Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 377
Filtrar
1.
J Virol ; : e0098624, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39230304

RESUMEN

Nipah virus (NiV) is a highly pathogenic paramyxovirus causing frequently lethal encephalitis in humans. The NiV genome is encapsidated by the nucleocapsid (N) protein. RNA synthesis is mediated by the viral RNA-dependent RNA polymerase (RdRP), consisting of the polymerase (L) protein complexed with the homo-tetrameric phosphoprotein (P). The advance of the polymerase along its template requires iterative dissolution and reformation of transient interactions between P and N protomers in a highly regulated process that remains poorly understood. This study applied functional and biochemical NiV polymerase assays to the problem. We mapped three distinct protein interfaces on the C-terminal P-X domain (P-XD), which form a triangular prism and engage L, the C-terminal N tail, and the globular N core, respectively. Transcomplementation assays using NiV L and N-tail binding-deficient mutants revealed that only one XD of a P tetramer binds to L, whereas three must be available for N-binding for efficient polymerase activity. The dissolution of the N-tail complex with P-XD was coordinated by a transient interaction between N-core and the α-1/2 face of this XD but not unoccupied XDs of the P tetramer, creating a timer for coordinated polymerase advance. IMPORTANCE: Mononegaviruses comprise major human pathogens such as the Ebola virus, rabies virus, respiratory syncytial virus, measles virus, and Nipah virus (NiV). For replication and transcription, their polymerase complexes must negotiate a protein-encapsidated RNA genome, which requires the highly coordinated continuous formation and resolution of protein-protein interfaces as the polymerase advances along the template. The viral P protein assumes a central role in this process, but the molecular mechanism of ensuring polymerase mobility is poorly understood. Studying NiV polymerase complexes, we applied functional and biochemical assays to map three distinct interfaces in the NiV P XD and identified transient interactions between XD and the nucleocapsid core as instrumental in coordinating polymerase advance. These results define a conserved molecular principle regulating paramyxovirus polymerase dynamics and illuminate a promising druggable target for the structure-guided development of broad-spectrum polymerase inhibitors.

2.
J Virol ; : e0080924, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39177356

RESUMEN

Small hydrophobic (SH) proteins are a class of viral accessory proteins expressed by many members of the negative-stranded RNA viral families Paramyxoviridae and Pneumoviridae. Identified SH proteins are type I or II transmembrane (TM) proteins with a single-pass TM domain. Little is known about the functions of SH proteins; however, several possess viroporin activity, enhancing membrane permeability of infected cells or those expressing SH protein. Moreover, several SH proteins inhibit apoptosis and immune signaling pathways within infected cells, including TNF and interferon signaling, or activate inflammasomes. SH proteins are generally nonessential for viral replication in vitro, but loss of SH is often associated with reduced replication in vivo, suggesting a role in enhancing viral replication or evading host immunity. Analogous proteins are expressed by a variety of pathogens of public health importance; thus, understanding the functional importance and mechanisms of SH proteins provides insight into the pathogenesis and replication of negative-sense RNA viruses.

3.
Antimicrob Agents Chemother ; : e0080024, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39162479

RESUMEN

Small-molecule antivirals can be used as chemical probes to stabilize transitory conformational stages of viral target proteins, facilitating structural analyses. Here, we evaluate allosteric pneumo- and paramyxovirus polymerase inhibitors that have the potential to serve as chemical probes and aid the structural characterization of short-lived intermediate conformations of the polymerase complex. Of multiple inhibitor classes evaluated, we discuss in-depth distinct scaffolds that were selected based on well-understood structure-activity relationships, insight into resistance profiles, biochemical characterization of the mechanism of action, and photoaffinity-based target mapping. Each class is thought to block structural rearrangements of polymerase domains albeit target sites and docking poses are distinct. This review highlights validated druggable targets in the paramyxo- and pneumovirus polymerase proteins and discusses discrete structural stages of the polymerase complexes required for bioactivity.

4.
Animals (Basel) ; 14(16)2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39199956

RESUMEN

Newcastle disease (ND) is a highly contagious viral disease caused by the paramyxovirus, which is a single-stranded ribonucleic acid (RNA) virus. This study was conducted to investigate ND outbreaks in 10 vaccinated or non-vaccinated broiler farms, collectively housing 9840 birds of various ages in the Chitwan and Nawalpur districts of Nepal from July to December 2021. Clinically, the affected birds exhibited symptoms such as limb paralysis, greenish diarrhea (seven out of ten flocks), torticollis (two out of ten flocks), inappetence, and drowsiness (ten out of ten flocks). Birds that succumbed during the clinical course underwent a necropsy for gross pathology and samples were collected for the histopathology and molecular diagnosis. The gross and microscopic examination revealed hemorrhages in the proventriculus, erosions and ulcers in the small intestine, congestion, as well as sero-mucosal hemorrhages in the trachea of affected birds, which are typical of ND. Rapid test kits further confirmed the presence of the ND virus antigen while excluding the avian influenza virus. Furthermore, M gene-based real time polymerase chain reaction (RT-PCR) was performed in the pooled samples from the affected birds and the presence of a velogenic strain of the ND virus was identified. The phylogenetic analysis of the RT-PCR positive strain based on the partial F gene nucleotide sequence revealed these strains as genotype VII.2 (formerly VIIi). The findings highlight the occurrence of clinical ND outbreaks in farms despite adherence to recommended vaccination protocols in broiler flocks, underscoring the need for a regular comprehensive investigation involving in-depth examinations of available vaccines and genetic analyses.

5.
Euro Surveill ; 29(28)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38994600

RESUMEN

We investigated a variant of measles virus that encodes three mismatches to the reverse priming site for a widely used diagnostic real-time RT-PCR assay; reduction of sensitivity was hypothesised. We examined performance of the assay in context of the variant using in silico data, synthetic RNA templates and clinical specimens. Sensitivity was reduced observed at low copy numbers for templates encoding the variant sequence. We designed and tested an alternate priming strategy, rescuing the sensitivity of the assay.


Asunto(s)
Virus del Sarampión , Sarampión , ARN Viral , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Humanos , Sarampión/diagnóstico , Sarampión/virología , Virus del Sarampión/genética , Virus del Sarampión/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , ARN Viral/genética
6.
J Virol ; 98(8): e0033224, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39078194

RESUMEN

The Mononegavirales, or non-segmented negative-sense RNA viruses (nsNSVs), includes significant human pathogens, such as respiratory syncytial virus, parainfluenza virus, measles virus, Ebola virus, and rabies virus. Although these viruses differ widely in their pathogenic properties, they are united by each having a genome consisting of a single strand of negative-sense RNA. Consistent with their shared genome structure, the nsNSVs have evolved similar ways to transcribe their genome into mRNAs and replicate it to produce new genomes. Importantly, both mRNA transcription and genome replication are performed by a single virus-encoded polymerase. A fundamental and intriguing question is: how does the nsNSV polymerase commit to being either an mRNA transcriptase or a replicase? The polymerase must become committed to one process or the other either before it interacts with the genome template or in its initial interactions with the promoter sequence at the 3´ end of the genomic RNA. This review examines the biochemical, molecular biology, and structural biology data regarding the first steps of transcription and RNA replication that have been gathered over several decades for different families of nsNSVs. These findings are discussed in relation to possible models that could explain how an nsNSV polymerase initiates and commits to either transcription or genome replication.


Asunto(s)
Genoma Viral , ARN Viral , Replicación Viral , Humanos , ARN Viral/genética , ARN Viral/metabolismo , Transcripción Viral/genética , Virus ARN de Sentido Negativo/genética , Virus ARN de Sentido Negativo/metabolismo , Transcripción Genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , ARN Polimerasa Dependiente del ARN/metabolismo , ARN Polimerasa Dependiente del ARN/genética
7.
Cell ; 187(16): 4213-4230.e19, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39013471

RESUMEN

Foamy viruses (FVs) are an ancient lineage of retroviruses, with an evolutionary history spanning over 450 million years. Vector systems based on Prototype Foamy Virus (PFV) are promising candidates for gene and oncolytic therapies. Structural studies of PFV contribute to the understanding of the mechanisms of FV replication, cell entry and infection, and retroviral evolution. Here we combine cryoEM and cryoET to determine high-resolution in situ structures of the PFV icosahedral capsid (CA) and envelope glycoprotein (Env), including its type III transmembrane anchor and membrane-proximal external region (MPER), and show how they are organized in an integrated structure of assembled PFV particles. The atomic models reveal an ancient retroviral capsid architecture and an unexpected relationship between Env and other class 1 fusion proteins of the Mononegavirales. Our results represent the de novo structure determination of an assembled retrovirus particle.


Asunto(s)
Microscopía por Crioelectrón , Spumavirus , Ensamble de Virus , Internalización del Virus , Spumavirus/genética , Cápside/metabolismo , Cápside/química , Cápside/ultraestructura , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Humanos , Evolución Molecular , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Modelos Moleculares
8.
Microbiol Mol Biol Rev ; : e0000123, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38912755

RESUMEN

SUMMARYSeveral examples of high-impact cross-species transmission of newly emerging or re-emerging bat-borne viruses, such as Sudan virus, Nipah virus, and severe acute respiratory syndrome coronavirus 2, have occurred in the past decades. Recent advancements in next-generation sequencing have strengthened ongoing efforts to catalog the global virome, in particular from the multitude of different bat species. However, functional characterization of these novel viruses and virus sequences is typically limited with regard to assessment of their cross-species potential. Our understanding of the intricate interplay between virus and host underlying successful cross-species transmission has focused on the basic mechanisms of entry and replication, as well as the importance of host innate immune responses. In this review, we discuss the various roles of the respective molecular mechanisms underlying cross-species transmission using different recent bat-borne viruses as examples. To delineate the crucial cellular and molecular steps underlying cross-species transmission, we propose a framework of overall characterization to improve our capacity to characterize viruses as benign, of interest, or of concern.

9.
Proc Natl Acad Sci U S A ; 121(24): e2403389121, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38833471

RESUMEN

Cell-cell fusion mediated by most paramyxovirus requires fusion protein (F) and attachment protein (H, HN, or G). The F protein is proteolytic cleaved to be fusogenically active. J paramyxovirus (JPV) has a unique feature in the family Paramyxoviridae: It encodes an integral membrane protein, syncytial protein (SP, formerly known as transmembrane protein, TM), which is essential in JPV-promoted cell-cell fusion (i.e., syncytial). In this study, we report that cleavage of SP is essential for its syncytial-promoting activity. We have identified the cleavage site of SP at amino acid residues 172 to 175, LKTG, and deletion of the "LKTG" residues abolished SP protein cleavage and its ability to promote cell-cell fusion. Replacing the cleavage site LKTG with a factor Xa protease cleavage site allows cleavage of the SP with factor Xa protease and restores its ability to promote cell-cell fusion. Furthermore, results from a hemifusion assay indicate that cleavage of SP plays an important role in the progression from the intermediate hemifusion state to a complete fusion. This work indicates that SP has many characteristics of a fusion protein. We propose that SP is likely a cell-cell fusion-promoting protein.


Asunto(s)
Fusión Celular , Proteínas Virales de Fusión , Animales , Proteínas Virales de Fusión/metabolismo , Chlorocebus aethiops , Proteolisis , Células Vero , Internalización del Virus , Factor Xa/metabolismo , Humanos , Línea Celular
10.
Virol J ; 21(1): 146, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918816

RESUMEN

The genus Jeilongvirus comprises non-segmented negative-stranded RNA viruses that are classified within the Paramyxoviridae family by phylogeny. Jeilongviruses are found in various reservoirs, including rodents and bats. Rodents are typical viral reservoirs with diverse spectra and zoonotic potential. Little is currently known about jeilongviruses in rodents from central China. The study utilized high-throughput and Sanger sequencing to obtain jeilongvirus genomes, including those of two novel strains (HBJZ120/CHN/2021 (17,468 nt) and HBJZ157/CHN/2021 (19,143 nt)) and three known viruses (HBXN18/CHN/2021 (19,212 nt), HBJZ10/CHN/2021 (19,700 nt), HBJM106/CHN/2021 (18,871 nt)), which were characterized by genome structure, identity matrix, and phylogenetic analysis. Jeilongviruses were classified into three subclades based on their topology, phylogeny, and hosts. Based on the amino acid sequence identities and phylogenetic analysis of the L protein, HBJZ120/CHN/2021 and HBJZ157/CHN/2021 were found to be strains rather than novel species. Additionally, according to specific polymerase chain reaction screening, the positive percentage of Beilong virus in Hubei was 6.38%, suggesting that Beilong virus, belonging to the Jeilongvirus genus, is likely to be widespread in wild rodents. The identification of novel strains further elucidated the genomic diversity of jeilongviruses. Additionally, the prevalence of jeilongviruses in Hubei, China, was profiled, establishing a foundation for the surveillance and early warning of emerging paramyxoviruses.


Asunto(s)
Genoma Viral , Filogenia , Roedores , Animales , China , Roedores/virología , Animales Salvajes/virología , Paramyxovirinae/genética , Paramyxovirinae/clasificación , Paramyxovirinae/aislamiento & purificación , ARN Viral/genética , Infecciones por Paramyxoviridae/veterinaria , Infecciones por Paramyxoviridae/virología , Infecciones por Paramyxoviridae/epidemiología , Secuenciación de Nucleótidos de Alto Rendimiento , Reservorios de Enfermedades/virología , Análisis de Secuencia de ADN
11.
J Infect Dis ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38842160

RESUMEN

BACKGROUND: Nipah virus is an emerging zoonotic virus that causes severe respiratory disease and meningoencephalitis. The pathophysiology of Nipah virus meningoencephalitis is poorly understood. METHODS: We have collected the brains of African green monkeys during multiple Nipah virus, Bangladesh studies, resulting in 14 brains with Nipah virus-associated lesions. RESULTS: The lesions seen in the brain of African green monkeys infected with Nipah virus, Bangladesh were very similar to those observed in humans with Nipah virus, Malaysia infection. We observed viral RNA and antigen within neurons and endothelial cells, within encephalitis foci and in uninflamed portions of the CNS. CD8+ T cells had a consistently high prevalence in CNS lesions. We developed a UNet model for quantifying and visualizing inflammation in the brain in a high-throughput and unbiased manner. While CD8+ T cells had a consistently high prevalence in CNS lesions, the model revealed that CD68+ cells were numerically the immune cell with the highest prevalence in the CNS of NiV-infected animals. CONCLUSION: Our study provides an in-depth analysis on Nipah virus infection in the brains of primates, and similarities between lesions in patients and the animals in our study validate this model.

12.
Methods Mol Biol ; 2808: 141-152, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743368

RESUMEN

Measles virus (MeV) infection of airway surface epithelial cells provides a site for final amplification before being released back into the environment via coughing and sneezing. Multiple cell lines have served as models of polarized epithelia for MeV infection, such as Caco2 cells (intestinal derived human epithelia) or MDCK cells (kidney derived canine epithelia). In this chapter, we describe the materials and air-liquid interface (ALI) culture conditions for maintaining four different cell lines derived from human airway epithelial cells: 16HBE14o-, Calu-3, H358, and NuLi-1. We provide methods for confirming transepithelial electrical resistance (TER) and preparing samples for microscopy as well as expected results from apical or basolateral MeV delivery. Polarized human airway derived cells serve as tissue culture models for investigating targeted questions about how MeV exits a human host. In addition, these methods are generalizable to studies of other respiratory viruses or the biology of ALI airway epithelial cells.


Asunto(s)
Técnicas de Cultivo de Célula , Células Epiteliales , Virus del Sarampión , Humanos , Virus del Sarampión/fisiología , Células Epiteliales/virología , Células Epiteliales/citología , Técnicas de Cultivo de Célula/métodos , Sarampión/virología , Línea Celular , Perros , Animales , Mucosa Respiratoria/virología , Mucosa Respiratoria/citología , Impedancia Eléctrica
13.
Methods Mol Biol ; 2808: 129-140, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743367

RESUMEN

Many negative-sense single-stranded RNA viruses within the order Mononegavirales harm humans. A common feature shared among cells infected by these viruses is the formation of subcellular membraneless structures called biomolecular condensates, also known as inclusion bodies (IBs), that form through a process called liquid-liquid phase separation (LLPS). Like many other membraneless organelles, viral IBs enrich a specific subset of viral and host proteins involved in the formation of viral particles. Elucidation of the properties and regulation of these IBs as they mature throughout the viral replication process are important for our understanding of viral replication, which may also lead to the development of alternative antiviral treatments. The protocol outlined in this chapter aims to characterize the intrinsic properties of LLPS within the measles virus (MeV, a member of Mononegavirales) IBs by using an imaging approach that fluorescently tags an IB-associated host protein. This method uses common laboratory techniques and is generalizable to any host factors as well as other viral systems.


Asunto(s)
Recuperación de Fluorescencia tras Fotoblanqueo , Cuerpos de Inclusión Viral , Virus del Sarampión , Humanos , Cuerpos de Inclusión Viral/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Virus del Sarampión/fisiología , Virus del Sarampión/metabolismo , Replicación Viral , Cuerpos de Inclusión/metabolismo , Animales , Interacciones Huésped-Patógeno , Separación de Fases
14.
Methods Mol Biol ; 2808: 167-175, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743370

RESUMEN

Measles virus is one of the most contagious airborne human viruses which keeps causing outbreaks in numerous countries over the world despite the existence of an efficient vaccine. Fusion inhibitory lipopeptides were shown to inhibit viral entry into target cells, and their adequate administration into the respiratory tract may provide a novel preventive approach against airborne infections. Aerosol delivery presents the best administration route to deliver such preventive compounds to the upper and lower respiratory tract. This approach offers a conceptually new strategy to protect the population at risk against infection by respiratory viruses, including measles. It is a noninvasive needle-free approach, which may be used when antiviral protection is required, without any medical assistance. In this chapter, we describe the nebulization approach of lipopeptide compounds in nonhuman primates and the subsequent measles virus challenge.


Asunto(s)
Aerosoles , Modelos Animales de Enfermedad , Virus del Sarampión , Sarampión , Animales , Sarampión/prevención & control , Lipopéptidos/administración & dosificación , Humanos , Sistemas de Liberación de Medicamentos/métodos
15.
Sci Rep ; 14(1): 10741, 2024 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730036

RESUMEN

The majority of pigeon paramyxovirus type 1 (PPMV-1) strains are generally non-pathogenic to chickens; however, they can induce severe illness and high mortality rates in pigeons, leading to substantial economic repercussions. The genomes of 11 PPMV-1 isolates from deceased pigeons on meat pigeon farms during passive monitoring from 2009 to 2012 were sequenced and analyzed using polymerase chain reaction and phylogenetic analysis. The complete genome lengths of 11 isolates were approximately 15,192 nucleotides, displaying a consistent gene order of 3'-NP-P-M-F-HN-L-5'. ALL isolates exhibited the characteristic motif of 112RRQKRF117 at the fusion protein cleavage site, which is characteristic of velogenic Newcastle disease virus. Moreover, multiple mutations have been identified within the functional domains of the F and HN proteins, encompassing the fusion peptide, heptad repeat region, transmembrane domains, and neutralizing epitopes. Phylogenetic analysis based on sequences of the F gene unveiled that all isolates clustered within genotype VI in class II. Further classification identified at least two distinct sub-genotypes, with seven isolates classified as sub-genotype VI.2.1.1.2.2, whereas the others were classified as sub-genotype VI.2.1.1.2.1. This study suggests that both sub-genotypes were implicated in severe disease manifestation among meat pigeons, with sub-genotype VI.2.1.1.2.2 displaying an increasing prevalence among Shanghai's meat pigeon population since 2011. These results emphasize the value of developing pigeon-specific vaccines and molecular diagnostic tools for monitoring and proactively managing potential PPMV-1 outbreaks.


Asunto(s)
Columbidae , Genoma Viral , Enfermedad de Newcastle , Virus de la Enfermedad de Newcastle , Filogenia , Animales , Columbidae/virología , China/epidemiología , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/aislamiento & purificación , Virus de la Enfermedad de Newcastle/clasificación , Enfermedad de Newcastle/virología , Enfermedad de Newcastle/epidemiología , Genotipo , Granjas , Carne/virología
16.
Artículo en Inglés | MEDLINE | ID: mdl-38770087

RESUMEN

Henipaviruses are enveloped single-stranded, negative-sense RNA viruses of the paramyxovirus family. Two henipaviruses, Nipah virus and Hendra virus, cause a systemic respiratory and/or neurological disease in humans and ten additional species of mammals, with a high fatality rate. Because of their highly pathogenic nature, Nipah virus and Hendra virus are categorized as BSL-4 pathogens, which limits the number and scope of translational research studies on these important human pathogens. To begin to address this limitation, we are developing a BSL-2 model of authentic henipavirus infection in mice, using the non-pathogenic henipavirus, Cedar virus. Notably, wild-type mice are highly resistant to Hendra virus and Nipah virus infection. However, previous work has shown that mice lacking expression of the type I interferon receptor (IFNAR-KO mice) are susceptible to both viruses. Here, we show that luciferase-expressing recombinant Cedar virus (rCedV-luc) is also able to replicate and establish a transient infection in IFNAR-KO mice, but not in wild-type mice. Using longitudinal bioluminescence imaging (BLI) of luciferase expression, we detected rCedV-luc replication as early as 10 h post-infection. Viral replication peaks between days 1 and 3 post-infection, and declines to levels undetectable by BLI by 7 days post-infection. Immunohistochemistry is consistent with viral infection and replication in endothelial cells and other non-immune cell types within tissue parenchyma. Serology analyses demonstrate significant IgG responses to the Cedar virus surface glycoprotein with potent neutralizing activity in IFNAR-KO mice, whereas antibody responses in wild-type animals were non-significant. Overall, these data suggest that rCedV-luc infection of IFNAR-KO mice represents a viable platform for the study of in vivo henipavirus replication, anti-henipavirus host responses and henipavirus-directed therapeutics.

17.
Microb Genom ; 10(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38700925

RESUMEN

Paramyxoviruses are a group of single-stranded, negative-sense RNA viruses, some of which are responsible for acute human disease, including parainfluenza virus, measles virus, Nipah virus and Hendra virus. In recent years, a large number of novel paramyxoviruses, particularly members of the genus Jeilongvirus, have been discovered in wild mammals, suggesting that the diversity of paramyxoviruses may be underestimated. Here we used hemi-nested reverse transcription PCR to obtain 190 paramyxovirus sequences from 969 small mammals in Hubei Province, Central China. These newly identified paramyxoviruses were classified into four clades: genera Jeilongvirus, Morbillivirus, Henipavirus and Narmovirus, with most of them belonging to the genus Jeilongvirus. Using Illumina sequencing and Sanger sequencing, we successfully recovered six near-full-length genomes with different genomic organizations, revealing the more complex genome content of paramyxoviruses. Co-divergence analysis of jeilongviruses and their known hosts indicates that host-switching occurred more frequently in the evolutionary histories of the genus Jeilongvirus. Together, our findings demonstrate the high prevalence of paramyxoviruses in small mammals, especially jeilongviruses, and highlight the diversity of paramyxoviruses and their genome content, as well as the evolution of jeilongviruses.


Asunto(s)
Infecciones por Paramyxoviridae , Paramyxovirinae , Paramyxovirinae/genética , Infecciones por Paramyxoviridae/epidemiología , Infecciones por Paramyxoviridae/veterinaria , Mamíferos , China , Filogenia , Genoma Viral , Especificidad del Huésped
18.
Open Vet J ; 14(1): 12-18, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38633151

RESUMEN

Newcastle disease (ND) is a tremendously contagious avian infection with extensive monetary ramifications for the chicken zone. To reduce the effect of ND on the Saudi rooster enterprise, our analysis emphasizes the necessity of genotype-particular vaccinations, elevated surveillance, public recognition campaigns, and stepped-forward biosecurity. Data show that one-of-a-kind bird species, outdoor flocks, and nearby differences in susceptibility are all vulnerable. The pathogenesis consists of tropism in the respiratory and gastrointestinal structures and some genotypes boom virulence. Laboratory diagnostics use reverse transcription-polymerase chain reaction, sequencing, and serotyping among different strategies. Vital records are supplied through immune responses and serological trying out. Vaccination campaigns, biosecurity protocols, and emergency preparedness are all covered in prevention and manipulation techniques. Notably, co-circulating genotypes and disparities in immunization regulations worry Saudi Arabia. The effect of ND in Saudi Arabia is tested in this paper, with precise attention paid to immunological reaction, pathogenesis, susceptibility elements, laboratory analysis, and preventative and manipulation measures. Saudi Arabia can shield its bird region and beef up its defences against Newcastle's ailment, enforcing those hints into its policies.


Asunto(s)
Enfermedades de los Bovinos , Enfermedad de Newcastle , Enfermedades de las Aves de Corral , Bovinos , Animales , Masculino , Aves de Corral , Pollos , Arabia Saudita , Virus de la Enfermedad de Newcastle/genética , Enfermedades de las Aves de Corral/epidemiología , Enfermedad de Newcastle/epidemiología
19.
Microbiol Spectr ; 12(5): e0041724, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38606982

RESUMEN

Paramyxo- and filovirus genomes are equipped with bipartite promoters at their 3' ends to initiate RNA synthesis. The two elements, the primary promoter element 1 (PE1) and the secondary promoter element 2 (PE2), are separated by a spacer region that must be precisely a multiple of 6 nucleotides (nts), indicating these viruses adhere to the "rule of six." However, our knowledge of PE2 has been limited to a narrow spectrum of virus species. In this study, a comparative analysis of 1,647 paramyxoviral genomes from a public database revealed that the paramyxovirus PE2 can be clearly categorized into two distinct subcategories: one marked by C repeats at every six bases (exclusive to the subfamily Orthoparamyxovirinae) and another characterized by CG repeats every 6 nts (observed in the subfamilies Avulavirinae and Rubulavirinae). This unique pattern collectively mirrors the evolutionary lineage of these subfamilies. Furthermore, we showed that PE2 of the Rubulavirinae, with the exception of mumps virus, serves as part of the gene-coding region. This may be due to the fact that the Rubulavirinae are the only paramyxoviruses that cannot propagate without RNA editing. Filoviruses have three to eight consecutive uracil repeats every six bases (UN5) in PE2, which is located in the 3' end region of the genome. We obtained PE2 sequences from 2,195 filoviruses in a public database and analyzed the sequence conservation among virus species. Our results indicate that the continuity of UN5 hexamers is consistently maintained with a high degree of conservation across virus species. IMPORTANCE: The genomic intricacies of paramyxo- and filoviruses are highlighted by the bipartite promoters-promoter element 1 (PE1) and promoter element 2 (PE2)-at their 3' termini. The spacer region between these elements follows the "rule of six," crucial for genome replication. By a comprehensive analysis of paramyxoviral genome sequences, we identified distinct subcategories of PE2 based on C and CG repeats that were specific to Orthoparamyxovirinae and Avulavirinae/Rubulavirinae, respectively, mirroring their evolutionary lineages. Notably, the PE2 of Rubulavirinae is integrated into the gene-coding region, a unique trait potentially linked to its strict dependence on RNA editing for virus growth. This study also focused on the PE2 sequences in filovirus genomes. The strict conservation of the continuity of UN5 among virus species emphasizes its crucial role in viral genome replication.


Asunto(s)
Filoviridae , Genoma Viral , Filogenia , Regiones Promotoras Genéticas , Regiones Promotoras Genéticas/genética , Genoma Viral/genética , Filoviridae/genética , Filoviridae/clasificación , Paramyxoviridae/genética , Paramyxoviridae/clasificación , Humanos , ARN Viral/genética , Evolución Molecular , Animales
20.
Viruses ; 16(4)2024 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-38675988

RESUMEN

Sosuga virus (SOSV), a rare human pathogenic paramyxovirus, was first discovered in 2012 when a person became ill after working in South Sudan and Uganda. During an ecological investigation, several species of bats were sampled and tested for SOSV RNA and only one species, the Egyptian rousette bat (ERBs; Rousettus aegyptiacus), tested positive. Since that time, multiple other species have been sampled and ERBs in Uganda have continued to be the only species of bat positive for SOSV infection. Subsequent studies of ERBs with SOSV demonstrated that ERBs are a competent host for SOSV and shed this infectious virus while exhibiting only minor infection-associated pathology. Following the 2014 Ebola outbreak in West Africa, surveillance efforts focused on discovering reservoirs for zoonotic pathogens resulted in the capture and testing of many bat species. Here, SOSV RNA was detected by qRT-PCR only in ERBs captured in the Moyamba District of Sierra Leone in the central region of the country. These findings represent a substantial range extension from East Africa to West Africa for SOSV, suggesting that this paramyxovirus may occur in ERB populations throughout its sub-Saharan African range.


Asunto(s)
Quirópteros , Animales , Quirópteros/virología , Sierra Leona/epidemiología , Infecciones por Paramyxoviridae/veterinaria , Infecciones por Paramyxoviridae/virología , Infecciones por Paramyxoviridae/epidemiología , ARN Viral/genética , Filogenia , Reservorios de Enfermedades/virología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA