio ranging from 24% to 90%. While the potential nonzoonotic routes of virus spread are plausible, the risk is not yet fully determined. Here, we described the ways by which MARV spreads within the human population focusing mainly on the potential of sexual transmission. In addition, we addressed some measures that should be taken to minimize the risk of sexual spread of the virus and proposed a future research agenda on the risk of sexual transmission. Methods: For this perspective, we searched four electronic databases (i.e., PubMed, Scopus, Web of Science, and Google Scholar) and included the most relevant studies published since the first identification of the virus in 1967. We used "Marburg virus," "Marburg virus disease," "Seminal fluid," "Sexually-transmitted virus," "Sexual transmission," and "Emerging infectious disease" as keywords. Results: MARV is transmitted to humans via both direct and indirect contact with infected animals (most importantly bats) and individuals who have recently been diagnosed with or died of the disease. The virus transmission through sexual contact has been previously suspected (exclusively from men to their sexual partners). Studies suggest that this virus persists predominantly in testicular Sertoli cells within seminiferous tubules over a relatively long period and is released through seminal fluid (in some reports >200 days post onset of infection) both could potentially threaten sexual health. In addition to men, women could theoretically, although less probably contribute to the sexual transmission of the disease. Conclusion: MVD, however, rarely, could be passed through sex, and men appear to be the main carriers in this regard. Taking preventive countermeasures and practicing safe sex are recommended to reduce the risk of interhuman transmission.

11.
Methods Mol Biol ; 2829: 277-286, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38951344

RESUMO

Quantitative immunoassays, such as the traditional enzyme-linked immunosorbent assay (ELISA), are used to determine concentrations of an antigen in a matrix of unknown antigen concentration. Magnetic immunoassays, such as the Luminex xMAP technology, allow for the simultaneous detection of multiple analytes and offer heightened sensitivity, specificity, low sample volume requirements, and high-throughput capabilities. Here, we describe a quantitative immunoassay using the Luminex MAGPIX® System to determine the antigen concentration from liquid samples with unknown concentrations. In detail, we describe a newly developed assay for determining production yields of Drosophila S2-produced Marburg virus (MARV) glycoprotein in insect-cell-culture-derived supernatant. The potential applications of this assay could extend to the quantification of viral antigens in fluids derived from both in vitro and in vivo models infected with live MARV, thereby providing additional applications for virological research.


Assuntos
Antígenos Virais , Microesferas , Animais , Imunoensaio/métodos , Antígenos Virais/imunologia , Antígenos Virais/análise , Marburgvirus/imunologia , Marburgvirus/isolamento & purificação , Drosophila , Técnicas de Cultura de Células/métodos , Linhagem Celular , Ensaio de Imunoadsorção Enzimática/métodos
13.
BMC Biotechnol ; 24(1): 45, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38970027

RESUMO

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


Assuntos
Biologia Computacional , Doença do Vírus de Marburg , Marburgvirus , Vacinas Virais , Marburgvirus/imunologia , Doença do Vírus de Marburg/prevenção & controle , Doença do Vírus de Marburg/imunologia , Vacinas Virais/imunologia , Biologia Computacional/métodos , Animais , Humanos , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/genética , Epitopos/imunologia , Epitopos/genética , Epitopos/química , Escherichia coli/genética , Escherichia coli/metabolismo , Imunoinformática
14.
Microbiol Spectr ; 12(9): e0026924, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39078193

RESUMO

Filoviruses produce viral particles with characteristic filamentous morphology. The major viral matrix protein, VP40, is trafficked to the plasma membrane and promotes viral particle formation and subsequent viral egress. In the present study, we assessed the role of the small GTPase Rab11-mediated endocytic pathway in Marburg virus (MARV) particle formation and budding. Although Rab11 was predominantly localized in the perinuclear region, it exhibited a more diffuse distribution in the cytoplasm of cells transiently expressing MARV VP40. Rab11 was incorporated into MARV-like particles. Expression of the dominant-negative form of Rab11 and knockdown of Rab11 decreased the amount of VP40 fractions in the cell periphery. Moreover, downregulation of Rab11 moderately reduced the release of MARV-like particles and authentic MARV. We further demonstrated that VP40 induces the distribution of the microtubule network toward the cell periphery, which was partly associated with Rab11. Depolymerization of microtubules reduced the accumulation of VP40 in the cell periphery along with viral particle formation. VP40 physically interacted with α-tubulin, a major component of microtubules, but not with Rab11. Taken together, these results suggested that VP40 partly interacts with microtubules and facilitates their distribution toward the cell periphery, leading to the trafficking of transiently tethering Rab11-positive vesicles toward the cell surface. As we previously demonstrated the role of Rab11 in the formation of Ebola virus particles, the results here suggest that filoviruses in general exploit the vesicle-trafficking machinery for proper virus-particle formation and subsequent egress. These pathways may be a potential target for the development of pan-filovirus therapeutics.IMPORTANCEFiloviruses, including Marburg and Ebola viruses, produce distinct filamentous viral particles. Although it is well known that the major viral matrix protein of these viruses, VP40, is trafficked to the cell surface and promotes viral particle production, details regarding the associated molecular mechanisms remain unclear. To address this knowledge gap, we investigated the role of the small GTPase Rab11-mediated endocytic pathway in this process. Our findings revealed that Marburg virus exploits the Rab11-mediated vesicle-trafficking pathway for the release of virus-like particles and authentic virions in a microtubule network-dependent manner. Previous findings demonstrated that Rab11 is also involved in Ebola virus-particle production. Taken together, these data suggest that filoviruses, in general, may hijack the microtubule-dependent vesicle-trafficking machinery for productive replication. Therefore, this pathway presents as a potential target for the development of pan-filovirus therapeutics.


Assuntos
Endocitose , Marburgvirus , Proteínas da Matriz Viral , Vírion , Proteínas rab de Ligação ao GTP , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab de Ligação ao GTP/genética , Marburgvirus/fisiologia , Marburgvirus/genética , Marburgvirus/metabolismo , Humanos , Proteínas da Matriz Viral/metabolismo , Proteínas da Matriz Viral/genética , Animais , Vírion/metabolismo , Microtúbulos/metabolismo , Microtúbulos/virologia , Liberação de Vírus , Linhagem Celular , Células HEK293 , Replicação Viral
15.
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
16.
Stud Hist Philos Sci ; 106: 155-164, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38986223

RESUMO

A trained physicist, Kurd Lasswitz (1848-1910) is best known as a novelist, the father of modern German science fiction, and as a historian of science, the initiator of the modern historiography of atomism. In the late 19th century, Lasswitz engaged in an intense dialogue with the emerging Marburg school of neo-Kantianism, contributing to shaping most of its defining tenets. By the end of the decade, this research had grown into a two-volume Geschichte der Atomistik (1890), which remains the most successful example of neo-Kantian historiography of science. Lasswitz combined attention to historical detail with the search for the intellectual tools (Denkmittel) without which the 'fact of science' would be impossible. In particular, Lasswitz regarded Huygens' kinetic atomism as a historical model of a successful scientific theory, shaped by the interplay of two conceptual tools: (a) substantiality, the requirement for identity of the subject of motion through time, which found its scientific expression in the extensive atom; (b) variability, the intensive tendency to continue in an instant, which found its conceptual fixation in the notion of 'differential'. By raising the problem of individuality in physics, Lasswitz offers a unique perspective on the utilization of the history of science in 19th-century neo-Kantian thought.


Assuntos
Historiografia , História do Século XIX , História do Século XX , Alemanha , Física/história , Ciência/história
17.
Eur J Med Chem ; 275: 116596, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-38889610

RESUMO

The increasing frequency of filovirus outbreaks in African countries has led to a pressing need for the development of effective antifilovirus agents. In continuation of our previous research on the antifilovirus activity of monoterpenoid derivatives, we synthesized a series of (+)-fenchol and (-)-isopinocampheol derivatives by varying the type of heterocycle and linker length. Derivatives with an N-alkylpiperazine cycle proved to be the most potent antiviral compounds, with half-maximal inhibitory concentration (IC50) 1.4-20 µÐœ against Lenti-EboV-GP infection and 11.3-47 µÐœ against Lenti-MarV-GP infection. Mechanism-of-action experiments revealed that the compounds may exert their action by binding to surface glycoproteins (GPs). It was demonstrated that the binding of the synthesized compounds to the Marburg virus GP is less efficient as compared to the Ebola virus GP. Furthermore, it was shown that the compounds possess lysosomotropic properties. Thus, the antiviral activity may be due to dual effects. This study offers new antiviral agents that are worthy of further exploration.


Assuntos
Antivirais , Antivirais/farmacologia , Antivirais/química , Antivirais/síntese química , Humanos , Internalização do Vírus/efeitos dos fármacos , Relação Estrutura-Atividade , Ebolavirus/efeitos dos fármacos , Estrutura Molecular , Relação Dose-Resposta a Droga , Animais , Testes de Sensibilidade Microbiana , Chlorocebus aethiops , Marburgvirus/efeitos dos fármacos
18.
BMC Infect Dis ; 24(1): 628, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38914946

RESUMO

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


Assuntos
Surtos de Doenças , Doença do Vírus de Marburg , Tanzânia/epidemiologia , Humanos , Surtos de Doenças/prevenção & controle , Doença do Vírus de Marburg/epidemiologia , Doença do Vírus de Marburg/prevenção & controle , Controle de Infecções/métodos , Animais , Países em Desenvolvimento
19.
J Virol ; 98(7): e0015524, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38832790

RESUMO

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


Assuntos
Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Reações Cruzadas , Macaca mulatta , Doença do Vírus de Marburg , Marburgvirus , Animais , Marburgvirus/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Monoclonais/imunologia , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/prevenção & controle , Reações Cruzadas/imunologia , Glicoproteínas/imunologia , Proteínas do Envelope Viral/imunologia , Imunização , Humanos , Ebolavirus/imunologia , Antígenos Virais/imunologia
20.
Front Microbiol ; 15: 1387628, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38725678

RESUMO

Marburg virus disease (MVD) presents a significant global health threat, lacking effective antivirals and with current supportive care offering limited therapeutic options. This mini review explores the emerging landscape of novel antiviral strategies against MVD, focusing on promising therapeutics currently in the development pipeline. We delve into direct-acting antiviral approaches, including small molecule inhibitors targeting viral entry, replication, and assembly, alongside nucleic acid antisense and RNA interference strategies. Host-targeting antivirals are also considered, encompassing immune modulators like interferons and cytokine/chemokine modulators, broad-spectrum antivirals, and convalescent plasma and antibody-based therapies. The paper then examines preclinical and clinical development for the novel therapeutics, highlighting in vitro and in vivo models for antiviral evaluation, safety and efficacy assessments, and the critical stages of clinical trials. Recognizing the challenges of drug resistance and viral escape, the mini review underscores the potential of combination therapy strategies and emphasizes the need for rapid diagnostic tools to optimize treatment initiation. Finally, we discuss the importance of public health preparedness and equitable access to these promising therapeutics in achieving effective MVD control and global health security. This mini review presents a comprehensive overview of the burgeoning field of MVD antivirals, highlighting the potential of these novel approaches to reshape the future of MVD treatment and prevention.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA