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








Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 12(3): e0365823, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38323828

RESUMO

The internal ribosome entry site (IRES) element constitutes a cis-acting RNA regulatory sequence that recruits the ribosomal initiation complex in a cap-independent manner, assisted by various RNA-binding proteins and IRES trans-acting factors. Foot-and-mouth disease virus (FMDV) contains a functional IRES element and takes advantage of this element to subvert host translation machinery. Our study identified a novel mechanism wherein RALY, a member of the heterogeneous nuclear ribonucleoproteins (hnRNP) family belonging to RNA-binding proteins, binds to the domain 3 of FMDV IRES via its RNA recognition motif residue. This interaction results in the downregulation of FMDV replication by inhibiting IRES-driven translation. Furthermore, our findings reveal that the inhibitory effect exerted by RALY on FMDV replication is not attributed to the FMDV IRES-mediated assembly of translation initiation complexes but rather to the impediment of 80S ribosome complex formation after binding with 40S ribosomes. Conversely, 3Cpro of FMDV counteracts RALY-mediated inhibition by the ubiquitin-proteasome pathway. Therefore, these results indicate that RALY, as a novel critical IRES-binding protein, inhibits FMDV replication by blocking the formation of 80S ribosome, providing a deeper understanding of how viruses recruit and manipulate host factors. IMPORTANCE: The translation of FMDV genomic RNA driven by IRES element is a crucial step for virus infections. Many host proteins are hijacked to regulate FMDV IRES-dependent translation, but the regulatory mechanism remains unknown. Here, we report for the first time that cellular RALY specifically interacts with the IRES of FMDV and negatively regulates viral replication by blocking 80S ribosome assembly on FMDV IRES. Conversely, RALY-mediated inhibition is antagonized by the viral 3C protease by the ubiquitin-proteasome pathway. These results would facilitate further understanding of virus-host interactions and translational control during viral infection.


Assuntos
Vírus da Febre Aftosa , Animais , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas de Ligação a RNA/genética , Ribossomos/genética , Endopeptidases/metabolismo , Sítios Internos de Entrada Ribossomal , Proteases Virais 3C , Ubiquitinas/genética , Ubiquitinas/metabolismo
2.
J Virol ; 97(8): e0018123, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37565750

RESUMO

Vacuolar protein sorting 28 (Vps28), a component of the ESCRT-I (endosomal sorting complex required for transport I), plays an important role in the pathogen life cycle. Here, we investigated the reciprocal regulation between Vps28 and the foot-and-mouth disease virus (FMDV). Overexpression of Vps28 decreased FMDV replication. On the contrary, the knockdown of Vps28 increased viral replication. Subsequently, the mechanistic study showed that Vps28 destabilized the replication complex (RC) by associating with 3A rather than 2C protein. In addition, Vps28 targeted FMDV VP0, VP1, and VP3 for degradation to inhibit viral replication. To counteract this, FMDV utilized tactics to restrict Vps28 to promote viral replication. FMDV degraded Vps28 mainly through the ubiquitin-proteasome pathway. Additional data demonstrated that 2B and 3A proteins recruited E3 ubiquitin ligase tripartite motif-containing protein 21 to degrade Vps28 at Lys58 and Lys25, respectively, and FMDV 3Cpro degraded Vps28 through autophagy and its protease activity. Meantime, the 3Cpro-mediated Vps28 degradation principally alleviated the ability to inhibit viral propagation. Intriguingly, we also demonstrated that the N-terminal and C-terminal domains of Vps28 were responsible for the suppression of FMDV replication, which suggested the elaborated counteraction between FMDV and Vps28. Collectively, our results first investigate the role of ESCRTs in host defense against picornavirus and unveil underlying strategies utilized by FMDV to evade degradation machinery for triumphant propagation. IMPORTANCE ESCRT machinery plays positive roles in virus entry, replication, and budding. However, little has been reported on its negative regulation effects during viral infection. Here, we uncovered the novel roles of ESCRT-I subunit Vps28 on FMDV replication. The data indicated that Vps28 destabilized the RC and impaired viral structural proteins VP0, VP1, and VP3 to inhibit viral replication. To counteract this, FMDV hijacked intracellular protein degradation pathways to downregulate Vps28 expression and thus promoted viral replication. Our findings provide insights into how ESCRT regulates pathogen life cycles and elucidate additional information regarding FMDV counteraction of host antiviral activity.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Animais , Vírus da Febre Aftosa/metabolismo , Proteínas Virais/metabolismo , Transdução de Sinais , Transporte Proteico , Replicação Viral/fisiologia
3.
mBio ; 14(2): e0035823, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-36939331

RESUMO

Viruses lack the properties to replicate independently due to the limited resources encoded in their genome; therefore, they hijack the host cell machinery to replicate and survive. Picornaviruses get the prerequisite for effective protein synthesis through specific sequences known as internal ribosome entry sites (IRESs). In the past 2 decades, significant progress has been made in identifying different types of IRESs in picornaviruses. This review will discuss the past and current findings related to the five different types of IRESs and various internal ribosome entry site trans-acting factors (ITAFs) that either promote or suppress picornavirus translation and replication. Some IRESs are inefficient and thus require ITAFs. To achieve their full efficiency, they recruit various ITAFs, which enable them to translate more effectively and efficiently, except type IV IRES, which does not require any ITAFs. Although there are two kinds of ITAFs, one promotes viral IRES-dependent translation, and the second type restricts. Picornaviruses IRESs are classified into five types based on their use of sequence, ITAFs, and initiation factors. Some ITAFs regulate IRES activity by localizing to the viral replication factories in the cytoplasm. Also, some drugs, chemicals, and herbal extracts also regulate viral IRES-dependent translation and replication. Altogether, this review will elaborate on our understanding of the past and recent advancements in the IRES-dependent translation and replication of picornaviruses. IMPORTANCE The family Picornaviridae is divided into 68 genera and 158 species. The viruses belonging to this family range from public health importance, such as poliovirus, enterovirus A71, and hepatitis A virus, to animal viruses of great economic importance, such as foot-and-mouth disease virus. The genomes of picornaviruses contain 5' untranslated regions (5' UTRs), which possess crucial and highly structured stem-loops known as IRESs. IRES assemble the ribosomes and facilitate the cap-independent translation. Virus-host interaction is a hot spot for researchers, which warrants deep insight into understanding viral pathogenesis better and discovering new tools and ways for viral restriction to improve human and animal health. The cap-independent translation in the majority of picornaviruses is modulated by ITAFs, which bind to various IRES regions to initiate the translation. The discoveries of ITAFs substantially contributed to understanding viral replication behavior and enhanced our knowledge about virus-host interaction more effectively than ever before. This review discussed the various types of IRESs found in Picornaviridae, past and present discoveries regarding ITAFs, and their mechanism of action. The herbal extracts, drugs, and chemicals, which indicated their importance in controlling viruses, were also summarized. In addition, we discussed the movement of ITAFs from the nucleus to viral replication factories. We believe this review will stimulate researchers to search for more novel ITAFs, drugs, herbal extracts, and chemicals, enhancing the understanding of virus-host interaction.


Assuntos
Vírus da Febre Aftosa , Vírus da Hepatite A , Picornaviridae , Animais , Humanos , Picornaviridae/genética , Sítios Internos de Entrada Ribossomal , Vírus da Febre Aftosa/fisiologia , Ribossomos/genética , Ribossomos/metabolismo , Vírus da Hepatite A/metabolismo , Biossíntese de Proteínas , RNA Viral/metabolismo
4.
mBio ; 13(5): e0143422, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36106732

RESUMO

The maintenance of viral protein homeostasis depends on the machinery of the infected host cells, giving us an insight into the interplay between host and virus. Accumulating evidence suggests that heat shock protein 60 (HSP60), as one molecular chaperone, is involved in regulating virus infection. However, the role of HSP60 during foot-and-mouth disease virus (FMDV) replication and its specific mechanisms have not been reported. We demonstrate that HSP60 modulates the FMDV life cycle. HSP60 plays a role at the postentry stage of the viral life cycle, including RNA replication and mRNA translation; however, HSP60 does not affect viral replication of Seneca Valley virus (SVA) or encephalomyocarditis virus (EMCV). We found that HSP60 is involved in FMDV replication complex (RC) formation. Furthermore, our results indicate that HSP60 interacts with FMDV nonstructural proteins 3A and 2C, key elements of the viral replication complex. We also show that HSP60 regulates the stability of 3A and 2C via caspase-dependent and autophagy-lysosome-dependent degradation, thereby promoting FMDV RNA synthesis and mRNA translation mediated by the RC. Additionally, we determined that the apical domain of HSP60 is responsible for interacting with 3A and 2C. The N terminus of 3A and ATPase domain of 2C are involved in binding to HSP60. Importantly, HSP60 depletion potently reduced FMDV pathogenicity in infected mice. Altogether, this study demonstrates a specific role of HSP60 in promoting FMDV replication. Furthermore, targeting host HSP60 will help us design the FMDV-specific antiviral drugs. IMPORTANCE FMDV is the leading cause of the foot-and-mouth disease (FMD), affecting cloven-hoofed animals with high morbidity and mortality. We determined that HSP60 is required for efficient viral RNA replication and mRNA translation during FMDV infection. Furthermore, we demonstrate that HSP60 interacts with FMDV nonstructural proteins 3A and 2C, the elements of the RC; HSP60 contributes to the stability of 3A and 2C to affect the formation and function of the RC. We also validated the potential role of HSP60 as the antiviral target in vivo using small interfering RNA. These findings deepen the understanding of the host-virus interaction and provide information supporting the design of novel therapeutics for FMDV infection.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Animais , Camundongos , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Chaperonina 60/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , RNA Interferente Pequeno/metabolismo , Linhagem Celular , Vírus da Febre Aftosa/genética , Replicação Viral/fisiologia , Antivirais/metabolismo , Caspases/metabolismo
5.
Front Cell Infect Microbiol ; 11: 707107, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34532300

RESUMO

Endoplasmic reticulum (ER) stress-induced autophagy is closely associated with viral infection and propagation. However, the intrinsic link between ER stress, autophagy, and viral replication during foot-and-mouth disease virus (FMDV) infection is not fully elucidated. Our previous studies demonstrated that FMDV infection activated the ER stress-associated UPR of the PERK-eIF2a and ATF6 signaling pathway, whereas the IRE1a signaling was suppressed. We found that the activated-ATF6 pathway participated in FMDV-induced autophagy and FMDV replication, while the IRE1α pathway only affected FMDV replication. Further studies indicated that Sec62 was greatly reduced in the later stages of FMDV infection and blocked the activation of the autophagy-related IRE1α-JNK pathway. Moreover, it was also found that Sec62 promoted IRE1a phosphorylation and negatively regulated FMDV proliferation. Importantly, Sec62 may interact with LC3 to regulate ER stress and autophagy balance and eventually contribute to FMDV clearance via fusing with lysosomes. Altogether, these results suggest that Sec62 is a critical molecule in maintaining and recovering ER homeostasis by activating the IRE1α-JNK pathway and delivering autophagosome into the lysosome, thus providing new insights on FMDV-host interactions and novel antiviral therapies.


Assuntos
Autofagia , Estresse do Retículo Endoplasmático , Vírus da Febre Aftosa , Proteínas de Membrana Transportadoras/metabolismo , Replicação Viral , Animais , Endorribonucleases , Vírus da Febre Aftosa/fisiologia , Proteínas Serina-Treonina Quinases
6.
Viruses ; 13(9)2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34578346

RESUMO

In cells, the contributions of DEAD-box helicases (DDXs), without which cellular life is impossible, are of utmost importance. The extremely diverse roles of the nucleolar helicase DDX21, ranging from fundamental cellular processes such as cell growth, ribosome biogenesis, protein translation, protein-protein interaction, mediating and sensing transcription, and gene regulation to viral manipulation, drew our attention. We designed this project to study virus-host interactions and viral pathogenesis. A pulldown assay was used to investigate the association between foot-and-mouth disease virus (FMDV) and DDX21. Further insight into the DDX21-FMDV interaction was obtained through dual-luciferase, knockdown, overexpression, qPCR, and confocal microscopy assays. Our results highlight the antagonistic feature of DDX21 against FMDV, as it progressively inhibited FMDV internal ribosome entry site (IRES) -dependent translation through association with FMDV IRES domains 2, 3, and 4. To subvert this host helicase antagonism, FMDV degraded DDX21 through its non-structural proteins 2B, 2C, and 3C protease (3Cpro). Our results suggest that DDX21 is degraded during 2B and 2C overexpression and FMDV infection through the caspase pathway; however, DDX21 is degraded through the lysosomal pathway during 3Cpro overexpression. Further investigation showed that DDX21 enhanced interferon-beta and interleukin-8 production to restrict viral replication. Together, our results demonstrate that DDX21 is a novel FMDV IRES trans-acting factor, which negatively regulates FMDV IRES-dependent translation and replication.


Assuntos
RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/metabolismo , Animais , Linhagem Celular , Febre Aftosa/virologia , Regulação Viral da Expressão Gênica , Técnicas de Silenciamento de Genes , Ribonucleoproteínas Nucleares Heterogêneas , Interações entre Hospedeiro e Microrganismos , Interações Hospedeiro-Patógeno , Humanos , Interferon beta/genética , Sítios Internos de Entrada Ribossomal , Proteína de Ligação a Regiões Ricas em Polipirimidinas , Mapas de Interação de Proteínas , Proteínas Virais/genética , Replicação Viral/genética
7.
Front Immunol ; 12: 616402, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093518

RESUMO

In addition to ribosomal protein synthesis and protein translation, ribosomal proteins also participate in tumorigenesis and tumor progression, immune responses, and viral replication. Here, we show that ribosomal protein L13 (RPL13) participates in the antiviral immune response induced by foot-and-mouth disease virus (FMDV), inhibiting FMDV replication. The overexpression of RPL13 promoted the induction and activation of the promoters of the nuclear factor-κB (NF-κB) and interferon-ß (IFN-ß) genes, and the expression and protein secretion of the antiviral factor IFN-ß and proinflammatory cytokine interleukin-6 (IL-6). The knockdown of RPL13 had the opposite effects. We also found that the FMDV 3Cpro protease interacts with RPL13, and that its activity reduces the expression of RPL13, thus antagonizing the RPL13-mediated antiviral activity. This study extends our knowledge of the extraribosomal functions of ribosomal proteins and provides new scientific information on cellular antiviral defenses and virus-antagonizing mechanisms.


Assuntos
Vírus da Febre Aftosa/imunologia , Febre Aftosa/imunologia , Febre Aftosa/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Proteínas de Neoplasias/metabolismo , Proteínas Ribossômicas/metabolismo , Animais , Biomarcadores , Linhagem Celular , RNA Helicases DEAD-box/metabolismo , Febre Aftosa/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas de Neoplasias/genética , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Ribossômicas/genética , Transdução de Sinais , Replicação Viral
8.
FEMS Microbiol Ecol ; 97(4)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33705527

RESUMO

The gut microbiota plays an important role in human health. In modern life, with the improvement of living conditions, the intake of high-sugar and high-fat diets as well as the large-scale use of antibacterial drugs have an extensive impact on the gut microbiota, even leading to gut microbiota-orchestrating disorders. This review discusses the effects of various factors, including geographic location, age, diet, antibacterial drugs, psychological situation and exercise on gut bacteria, which helps us profoundly to understand the significance of gut bacteria to human health and to find effective solutions to prevent or treat related diseases.


Assuntos
Microbioma Gastrointestinal , Bactérias , Dieta Hiperlipídica , Humanos
9.
Front Immunol ; 12: 602717, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33708201

RESUMO

Echinococcosis, mainly caused by Echinococcus granulosus, is one of the 17 neglected tropical diseases. Extracellular vesicles (EVs) play an essential role in the host-parasite interplay. However, the EVs in the hydatid fluid (HF) of E. granulosus are not fully characterized. Herein, three different types of HF EVs, designated as 2 K, 10 K, and 110 K EVs based on the centrifugal force used, were morphologically identified. A total of 97, 80, and 581 proteins were identified in 2 K, 10 K, and 110 K EVs, respectively, 39 of which were commonly shared. Moreover, 11, 8, and 25 miRNAs were detected, respectively, and all of the 7 selected miRNAs were validated by qPCR to be significantly lower abundant than that in protoscoleces. It was further deemed that 110 K EVs were internalized by sheep peripheral blood mononuclear cells (PBMCs) in a time-dependent manner and thus induced interleukin (IL)-10, tumor necrosis factor-α (TNF-α), and IRF5 were significantly upregulated and IL-1ß, IL-17, and CD14 were significantly downregulated (p < 0.05). These data demonstrate the physical discrepancy of three HF EVs and an immunomodulatory effect of 110 K EVs on sheep PMBCs, suggesting a role in immune responses during E. granulosus infection.


Assuntos
Equinococose/imunologia , Echinococcus granulosus/imunologia , Vesículas Extracelulares/imunologia , Imunomodulação , Leucócitos Mononucleares/imunologia , Ovinos , Animais , Citocinas/imunologia , Equinococose/parasitologia , Proteínas de Helminto/imunologia , Ovinos/imunologia , Ovinos/parasitologia
10.
Acta Trop ; 213: 105756, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33189712

RESUMO

Extracellular vesicles (EVs) are heterogeneous populations of different membrane-wrapped vesicles in size and encapsulated cargo and have recently emerged as a crucial carrier with the functions in intercellular communication, being involved in host-parasite interactions. However, Echinococcus granulosus EVs are not fully described. To separate EVs with a different size, the culture supernatant of E. granulosus protoscoleces (PSCs) was sequentially centrifuged at 2,000g, 10,000g and 110,000g, and the resulting precipitates were accordingly named as 2K, 10K and 110K EVs, respectively. The size and morphology of three different EVs were identified using ZETASIZER NANO and transmission electron microscopy (TEM), respectively. Then mass spectrometry was applied to define protein cargo of EVs and EV internalization was assessed using fluorescent microscopy and flow cytometry. The results showed that 2K EVs mainly ranged from 450 to 950 nm in diameter, 10K EVs ranged from 220 to 390 nm and 110K EVs from 60 to 150 nm. A total of 901 EV proteins were identified, 328 of which were commonly found in the three types of EVs. GO analysis revealed that these proteins were mainly involved in binding (44%) and catalytic activity (44%). Three types of EVs were different in biomarkers (Enolase and 14-3-3) and in reactivity with anti-echinococcosis positive serum. Moreover, 110K EVs were more easily internalized by hepatic cells than 10K EVs as well as 2K EVs (p < 0.0001). These results reveal the physical and biological discrepancy among 2K, 10K and 110K EVs, suggesting a distinct role in host-parasite interactions.


Assuntos
Equinococose/parasitologia , Echinococcus granulosus/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas de Helminto/metabolismo , Interações Hospedeiro-Parasita , Animais , Transporte Biológico , Células Cultivadas , Vesículas Extracelulares/química , Citometria de Fluxo , Hepatócitos/parasitologia , Camundongos , Microscopia Eletrônica de Transmissão , Ovinos
11.
Viruses ; 12(12)2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33255534

RESUMO

DEAD-box helicase 23 (DDX23) is a host nuclear helicase, which is a part of the spliceosomal complex and involved in pre-mRNA splicing. To investigate whether DDX23, an internal ribosomal entry sites transacting factor (ITAF) affects foot-and-mouth disease virus (FMDV) replication and translation through internal ribosome entry site (IRES)-dependent manner. For this, we utilized a pull-down assay, Western blotting, quantitative real-time PCR, confocal microscopy, overexpression and small interfering RNA knockdown, as well as the median tissue culture infective dose. Our findings showed that FMDV infection inhibited DDX23 expression and the overexpression of DDX23 reduced viral replication, however, CRISPR Cas9 knockout/small interfering RNA knockdown increased FMDV replication. FMDV IRES domain III and IV interacted with DDX23, whereas DDX23 interacted with FMDV 3C proteinase and significantly degraded. The enzymatic activity of FMDV 3C proteinase degraded DDX23, whereas FMDV degraded DDX23 via the lysosomal pathway. Additionally, IRES-driven translation was suppressed in DDX23-overexpressing cells, and was enhanced in DDX23 knocked down. Collectively, our results demonstrated that DDX23 negatively affects FMDV IRES-dependent translation, which could be a useful target for the design of antiviral drugs.


Assuntos
Cisteína Endopeptidases/metabolismo , RNA Helicases DEAD-box/metabolismo , Vírus da Febre Aftosa/fisiologia , Febre Aftosa/metabolismo , Febre Aftosa/virologia , Regulação Viral da Expressão Gênica , Proteínas Virais/metabolismo , Replicação Viral , Proteases Virais 3C , Animais , Linhagem Celular , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Sítios Internos de Entrada Ribossomal , Lisossomos , Ligação Proteica , Biossíntese de Proteínas , Proteólise
12.
J Adv Res ; 24: 317-324, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32455006

RESUMO

Argonaute proteins are highly conserved in almost all organisms. They not only involve in the biogenesis of small regulatory RNAs, but also regulate gene expression and defend against foreign pathogen invasion via small RNA-mediated gene silencing pathways. As a key player in these pathways, the abnormal expression and/or mis-modifications of Argonaute proteins lead to the disorder of small RNA biogenesis and functions, thus influencing multiply biological processes and disease development, especially cancer. In this review, we focus on the post-translational modifications and novel functions of Argonaute proteins in alternative splicing, host defense and genome editing.

13.
Front Microbiol ; 10: 2707, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31849869

RESUMO

Alveolar echinococcosis caused by Echinococcus multilocularis is an important zoonotic disease. In the infected mice, emu-miR-4989-3p is present in sera, but its role remains unknown. Using high-throughput sequencing and qPCR, emu-miR-4989-3p was herein confirmed to be encapsulated into E. multilocularis extracellular vesicles. In the transfected macrophages, emu-miR-4989-3p was demonstrated to significantly inhibit NO production compared to the control (p < 0.05). Moreover, transfection of emu-miR-4989-3p also gave rise to the increased expression of TNF-α (p < 0.01). Furthermore, emu-miR-4989-3p induced the dysregulation of several key components in the LPS/TLR4 signaling pathway compared with the control, especially TLR4 and NF-κB that both were upregulated. Conversely, the NO production and the expression of TNF-α, TLR4 and NF-κB tended to be increased and decreased in the mimics-transfected cells upon emu-miR-4989-3p low expression, respectively. These results suggest that emu-miR-4989-3p is one of 'virulence' factors encapsulated into the extracellular vesicles, potentially playing a role in the pathogenesis of E. multilocularis.

14.
Comp Immunol Microbiol Infect Dis ; 67: 101363, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31600681

RESUMO

Exosomes, secreted by various cells, are nanometer-scale vesicles with the functions in intercellular communication. To understand a role of exosomal miRNAs in the sheeppox virus infection, exosomes were isolated from sheeppox virus-infected sheep testicular cells 0 h, 24 h and 72 h post infection. The results of transmission electron microscopy and size distribution showed that all three exosome samples were spherical particles with negatively-stained membrane, ranging from 39 nm to 127 nm in diameter. A total of 106 known and 279 novel miRNAs were identified, and 78 known and 54 novel miRNAs were commonly detected in three exosome samples. Compared with the exosomes by the uninfected controls, a total of 34 known miRNAs were aberrantly expressed in the exosomes from infected cells. In agreement with the sequencing data, the expression of oar-miR-21 and oar-miR-10b was shown to be the highest in exosomes at 24 h after SPPV-infected, and the expression of oar-let-7f was the highest in exosomes at 72 h. Conversely, the expression of oar-let-7b and oar-miR-221 was significantly decreased 24 h and 72 h post infection compared with 0 h. The analysis results also revealed that differentially expressed miRNAs were mostly involved in an immune system process and stimulus response. These results provide rich data to further investigate a role of exosomal miRNAs in SPPV-host interactions.


Assuntos
Exossomos/genética , MicroRNAs/genética , MicroRNAs/isolamento & purificação , Infecções por Poxviridae/veterinária , Animais , Capripoxvirus/genética , Masculino , Ovinos , Doenças dos Ovinos/virologia , Testículo/citologia , Testículo/virologia
15.
Infect Genet Evol ; 75: 103985, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31362070

RESUMO

Infection of Cysticercus tenuicollis, the larval stage of Taenia hydatigena, is extensively found in sheep and pigs and jeopardizes the breeding and meat industry. miRNAs are a subclass of small noncoding regulatory RNAs and closely associated with the pathogenesis and biology of parasites. Here, using HiSeq sequencing we identified 49 known and 2 potential novel miRNAs in C. tenuicollis, of which both thy-miR-71 and -87 were predominant. Using RT-qPCR, 6 selected miRNAs were validated, and thy-miR-71 and -miR-87 were confirmed to be highly expressed, with the copy number of approximately 82,340 ±â€¯2079 and 19,580 ±â€¯609 per 1 ng total RNA, respectively. Similar to other cestodes, T. hydatigena was predicted to have two conserved miRNA clusters thy-miR-71/2c/2b and thy-miR-4989/277, and three members of the former were confirmed to reside sequentially within the genomic region of 253 bp by PCR. The current data provide us a valuable resource for further studies of a role of miRNAs in T. hydatigena biology and infection.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , Gado/parasitologia , MicroRNAs/genética , Taenia/genética , Animais , Cruzamento , Indústria Alimentícia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Família Multigênica , RNA de Helmintos/genética , Reação em Cadeia da Polimerase em Tempo Real , Sequenciamento Completo do Genoma
16.
Cancer Lett ; 438: 44-51, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30219505

RESUMO

Exosomes are phospholipid bilayer membrane-enclosed vesicles in a size from 30 to 150 nm, carrying a variety of active components, such as proteins, mRNA and miRNAs, and are involved in intercellular communication. Exosomes are released by almost all living cells and detected in various biological fluids. Viruses especially oncogenic viruses have been reported to influence the formation of virus-associated cancer through reshaping the tumor microenvironment via exosomes. In this review, a role of exosomes released by oncogenic virus-infected cells in promoting or inhibiting cancer formation is outlined. Moreover, the prospects and challenges of exosome applications in cancer therapies are critically discussed.


Assuntos
Exossomos/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Microambiente Tumoral/genética , Infecções Tumorais por Vírus/genética , Comunicação Celular/genética , Exossomos/metabolismo , Exossomos/virologia , Humanos , MicroRNAs/genética , Neoplasias/metabolismo , Neoplasias/virologia , Vírus Oncogênicos/fisiologia , RNA Mensageiro/genética , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/virologia
17.
Front Immunol ; 9: 546, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29616037

RESUMO

Nuclear factor kappa B (NF-κB) is a pluripotent and crucial dimer transcription factor that orchestrates various physiological and pathological processes, especially cell proliferation, inflammation, and cancer development and progression. NF-κB expression is transient and tightly regulated in normal cells, but it is activated in cancer cells. Recently, numerous studies have demonstrated microRNAs (miRNAs) play a vital role in the NF-κB signaling pathway and NF-κB-associated immune responses, radioresistance and drug resistance of cancer, some acting as inhibitors and the others as activators. Although it is still in infancy, targeting NF-κB or the NF-κB signaling pathway by miRNAs is becoming a promising strategy of cancer treatment.


Assuntos
MicroRNAs/genética , NF-kappa B/metabolismo , Neoplasias/metabolismo , Animais , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade/genética , Inflamação/genética , Terapia de Alvo Molecular , NF-kappa B/genética , Neoplasias/genética , Neoplasias/imunologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA