Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
PLoS Pathog ; 19(3): e1011295, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36972295

RESUMO

Calcium (Ca2+), a ubiquitous second messenger, plays a crucial role in many cellular functions. Viruses often hijack Ca2+ signaling to facilitate viral processes such as entry, replication, assembly, and egress. Here, we report that infection by the swine arterivirus, porcine reproductive and respiratory syndrome virus (PRRSV), induces dysregulated Ca2+ homeostasis, subsequently activating calmodulin-dependent protein kinase-II (CaMKII) mediated autophagy, and thus fueling viral replication. Mechanically, PRRSV infection induces endoplasmic reticulum (ER) stress and forms a closed ER-plasma membrane (PM) contacts, resulting the opening of store operated calcium entry (SOCE) channel and causing the ER to take up extracellular Ca2+, which is then released into the cytoplasm by inositol trisphosphate receptor (IP3R) channel. Importantly, pharmacological inhibition of ER stress or CaMKII mediated autophagy blocks PRRSV replication. Notably, we show that PRRSV protein Nsp2 plays a dominant role in the PRRSV induced ER stress and autophagy, interacting with stromal interaction molecule 1 (STIM1) and the 78 kDa glucose-regulated protein 78 (GRP78). The interplay between PRRSV and cellular calcium signaling provides a novel potential approach to develop antivirals and therapeutics for the disease outbreaks.


Assuntos
Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Animais , Suínos , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Sinalização do Cálcio , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Retículo Endoplasmático/metabolismo , Autofagia , Replicação Viral , Síndrome Respiratória e Reprodutiva Suína/metabolismo
2.
J Virol ; 97(10): e0104523, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37811994

RESUMO

IMPORTANCE: Senecavirus A (SVA) is an emerging picornavirus associated with vesicular disease, which wide spreads around the world. It has evolved multiple strategies to evade host immune surveillance. The mechanism and pathogenesis of the virus infection remain unclear. In this study, we show that SERPINB1, a member of the SERPINB family, promotes SVA replication, and regulates both innate immunity and the autophagy pathway. SERPINB1 catalyzes K48-linked polyubiquitination of IκB kinase epsilon (IKBKE) and degrades IKBKE through the proteasome pathway. Inhibition of IKBKE expression by SERPINB1 induces autophagy to decrease type I interferon signaling, and ultimately promotes SVA proliferation. These results provide importantly the theoretical basis of SVA replication and pathogenesis. SERPINB1 could be a potential therapeutic target for the control of viral infection.


Assuntos
Quinase I-kappa B , Picornaviridae , Serpinas , Replicação Viral , Autofagia , Quinase I-kappa B/genética , Imunidade Inata , Picornaviridae/fisiologia , Transdução de Sinais , Serpinas/genética , Interferon Tipo I
3.
J Virol ; 97(4): e0018823, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37039642

RESUMO

Stimulator of interferon (IFN) genes (STING) was recently pinpointed as an antiviral innate immune factor during the infection of RNA viruses. Porcine reproductive and respiratory syndrome virus (PRRSV), the swine arterivirus, is an enveloped RNA virus which has evolved many strategies to evade innate immunity. To date, the interactive network between PRRSV and STING remains to be fully established. Herein, we report that STING suppresses PRRSV replication through type I interferon signaling. However, PRRSV impedes STING trafficking from the endoplasmic reticulum (ER) to the Golgi apparatus, leading to the decreased phosphorylation of TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3). Furthermore, PRRSV nonstructural protein 2 (Nsp2) colocalizes with STING, blocks STING translocation, and disrupts the STING-TBK1-IRF3 complex. Mechanistically, PRRSV Nsp2 retains STING at the ER by increasing the level of Ca2+ sensor stromal interaction molecule 1 (STIM1) protein. Functional analysis reveals that PRRSV Nsp2 deubiquitinates STIM1 by virtue of its papain-like protease 2 (PLP2) deubiquitinating (DUB) activity. Finally, we demonstrate that loss of STIM1 is associated with an elevated IFN response and restricts PRRSV replication. This work delineates the relationship between PRRSV infection and STING signaling and the importance of papain-like proteases (PLPs) in interfering in this axis. IMPORTANCE Porcine reproductive and respiratory syndrome virus (PRRSV), a member of the family Arteriviridae, is responsible for reproductive disorders in pregnant sows and respiratory problems in piglets, resulting in huge losses in the swine industry worldwide. Of note, PRRSV infection causes immunosuppression, of which the mechanism is not completely understood. Here, we demonstrate for the first time that STING, a protein typically associated with the antiviral response in DNA viruses, plays a critical role in controlling PRRSV infection. However, PRRSV utilizes its encoded protein Nsp2 to inhibit STING activity by blocking its translocation from the ER to the Golgi apparatus. In particular, Nsp2 retains STING at the ER by interacting with and further deubiquitinating STIM1. For this process, the activity of the viral PLP2 DUB enzyme is indispensable. The study describes a novel mechanism by which PLP2 plays a critical role in suppressing the innate immune response against arteriviruses and potentially other viruses that encode similar proteases.


Assuntos
Proteínas de Membrana , Peptídeo Hidrolases , Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Molécula 1 de Interação Estromal , Animais , Feminino , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Papaína/metabolismo , Peptídeo Hidrolases/metabolismo , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Suínos , Proteínas não Estruturais Virais/metabolismo , Proteínas de Membrana/metabolismo , Imunidade Inata/imunologia , Ubiquitinação/fisiologia
4.
Vet Res ; 55(1): 67, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38783392

RESUMO

Porcine reproductive and respiratory syndrome (PRRS), caused by the PRRS virus (PRRSV), has caused substantial economic losses to the global swine industry due to the lack of effective commercial vaccines and drugs. There is an urgent need to develop alternative strategies for PRRS prevention and control, such as antiviral drugs. In this study, we identified ursonic acid (UNA), a natural pentacyclic triterpenoid from medicinal herbs, as a novel drug with anti-PRRSV activity in vitro. Mechanistically, a time-of-addition assay revealed that UNA inhibited PRRSV replication when it was added before, at the same time as, and after PRRSV infection was induced. Compound target prediction and molecular docking analysis suggested that UNA interacts with the active pocket of PTPN1, which was further confirmed by a target protein interference assay and phosphatase activity assay. Furthermore, UNA inhibited PRRSV replication by targeting PTPN1, which inhibited IFN-ß production. In addition, UNA displayed antiviral activity against porcine epidemic diarrhoea virus (PEDV) and Seneca virus A (SVA) replication in vitro. These findings will be helpful for developing novel prophylactic and therapeutic agents against PRRS and other swine virus infections.


Assuntos
Antivirais , Imunidade Inata , Vírus da Síndrome Respiratória e Reprodutiva Suína , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Triterpenos , Replicação Viral , Animais , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Antivirais/farmacologia , Suínos , Triterpenos/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Plantas Medicinais/química , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/virologia
5.
J Environ Manage ; 366: 121906, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39032258

RESUMO

Increased ecological land (IEL) such as forests and grasslands can greatly enhance ecosystem carbon sinks. Understanding the mechanisms for the magnitude of IEL-induced ecosystem carbon sinks is crucial for achieving carbon neutrality. We estimated the impact of IEL, specifically the increase in forests and grasslands, as well as global changes including atmospheric CO2 concentration, nitrogen deposition, and climate change on net ecosystem productivity (NEP) in National Key Ecological Function Zones (NKEFZs) in China using a calibrated ecological process model. The NEP in NKEFZs in China was calculated to be 119.4 Tg C yr-1, showing an increase of 42.6 Tg C yr-1 from 2001 to 2021. Compared to the slight contributions of climate change (-8.0%), nitrogen deposition (11.5%), and reduction in ecological land (-3.5%), the increase in NEP was primarily attributed to CO2 (66.5%) and IEL (33.5%). Moreover, the effect of IEL (14.8 Tg C yr-1) surpassed that of global change (13.1 Tg C yr-1) in the land use change zone. The IEL-induced NEP is significantly associated with CO2 fertilization, regulated by precipitation and nitrogen deposition. The high values of IEL-induced NEP occurred in areas with precipitation exceeding 800 mm and nitrogen deposition exceeding 25 kg N ha-1 yr-1. We recommend prioritizing the expansion of ecological land in areas with sufficient water and nutrients to enhance CO2 fertilization, while avoiding increasing ecological land in regions facing unfavorable climate change conditions. This study serves as a foundation for comprehending the NEP response to ecological restoration and global change.

6.
Vet Res ; 54(1): 124, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38124181

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) infection has caused huge economic losses in global swine industry over the last 37 years. PRRSV commercial vaccines are not effective against all epidemic PRRSV strains. In this study we performed a high-throughput screening (HTS) of an FDA-approved drug library, which contained 2339 compounds, and found vidofludimus (Vi) could significantly inhibits PRRSV replication in Marc-145 cells and primary porcine alveolar macrophages (PAMs). Compounds target prediction, molecular docking analysis, and target protein interference assay showed that Vi interacts with dihydroorotate dehydrogenase (DHODH), a rate-limiting enzyme in the de novo pyrimidine synthesis pathway. Furthermore, PRRSV infection was restored in the presence of excess uridine and cytidine which promote pyrimidine salvage, or excess orotate which is the product of DHODH in the de novo pyrimidine biosynthesis pathway, thus confirming that the antiviral effect of Vi against PRRSV relies on the inhibition of DHODH. In addition, Vi also has antiviral activity against Seneca virus A (SVA), encephalomyocarditis virus (EMCV), porcine epidemic diarrhea virus (PEDV), and pseudorabies virus (PRV) in vitro. These findings should be helpful for developing a novel prophylactic and therapeutic strategy against PRRSV and other swine viral infections.


Assuntos
Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Doenças dos Suínos , Animais , Suínos , Di-Hidro-Orotato Desidrogenase , Simulação de Acoplamento Molecular , Linhagem Celular , Replicação Viral/fisiologia , Antivirais/farmacologia , Antivirais/uso terapêutico , Pirimidinas/farmacologia
7.
J Virol ; 95(19): e0092321, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34260286

RESUMO

Peroxiredoxin 1 (PRDX1) is a cellular antioxidant enzyme that is crucial for diverse fundamental biological processes, such as autophagy, inflammation, and carcinogenesis. However, molecular mechanisms underpinning its diverse roles are not well understood. Here, we report that PRDX1 positively regulates interferon (IFN) induction and that pseudorabies virus (PRV) targets PRDX1 to evade IFN induction. PRV UL13 encodes a serine/threonine kinase important for PRV infection, although its biological function remains obscure. We identified PRDX1 as a UL13-interacting protein. Virological and biochemical assays demonstrate that PRDX1 promotes IFN induction by interacting with TANK-binding kinase 1 (TBK1) and IκB kinase ε (IKKε). Conversely, UL13 accelerates PRDX1 degradation via the ubiquitin-proteosome pathway in a kinase-dependent manner. In doing so, PRV inhibits IFN induction during productive infection, which requires PRDX1 expression. This study uncovers an essential role of PRDX1 in the innate immune response and reveals a new viral immune evasion strategy to counteract cellular defenses. IMPORTANCE PRV interacts with numerous cellular proteins during productive infection. Here, we demonstrated the interaction of viral protein UL13 with the antioxidant enzyme PRDX1, which functions in multiple signal transduction pathways. We found that PRDX1 participates in the type I IFN pathway by interacting with TBK1 and IKKε, thereby negatively regulating PRV propagation. However, UL13 ubiquitinates PRDX1, which routes PRDX1 into proteasomes for degradation and effectively reduces its expression. These results illuminate the fundamental role that PRDX1 plays in the IFN pathway, and they identify a potential target for the control of PRV infection.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Quinase I-kappa B/metabolismo , Imunidade Inata , Peroxirredoxinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Células HEK293 , Herpesvirus Suídeo 1/imunologia , Humanos , Evasão da Resposta Imune , Interferon Tipo I/biossíntese , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Ubiquitinação , Proteínas Virais/genética , Replicação Viral
8.
J Virol ; 95(18): e0021021, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34160254

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important endemic swine pathogens, causing enormous losses in the global swine industry. Commercially available vaccines only partially prevent or counteract the virus infection and correlated losses. PRRSV's replication mechanism has not been well understood. In this study, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was screened to bind with the viral major envelope glycoprotein 5 (GP5) after PRRSV infection. The interacting sites are located within a 13-amino-acid (aa) region (aa 93 to 105) of GP5 and at Lys227 of GAPDH. Interestingly, viral GP5 restricts the translocation of GAPDH from the cytoplasm to the nucleus. Moreover, cytoplasmic GAPDH facilitates PRRSV replication by virtue of its glycolytic activity. The results suggest that PRRSV GP5 restricts GAPDH to the nucleus and exploits its glycolytic activity to stimulate virus replication. The data provide insight into the role of GAPDH in PRRSV replication and reveal a potential target for controlling viral infection. IMPORTANCE PRRSV poses a severe economic threat to the pig industry. PRRSV GP5, the major viral envelope protein, plays an important role in viral infection, pathogenicity, and immunity. However, interactions between GP5 and host proteins have not yet been well studied. Here, we show that GAPDH interacts with GP5 through binding a 13-aa sequence (aa 93 to 105) in GP5, while GP5 interacts with GAPDH at the K277 amino acid residue of GAPDH. We demonstrate that GP5 interacts with GAPDH in the cytoplasm during PPRSV infection, inhibiting GAPDH entry into the nucleus. PRRSV exploits the glycolytic activity of GAPDH to promote viral replication. These results enrich our understanding of PRRSV infection and pathogenesis and open a new avenue for antiviral prevention and PRRSV treatment strategies.


Assuntos
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Animais , Gliceraldeído-3-Fosfato Desidrogenases/genética , Células HEK293 , Humanos , Síndrome Respiratória e Reprodutiva Suína/genética , Síndrome Respiratória e Reprodutiva Suína/metabolismo , Suínos , Proteínas do Envelope Viral/genética
9.
Microb Pathog ; 168: 105607, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35640766

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) is an important pathogen causing huge economic losses to the swine industry worldwide, because of its rapid evolution and variation in genomes. It is necessary to monitor the newly emerging epidemic strains and compare their virulence diversity in swine. In this study, two strains of PRRSV2 were isolated from clinical samples by using primary porcine alveolar macrophage (PAMs) cells and designated as SD1805 and FJ1805, respectively. They were unable to grow in MARC-145 cells. Full-length genomes sequence analysis revealed that SD1805 strain has the molecular characterization of NADC30 strain from American, and belongs to the branch of NADC30-like PRRSV (NL-PRRSV) (lineagea 1). FJ1805 isolate came from the inter-subgenotype recombinant of NADC30 strain with a highly pathogenic PRRSV (HP-PRRSV) strain HUN4. Moreover, it belongs to a lineagea 3 represented by the Chinese isolate QYYZ. Pathogenicity analysis showed that SD1805 strain had similar mortality and viral loads in lungs to HP-PRRSV BB0907 strain. FJ1805 strain showed milder pathogenicity compared to NL-PRRSV strain FJ1402 that was previously isolated with Marc-145 cells. It provides evidence of the circulation of the different subgenotype PRRSV strains in China with variations in cell adaption and pathogenic abilities.


Assuntos
Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Sequência de Aminoácidos , Animais , China/epidemiologia , Genoma Viral , Filogenia , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Suínos , Virulência
10.
PLoS Pathog ; 15(1): e1007239, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668592

RESUMO

Chikungunya virus (CHIKV) is a re-emerging Alphavirus causing fever, joint pain, skin rash, arthralgia, and occasionally death. Antiviral therapies and/or effective vaccines are urgently required. CHIKV biology is poorly understood, in particular the functions of the non-structural protein 3 (nsP3). Here we present the results of a mutagenic analysis of the alphavirus unique domain (AUD) of nsP3. Informed by the structure of the Sindbis virus AUD and an alignment of amino acid sequences of multiple alphaviruses, a series of mutations in the AUD were generated in a CHIKV sub-genomic replicon. This analysis revealed an essential role for the AUD in CHIKV RNA replication, with mutants exhibiting species- and cell-type specific phenotypes. To test if the AUD played a role in other stages of the virus lifecycle, the mutants were analysed in the context of infectious CHIKV. This analysis indicated that the AUD was also required for virus assembly. In particular, one mutant (P247A/V248A) exhibited a dramatic reduction in production of infectious virus. This phenotype was shown to be due to a block in transcription of the subgenomic RNA leading to reduced synthesis of the structural proteins and a concomitant reduction in virus production. This phenotype could be further explained by both a reduction in the binding of the P247A/V248A mutant nsP3 to viral genomic RNA in vivo, and the reduced affinity of the mutant AUD for the subgenomic promoter RNA in vitro. We propose that the AUD is a pleiotropic protein domain, with multiple functions during CHIKV RNA synthesis.


Assuntos
Vírus Chikungunya/genética , Proteínas não Estruturais Virais/metabolismo , Alphavirus/genética , Alphavirus/metabolismo , Febre de Chikungunya/genética , Febre de Chikungunya/metabolismo , Vírus Chikungunya/metabolismo , Genoma Viral , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Domínios Proteicos , Replicon , Proteínas não Estruturais Virais/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia , Vírus/genética
11.
Vet Res ; 52(1): 52, 2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33766129

RESUMO

Long non-coding RNAs (lncRNAs) are a new arm of gene regulatory mechanism as discovered by sequencing techniques and follow-up functional studies. The lncRNAs regulation of pseudorabies virus (PRV) infection has rarely been reported so far. Using RNA sequencing analysis, 225 lncRNAs with significant altered expressions in 3D4/21 cells infected with PRV (ZJ01) were identified. Five lncRNAs upregulated in PRV-infected cells were verified in cells infected with different PRV strains by qRT-PCR. By down- and up-regulation of lnc641, the accelerating effect of lnc641 on PRV replication was confirmed. Furthermore, we found that lnc641 regulated PRV replication by inhibiting the JAK-STAT1 pathway. This study suggests that lnc641 could be a new host factor target for developing antiviral therapies against PRV infection.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Suídeo 1/fisiologia , RNA Longo não Codificante/genética , RNA Viral/genética , Replicação Viral/genética , Animais , Linhagem Celular , Herpesvirus Suídeo 1/genética , RNA Longo não Codificante/metabolismo , RNA Viral/metabolismo , Sus scrofa
12.
J Virol ; 92(16)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29875241

RESUMO

Chikungunya virus (CHIKV), a mosquito-borne human pathogen, causes a disabling disease characterized by severe joint pain that can persist for weeks, months, or even years in patients. The nonstructural protein 3 (nsP3) plays essential roles during acute infection, but little is known about the function of nsP3 during chronic disease. Here, we used subdiffraction multicolor microscopy for spatial and temporal analysis of CHIKV nsP3 within human cells that persistently replicate replicon RNA. Round cytoplasmic granules of various sizes (i) contained nsP3 and stress granule assembly factors 1 and 2 (G3BP1/2), (ii) were next to double-stranded RNA foci and nsP1-positive structures, and (iii) were close to the nuclear membrane and the nuclear pore complex protein Nup98. Analysis of protein turnover and mobility by live-cell microscopy revealed that the granules could persist for hours to days, accumulated newly synthesized protein, and moved through the cytoplasm at various speeds. The granules also had a static internal architecture and were stable in cell lysates. Refractory cells that had cleared the noncytotoxic replicon regained the ability to respond to arsenite-induced stress. In summary, nsP3 can form uniquely stable granular structures that persist long-term within the host cell. This continued presence of viral and cellular protein complexes has implications for the study of the pathogenic consequences of lingering CHIKV infection and the development of strategies to mitigate the burden of chronic musculoskeletal disease brought about by a medically important arthropod-borne virus (arbovirus).IMPORTANCE Chikungunya virus (CHIKV) is a reemerging alphavirus transmitted by mosquitos and causes transient sickness but also chronic disease affecting muscles and joints. No approved vaccines or antivirals are available. Thus, a better understanding of the viral life cycle and the role of viral proteins can aid in identifying new therapeutic targets. Advances in microscopy and development of noncytotoxic replicons (A. Utt, P. K. Das, M. Varjak, V. Lulla, A. Lulla, A. Merits, J Virol 89:3145-3162, 2015, https://doi.org/10.1128/JVI.03213-14) have allowed researchers to study viral proteins within controlled laboratory environments over extended durations. Here we established human cells that stably replicate replicon RNA and express tagged nonstructural protein 3 (nsP3). The ability to track nsP3 within the host cell and during persistent replication can benefit fundamental research efforts to better understand long-term consequences of the persistence of viral protein complexes and thereby provide the foundation for new therapeutic targets to control CHIKV infection and treat chronic disease symptoms.


Assuntos
Vírus Chikungunya/fisiologia , Grânulos Citoplasmáticos/química , Proteínas não Estruturais Virais/análise , Replicação Viral , Humanos , Análise Espaço-Temporal
13.
J Virol ; 92(1)2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29070685

RESUMO

Chicken Na+/H+ exchanger type I (chNHE1), a multispan transmembrane protein, is a cellular receptor of the subgroup J avian leukosis virus (ALV-J). To identify the functional determinants of chNHE1 responsible for the ALV-J receptor activity, a series of chimeric receptors was created by exchanging the extracellular loops (ECL) of human NHE1 (huNHE1) and chNHE1 and by ECL replacement with a hemagglutinin (HA) tag. These chimeric receptors then were used in binding and entry assays to map the minimal ALV-J gp85-binding domain of chNHE1. We show that ECL1 of chNHE1 (chECL1) is the critical functional ECL that interacts directly with ALV-J gp85; ECL3 is also involved in ALV-J gp85 binding. Amino acid residues 28 to 39 of the N-terminal membrane-proximal region of chECL1 constitute the minimal domain required for chNHE1 binding of ALV-J gp85. These residues are sufficient to mediate viral entry into ALV-J nonpermissive cells. Point mutation analysis revealed that A30, V33, W38, and E39 of chECL1 are the key residues mediating the binding between chNHE1 and ALV-J gp85. Further, the replacement of residues 28 to 39 of huNHE1 with the corresponding chNHE1 residues converted the nonfunctional ALV-J receptor huNHE1 to a functional one. Importantly, soluble chECL1 and huECL1 harboring chNHE1 residues 28 to 39 both could effectively block ALV-J infection. Collectively, our findings indicate that residues 28 to 39 of chNHE1 constitute a domain that is critical for receptor function and mediate ALV-J entry.IMPORTANCE chNHE1 is a cellular receptor of ALV-J, a retrovirus that causes infections in chickens and serious economic losses in the poultry industry. Until now, the domains determining the chNHE1 receptor function remained unknown. We demonstrate that chECL1 is critical for receptor function, with residues 28 to 39 constituting the minimal functional domain responsible for chNHE1 binding of ALV-J gp85 and efficiently mediating ALV-J cell entry. These residues are located in the membrane-proximal region of the N terminus of chECL1, suggesting that the binding site of ALV-J gp85 on chNHE1 is probably located on the apex of the molecule; the receptor-binding mode might be different from that of retroviruses. We also found that soluble chECL1, as well as huECL1 harboring chNHE1 residues 28 to 39, effectively blocked ALV-J infection. These findings contribute to a better understanding of the ALV-J infection mechanism and also provide new insights into the control strategies for ALV-J infection.


Assuntos
Aminoácidos/química , Vírus da Leucose Aviária/metabolismo , Receptores Virais/metabolismo , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/metabolismo , Ligação Viral , Internalização do Vírus , Aminoácidos/metabolismo , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Galinhas , Humanos , Mutação Puntual , Receptores Virais/genética , Trocadores de Sódio-Hidrogênio/genética
14.
J Gen Virol ; 96(Pt 1): 150-158, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25274857

RESUMO

Avian leukosis virus subgroup J (ALV-J) was first isolated from meat-producing chickens that had developed myeloid leukosis. However, ALV-J infections associated with hemangiomas have occurred in egg-producing (layer) flocks in China. In this study, we identified an ALV-J layer isolate (HLJ13SH01) as a recombinant of ALV-J and a Rous sarcoma virus Schmidt-Ruppin B strain (RSV-SRB), which contained the RSV-SRB 5'-LTR and the other genes of ALV-J. Replication kinetic testing indicated that the HLJ13SH01 strain replicated faster than other ALV-J layer isolates in vitro. Sequence analysis indicated that the main difference between the two isolates was the 5'-LTR sequences, particularly the U3 sequences. A 19 nt insertion was uniquely found in the U3 region of the HLJ13SH01 strain. The results of a Dual-Glo luciferase assay revealed that the 19 nt insertion in the HLJ13SH01 strain increased the enhancer activity of the U3 region. Moreover, an additional CCAAT/enhancer element was found in the 19 nt insertion and the luciferase assay indicated that this element played a key role in increasing the enhancer activity of the 5'-U3 region. To confirm the potentiation effect of the 19 nt insertion and the CCAAT/enhancer element on virus replication, three infectious clones with 5'-U3 region variations were constructed and rescued. Replication kinetic testing of the rescued viruses demonstrated that the CCAAT/enhancer element in the 19 nt insertion enhanced the replication capacity of the ALV-J recombinant in vitro.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas/virologia , Replicação do DNA/genética , Doenças das Aves Domésticas/virologia , Vírus do Sarcoma de Rous/genética , Replicação Viral/genética , Animais , China
15.
Arch Virol ; 160(10): 2445-53, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26175070

RESUMO

The entry of enveloped viruses into host cells requires the fusion of viral and cell membranes. These membrane fusion reactions are mediated by virus-encoded glycoproteins. In the case of avian metapneumovirus (aMPV), the fusion (F) protein alone can mediate virus entry and induce syncytium formation in vitro. To investigate the fusogenic activity of the aMPV F protein, we compared the fusogenic activities of three subtypes of aMPV F proteins using a TCSD50 assay developed in this study. Interestingly, we found that the F protein of aMPV subtype B (aMPV/B) strain VCO3/60616 (aMPV/vB) was hyperfusogenic when compared with F proteins of aMPV/B strain aMPV/f (aMPV/fB), aMPV subtype A (aMPV/A), and aMPV subtype C (aMPV/C). We then further demonstrated that the amino acid (aa) residue 149F contributed to the hyperfusogenic activity of the aMPV/vB F protein. Moreover, we revealed that residue 149F had no effect on the fusogenic activities of aMPV/A, aMPV/C, and human metapneumovirus (hMPV) F proteins. Collectively, we provide the first evidence that the amino acid at position 149 affects the fusogenic activity of the aMPV/B F protein, and our findings will provide new insights into the fusogenic mechanism of this protein.


Assuntos
Variação Genética , Metapneumovirus/genética , Infecções por Paramyxoviridae/veterinária , Doenças das Aves Domésticas/virologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Humanos , Metapneumovirus/química , Metapneumovirus/classificação , Metapneumovirus/metabolismo , Dados de Sequência Molecular , Infecções por Paramyxoviridae/virologia , Alinhamento de Sequência , Perus/virologia , Células Vero , Proteínas Virais de Fusão/genética
16.
Arch Virol ; 160(4): 995-1004, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25655260

RESUMO

Avian leukosis virus subgroup J (ALV-J) is an avian oncogenic retrovirus that has caused severe economic losses in China. Gp85 protein is the main envelope protein and the most variable structural protein of ALV-J. It is also involved in virus neutralization. In this study, a specific monoclonal antibody, 4A3, was produced against the ALV-J gp85 protein. Immunofluorescence assays showed that 4A3 could react with different strains of ALV-J, including the British prototype isolate HPRS103, the American strains, an early Chinese broiler isolate, and layer isolates. A linear epitope on the gp85 protein was identified using a series of partially overlapping fragments spanning the gp85-encoding gene and subjecting them to western blot analysis. The results indicated that (134)AEAELRDFI(142) was the minimal linear epitope that could be recognized by mAb 4A3. Enzyme-linked immunosorbent assay (ELISA) revealed that chicken anti-ALV-J sera and mouse anti-ALV-J gp85 sera could also recognize the minimal linear epitope. Alignment analysis of amino acid sequences indicated that the epitope was highly conserved among 34 ALV-J strains. Furthermore, the epitope was not conserved among subgroup A and B of avian leukosis virus (ALV). Taken together, the mAb and the identified epitope may provide valuable tools for the development of new diagnostic methods for ALV-J.


Assuntos
Vírus da Leucose Aviária/imunologia , Leucose Aviária/virologia , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Doenças das Aves Domésticas/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Leucose Aviária/imunologia , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/isolamento & purificação , Galinhas , Mapeamento de Epitopos , Epitopos de Linfócito B/genética , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Doenças das Aves Domésticas/imunologia , Especificidade da Espécie , Proteínas do Envelope Viral/genética
17.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 32(3): 537-41, 2015 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-26485974

RESUMO

Multi-layer perceptron (MLP) neural network belongs to multi-layer feedforward neural network, and has the ability and characteristics of high intelligence. It can realize the complex nonlinear mapping by its own learning through the network. Bipolar disorder is a serious mental illness with high recurrence rate, high self-harm rate and high suicide rate. Most of the onset of the bipolar disorder starts with depressive episode, which can be easily misdiagnosed as unipolar depression and lead to a delayed treatment so as to influence the prognosis. The early identifica- tion of bipolar disorder is of great importance for patients with bipolar disorder. Due to the fact that the process of early identification of bipolar disorder is nonlinear, we in this paper discuss the MLP neural network application in early identification of bipolar disorder. This study covered 250 cases, including 143 cases with recurrent depression and 107 cases with bipolar disorder, and clinical features were statistically analyzed between the two groups. A total of 42 variables with significant differences were screened as the input variables of the neural network. Part of the samples were randomly selected as the learning sample, and the other as the test sample. By choosing different neu- ral network structures, all results of the identification of bipolar disorder were relatively good, which showed that MLP neural network could be used in the early identification of bipolar disorder.


Assuntos
Transtorno Bipolar/diagnóstico , Redes Neurais de Computação , Humanos
18.
J Clin Microbiol ; 52(1): 37-44, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24131697

RESUMO

Avian leukosis virus (ALV) subgroups A, B, and J are very common in poultry flocks and have caused serious economic losses in recent years. A multiplex PCR (mPCR) method for the detection of these three subgroups was developed and optimized in this study. We first designed a common forward primer, PF, and three downstream primers, AR, BR, and JR, which can amplify 715 bp for subgroup A, 515 bp for subgroup B, and 422 bp for subgroup J simultaneously in one reaction. The mPCR method produced neither cross-reactions with other subgroups of ALVs nor nonspecific reactions with other common avian viruses. The detection limit of the mPCR was as low as 1 × 10(3) viral DNA copies of each of the three subgroups. In animal experiments, the mPCR detected ALVs 2 to 4 days earlier than did virus isolation from whole-blood samples and cloaca swabs. Furthermore, a total of 346 clinical samples (including 127 tissue samples, 86 cloaca swabs, 59 albumen samples, and 74 whole-blood samples) from poultry flocks with suspected ALV infection were examined by mPCR, routine PCR, and virus isolation. The positive sample/total sample ratios for ALV-A, ALV-B, and ALV-J were 48% (166/346) as detected by mPCR and 48% (166/346) as detected by routine PCR. However, the positive sample/total sample ratio detected by virus isolation was 40% (138/346). The results of the mPCR and routine PCR were confirmed by sequencing the specific fragments. These results indicate that the mPCR method is rapid, specific, sensitive, and convenient for use in epidemiological studies of ALV, clinical detection of ALV, and ALV eradication programs.


Assuntos
Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/diagnóstico , Leucose Aviária/virologia , Técnicas de Diagnóstico Molecular/métodos , Reação em Cadeia da Polimerase Multiplex/métodos , Medicina Veterinária/métodos , Animais , Vírus da Leucose Aviária/genética , Epidemiologia Molecular/métodos , Aves Domésticas , Sensibilidade e Especificidade , Fatores de Tempo
19.
Gels ; 10(6)2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38920936

RESUMO

Effective forest fire suppression remains a critical challenge, necessitating innovative solutions. Temperature-sensitive hydrogels represent a promising avenue in this endeavor. Traditional firefighting methods often struggle to address forest fires efficiently while mitigating ecological harm and optimizing resource utilization. In this study, a novel intelligent temperature-sensitive hydrogel was prepared specially for forest fire extinguishment. Utilizing a one-pot synthesis approach, this material demonstrates exceptional fluidity at ambient temperatures, facilitating convenient application and transport. Upon exposure to elevated temperatures, it undergoes a phase transition to form a solid, barrier-like structure essential for containing forest fires. The incorporation of environmentally friendly phosphorus salts into the chitosan/hydroxypropyl methylcellulose gel system enhances the formation of temperature-sensitive hydrogels, thereby enhancing their structural integrity and firefighting efficacy. Morphological and thermal stability analyses elucidate the outstanding performance, with the hydrogel forming a dense carbonized layer that acts as a robust barrier against the spread of forest fires. Additionally, comprehensive evaluations employing rheological tests, cone calorimeter tests, a swelling test, and infrared thermography reveal the multifaceted roles of temperature-sensitive hydrogels in forest fire prevention and suppression strategies.

20.
Polymers (Basel) ; 16(13)2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-39000650

RESUMO

A novel self-gelatinizing powder was designed to accelerate wound healing through enhanced hemostasis and tissue recovery. Significantly, this research addresses the critical need for innovative wound management solutions by presenting a novel approach. Carboxymethylcellulose calcium (CMC-Ca) was synthesized using an ion exchange method, and lysine (Lys) was integrated through physical mixing to augment the material's functional characteristics. The prepared powder underwent comprehensive evaluation for its self-gelling capacity, gelation time, adhesion, swelling rate, coagulation efficiency, hemostatic effectiveness, and wound healing promotion. Results indicate that the self-gelatinizing powder exhibited remarkable water absorption capabilities, absorbing liquid up to 30 times its weight and achieving rapid coagulation within 3 min. The inclusion of Lys notably enhanced the powder's gel-forming properties. The gelation time was determined to be within 4 s using a rotational rheometer, with the powder rapidly forming a stable gel on the skin surface. Furthermore, in a mouse skin injury model, near-complete skin recovery was observed within 14 days, underscoring the powder's impressive self-healing attributes and promising application prospects in wound management.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA