Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vet Microbiol ; 288: 109932, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38043447

RESUMO

Bovine herpesvirus 1(BoHV-1) is an important bovine pathogen that causes great economic loss to cattle farms worldwide. The virus-productive infection in bovine kidney (MDBK) cells results in ATP depletion. The mechanisms are not well understood. Mitochondrial fatty acid ß-oxidation (FAO) is an important energy source in many tissues with high energy demand. Since carnitine palmitoyl-transferase 1 A (CPT1A) is the rate-limiting enzyme of FAO, we investigated the interactions between virus-productive infection and CPT1A signaling. Here, we found that virus-productive infection at the later stage significantly decreased CPT1A protein levels in all the detected cells, including MDBK, A549, and Neuro-2A cells, differentially altered the accumulation of CPT1A proteins in the nucleus and cytosol, and re-localized the protein in the nucleus. Etomoxir (ETO), an irreversible inhibitor of CPT1A, inhibited viral replication and partially interfered with the ability of BoHV-1 to alter CPT1A accumulation in the nucleus but not in the cytosol. Furthermore, ETO consistently reduced RNA levels of two viral regulatory proteins (bICP0 and bICP22) and protein expression of virion-associated proteins during productive infection, further supporting the important roles of CPT1A signaling in BoHV-1 productive infection. These data, for the first time, suggest that CPT1A is potentially involved in BoHV-1 productive infection.


Assuntos
Doenças dos Bovinos , Infecções por Herpesviridae , Herpesvirus Bovino 1 , Bovinos , Animais , Herpesvirus Bovino 1/genética , Replicação Viral , Infecções por Herpesviridae/veterinária , Transferases/metabolismo , Carnitina/metabolismo
2.
ACS Appl Mater Interfaces ; 15(41): 48601-48612, 2023 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-37787638

RESUMO

Chameleons are famous for their quick color changing abilities, and it is commonly assumed that they do this for camouflage. However, recent reports revealed that chameleons also change color for body temperature regulation. Inspired by the structure of the panther chameleon's skin, a stripe-patterned poly(N-isopropylacrylamide) (PNIPAM) and polyacrylamide (PAM) hydrogel film with a laminated structure is fabricated in this work; thus, both camouflage and thermoregulation can be achieved through controlling Vis and NIR light effectively. For the PNIPAM stripe, the upper layer is the native PNIPAM hydrogel and the lower layer is the carbon nanotube-composited PNIPAM hydrogel. Thus, the PNIPAM stripe is capable of reaching 28 °C at a low environmental temperature (12 °C) and a low radiation intensity (20 mW cm-2), while preventing the body temperature from rising by changing to white under a strong radiation intensity (100 mW cm-2). For the PAM stripe, the upper layer combines colloidal photonic crystals and displays a tunable structural color by stretching, and the lower layer is mixed with PNIPAM microgels for thermal regulation. Through the fabrication of multifunctional patterns, the film can achieve both dynamic structural color and thermoregulation by precisely controlling solar radiation absorption, scattering, and reflection. More importantly, in the stripe-patterned system, the shrinkage of the PNIPAM stripes can effectively trigger the elongation of the PAM stripe, which endows the structural color changing process to be self-powered completely. The performances show that the stripe-patterned film may have potential applications in intelligent coatings, especially in areas with large temperature differences during the day such as high plains.


Assuntos
Pele Artificial , Hidrogéis , Luz , Temperatura , Regulação da Temperatura Corporal
3.
Microbiol Spectr ; : e0196323, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37655900

RESUMO

Phospholipase C gamma 1 (PLC-γ1) may locate at distinct subcellular locations, such as cytosol, plasma membrane, and nucleus for varied biological functions. Bovine herpesvirus 1 (BoHV-1) productive infection activates PLC-γ1 signaling, as demonstrated by increased protein levels of phosphorylated-PLC-γ1 at Ser1248 [p-PLC-γ1(S1248)], which benefits virus productive infection. Here, for the first time, we reported that Golgi apparatus also contains activated p-PLC-γ1(S1248). And BoHV-1 productive infection at later stages (24 hpi) increased the accumulation of p-PLC-γ1(S1248) in the Golgi apparatus, where p-PLC-γ1(S1248) forms highlighted puncta observed via a confocal microscope. Coimmunoprecipitation studies demonstrated that the Golgi p-PLC-γ1(S1248) is specifically associated with the viral protein gD but not gC. In addition, we found that p-PLC-γ1(S1248) is consistently associated with both the plasma membrane-associated virions and the released virions. When the virus-infected cells were treated with PLC-γ1-specific inhibitor, U73122, for a short duration of 4 hours prior to the endpoint of virus infection, we found that the viral protein gD was trapped in the Golgi apparatus, suggesting that the PLC-γ1 signaling may facilitate trafficking of progeny virions out of this organelle. These findings provide a novel insight into the interplay between PLC-γ1 signaling and BoHV-1 replication. IMPORTANCE Bovine herpesvirus 1 (BoHV-1) productive infection increases protein levels of phosphorylated-phospholipase C gamma 1 at Ser1248 [p-PLC-γ1(S1248)]. However, whether it causes any variations to p-PLC-γ1(S1248) localization is not well understood. Here, for the first time, we found that partial p-PLC-γ1(S1248) is residing in the Golgi apparatus, where the accumulation is enhanced by virus infection. p-PLC-γ1(S1248) is consistently associated with virions, partially via binding to gD, in both the Golgi apparatus and cytoplasm membranes. Surprisingly, it also associates with the released virions. Of note, this is the first evidenced BoHV-1 virion-bound host protein. It seems that p-PLC-γ1(S1248) works as an escort during trafficking of progeny virions out of Golgi apparatus to the plasma membranes as well as releasing outside of the cell membranes. Furthermore, we showed that the activated p-PLC-γ1(S1248) is potentially implicated in the transport of virions out of Golgi apparatus, which may represent a novel mechanism to regulate virus productive infection.

4.
Microbiol Spectr ; 11(4): e0011723, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37227295

RESUMO

Bovine herpesvirus 1 (BoHV-1), an important bovine viral pathogen, causes severe disease in the upper respiratory tract and reproductive system. Tonicity-responsive enhancer-binding protein (TonEBP), also known as nuclear factor of activated T cells 5 (NFAT5), is a pleiotropic stress protein involved in a range of cellular processes. In this study, we showed that the knockdown of NFAT5 by siRNA increased BoHV-1 productive infection and overexpression of NFAT5 via plasmid transfection decreased virus production in bovine kidney (MDBK) cells. Virus productive infection at later stages significantly increased transcription of NFAT5 but not appreciably alter measurable NFAT5 protein levels. Virus infection relocalized NFAT5 protein and decreased the cytosol accumulation. Importantly, we found a subset of NFAT5 resides in mitochondria, and virus infection led to the depletion of mitochondrial NFAT5. In addition to full-length NFAT5, another two isoforms with distinct molecular weights were exclusively detected in the nucleus, where the accumulation was differentially affected following virus infection. In addition, virus infection differentially altered mRNA levels of PGK1, SMIT, and BGT-1, the canonical downstream targets regulated by NFAT5. Taken together, NFAT5 is a potential host factor that restricts BoHV-1 productive infection, and virus infection hijacks NFAT5 signaling transduction by relocalization of NFAT5 molecules in cytoplasm, nucleus, and mitochondria, as well as altered expression of its downstream targets. IMPORTANCE Accumulating studies have revealed that NFAT5 regulates disease development due to infection of numerous viruses, underlying the importance of the host factor in virus pathogenesis. Here, we report that NFAT5 has capacity to restrict BoHV-1 productive infection in vitro. And virus productive infection at later stages may alter NFAT5 signaling pathway as observed by relocalization of NFAT5 protein, reduced accumulation of NFAT5 in cytosol, and differential expression of NFAT5 downstream targets. Importantly, for the first time, we found that a subset of NFAT5 resides in mitochondria, implying that NFAT5 may regulate mitochondrial functions, which will extend our knowledge on NFAT5 biological activities. Moreover, we found two NFAT5 isoforms with distinct molecular weights were exclusively detected in the nucleus, where the accumulation was differentially affected following virus infection, representing a novel regulation mechanism on NFAT5 function in response to BoHV-1infection.


Assuntos
Infecções por Herpesviridae , Herpesvirus Bovino 1 , Humanos , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/metabolismo , Fatores de Transcrição NFATC/metabolismo , Citoplasma/metabolismo , Núcleo Celular/metabolismo , Técnicas de Cultura de Células , Fatores de Transcrição/metabolismo
5.
Vet Microbiol ; 276: 109626, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36502739

RESUMO

Bovine herpesvirus 1 (BoHV-1) is a significant risk factor for the bovine respiratory disease complex (BRDC), a severe disease causing great economic losses to the cattle industry worldwide. Previous studies have reported that both phospholipase C-γ1 (PLC-γ1) and ß-catenin are activated during BoHV-1 infection for efficient replication. However, the interplay between PLC-γ1 and ß-catenin as a consequence of virus infection remains to be elucidated. Here, we reported that PLC-γ1 interacted with ß-catenin, which was enhanced following virus infection. PLC-γ1-specific inhibitor, U73122, significantly reduced the mRNA levels of ß-catenin in BoHV-1-infected cells; however, the steady-state protein levels were not affected due to the virus infection. Interestingly, the treatment of virus-infected cells with U73122 reduced the accumulation of activated ß-catenin [p-ß-catenin(S552)] in fractions of the cytoplasmic membrane as that observed with the treatment of methyl-ß-cyclodextrin (MßCD), which can disrupt cytoplasmic membrane structure via sequestering cholesterol. Nucleus accumulation of p-ß-catenin(S552) was increased following U73122 treatment in virus-infected cells. In addition, the association of p-ß-catenin(S552) with cytoplasmic membrane induced by the virus infection was significantly disrupted by the treatment of U73122 and MßCD. These data indicated that the PLC-γ1 signaling is potentially involved in the regulation of ß-catenin signaling stimulated by BoHV-1 infection partially via affecting the subcellular localization of p-ß-catenin(S552).


Assuntos
Doenças dos Bovinos , Infecções por Herpesviridae , Herpesvirus Bovino 1 , Bovinos , Animais , beta Catenina/metabolismo , Herpesvirus Bovino 1/fisiologia , Transdução de Sinais , Membrana Celular , Infecções por Herpesviridae/veterinária , Fosfolipases Tipo C/metabolismo , Fosfolipase C gama/metabolismo
6.
Biomedicines ; 10(9)2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36140380

RESUMO

Bovine herpesvirus 1 (BoHV-1), an important pathogen of cattle, is also a promising oncolytic virus. Recent studies have demonstrated that the virus infection induces DNA damage and DNA damage response (DDR), potentially accounting for virus infection-induced cell death and oncolytic effects. However, whether the global DDR network affects BoHV-1 productive infection remains to be elucidated. In this study, we show that global DDR induced by ultraviolet (UV) irradiation prior to BoHV-1 infection differentially affected transcription of immediate early (IE) genes, such as infected cell protein 0 (bICP0) and bICP22, in a cell-type-dependent manner. In addition, UV-induced DDR may affect the stabilization of viral protein levels, such as glycoprotein C (gC) and gD, because the variation in mRNA levels of gC and gD as a consequence of UV treatment were not in line with the variation in individual protein levels. The virus productive infection also affects UV-primed DDR signaling, as demonstrated by the alteration of phosphorylated histone H2AX (γH2AX) protein levels and γH2AX formation following virus infection. Taken together, for the first time, we evidenced the interplay between UV-primed global DDR and BoHV-1 productive infection. UV-primed global DDR differentially modulates the transcription of virus genes and stabilization of virus protein. Vice versa, the virus infection may affect UV-primed DDR signaling.

7.
Rev Med Virol ; 32(6): e2394, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36069169

RESUMO

Herpes simplex virus type 1 (HSV-1) is an important human pathogen with neurotropism. Following lytic infection in mucosal or skin epithelium, life-long latency is established mainly in sensory neurons, which can periodically reactivate by stress, leading to recurrent disease and virus transmission. During the virus's productive infection, the tegument protein VP16, a component of HSV-1 virion, is physically associated with two cellular factors, host cell factor-1 (HCF-1), and POU domain protein Oct-1, to construct the VP16-induced complex, which is essential to stimulate immediate early (IE)-gene transcription as well as initiate the lytic programme. Apart from HCF-1 and Oct-1, VP16 also associates with a series of other host factors, making a VP16-induced regulatory switch to either activate or inactivate virus gene transcription. In addition, VP16 has effects on distinct signalling pathways via binding to various host molecules that are essentially related to innate immune responses, RNA polymerases, molecular chaperones, and virus infection-induced host shutoff. VP16 also functionally compensates for given host factors, such as PPAR-γ and ß-catenin. In this review, we provide an overview of the updated insights on the interplay between VP16 and the host factors that coordinate virus infection.


Assuntos
Herpesvirus Humano 1 , Fatores de Transcrição , Humanos , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Herpesvirus Humano 1/metabolismo , Proteína Vmw65 do Vírus do Herpes Simples/química , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Fator C1 de Célula Hospedeira , Etoposídeo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
8.
Int J Mol Sci ; 23(4)2022 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-35216447

RESUMO

Oncolytic bovine herpesvirus type 1 (BoHV-1) infection induces DNA damage in human lung adenocarcinoma cell line A549. However, the underlying mechanisms are not fully understood. We found that BoHV-1 infection decreased the steady-state protein levels of p53-binding protein 1 (53BP1), which plays a central role in dictating DNA damage repair and maintaining genomic stability. Furthermore, BoHV-1 impaired the formation of 53BP1 foci, suggesting that BoHV-1 inhibits 53BP1-mediated DNA damage repair. Interestingly, BoHV-1 infection redistributed intracellular ß-catenin, and iCRT14 (5-[[2,5-Dimethyl-1-(3-pyridinyl)-1H-pyrrol-3-yl]methylene]-3-phenyl-2,4-thiazolidinedione), a ß-catenin-specific inhibitor, enhanced certain viral protein expression, such as the envelope glycoproteins gC and gD, and enhanced virus infection-induced DNA damage. Therefore, for the first time, we provide evidence showing that BoHV-1 infection disrupts 53BP1-mediated DNA damage repair and suggest ß-catenin as a potential host factor restricting both virus replication and DNA damage in A549 cells.


Assuntos
Adenocarcinoma de Pulmão/genética , Dano ao DNA/efeitos dos fármacos , Infecções por Herpesviridae/genética , Neoplasias Pulmonares/genética , Piridinas/farmacologia , Pirróis/farmacologia , Tiazolidinedionas/farmacologia , Proteínas Virais/genética , beta Catenina/antagonistas & inibidores , Células A549 , Linhagem Celular Tumoral , Dano ao DNA/genética , Herpesvirus Bovino 1/patogenicidade , Humanos , Replicação Viral/efeitos dos fármacos
9.
Polymers (Basel) ; 13(23)2021 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-34883660

RESUMO

Traditional crosslinked diene rubber has excellent thermal-mechanical properties and solvent resistance, yet it is incapable of being recycled via universal molding or injecting. Vitrimers, a new class of covalently crosslinked polymer networks, can be topologically rearranged with the associative exchange mechanism, endowing them with thermoplasticity. Introducing the concept of vitrimers into crosslinked networks for the recycling of rubbers is currently an attractive research topic. However, designing tailored rubber vitrimers still remains a challenge. Herein, polybutadiene (PB) vitrimers with different structures were prepared via partial epoxidation of double bonds and ring-opening esterification reactions. Their mechanical and relaxation properties were investigated. It was found that the increasing crosslinking density can increase tensile strength and activation energy for altering the network topology. The influence of side-group effects on their relaxation properties shows that an increase in the number of epoxy groups on the polybutadiene chain can increase the chance of an effective exchange of disulfide units. This work provides a simple network design which can tune vitrimer properties via altering the crosslinking density and side-group effects.

10.
Cytokine Growth Factor Rev ; 62: 54-61, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34503914

RESUMO

Inflammatory responses, characterized by the overproduction of numerous proinflammatory mediators by immune cells, is essential to protect the host against invading pathogens. Excessive production of proinflammatory cytokines is a key pathogenic factor accounting for severe tissue injury and disease progression during the infection of multiple viruses, which are therefore termed as "cytokine storm". High mobility group box 1 (HMGB1), a ubiquitous DNA-binding protein released either over virus-infected cells or activated immune cells, may act as a proinflammatory cytokine with a robust capacity to potentiate inflammatory response and disease severity. Moreover, HMGB1 is a host factor that potentially participates in the regulation of viral replication cycles with complicated mechanisms. Currently, HMGB1 is regarded as a promising therapeutic target against virus infection. Here, we provide an overview of the updated studies on how HMGB1 is differentially manipulated by distinct viruses to regulate viral diseases.


Assuntos
Proteína HMGB1 , Viroses , Vírus , Citocinas , Humanos , Viroses/tratamento farmacológico , Replicação Viral
11.
Int J Mol Sci ; 22(16)2021 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-34445287

RESUMO

Bovine herpesvirus 1 (BoHV-1) is a promising oncolytic virus with broad antitumor spectrum; however, its oncolytic effects on human lung adenocarcinoma in vivo have not been reported. In this study, we report that BoHV-1 can be used as an oncolytic virus for human lung adenocarcinoma, and elucidate the underlying mechanism of how BoHV-1 suppresses tumor cell proliferation and growth. First, we examined the oncolytic activities of BoHV-1 in human lung adenocarcinoma A549 cells. BoHV-1 infection reduced the protein levels of histone deacetylases (HDACs), including HDAC1-4 that are promising anti-tumor drug targets. Furthermore, the HDAC inhibitor Trichostatin A (TSA) promoted BoHV-1 infection and exacerbated DNA damage and cytopathology, suggesting a synergy between BoHV-1 and TSA. In the A549 tumor xenograft mouse model, we, for the first time, showed that BoHV-1 can infect tumor and suppressed tumor growth with a similar high efficacy as the treatment of TSA, and HDACs have potential effects on the virus replication. Taken together, our study demonstrates that BoHV-1 has oncolytic effects against human lung adenocarcinoma in vivo.


Assuntos
Adenocarcinoma de Pulmão/patologia , Herpesvirus Bovino 1/fisiologia , Neoplasias Pulmonares/patologia , Células A549 , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/terapia , Adenocarcinoma de Pulmão/virologia , Animais , Proliferação de Células/genética , Células Cultivadas , Cricetinae , Dano ao DNA , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Int J Biol Macromol ; 183: 2376-2386, 2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-34111485

RESUMO

Bovine pestivirus A and B, previously known as bovine viral diarrhea virus (BVDV)-1 and 2, respectively, are important pathogens of cattle worldwide, which causes significant economic losses. B-cell epitopes in BVDV glycoprotein E2 and nonstructural protein NS2/3 have been extensively identified. In this study, we screened a 12-mer phage display peptide library using commercial goat anti-BVDV serum, and identified a mimotope "LTPHKHHKHLHA" referred to as P3. With sequence alignment, a putative B-cell epitope "77ESRKKLEKALLA88" termed as P3-BVDV1/2 residing in BVDV core protein was identified. The synthesized peptides of both P3 and P3-BVDV1/2 show strong reactivity with BVDV serum in immune blot assay. Immunization of mice with these individual peptides leads to the production of antibody that cannot neutralize virus infectivity. Thus for the first time we identified a B-cell epitope, "77ESRKKLEKALLA88", in BVDV core protein. Interestingly, the epitope was highly conserved in Pestivirus A, B, C, D, as well as emerging Pestivirus E and I, but highly variable in Pestiviruses H, G, F, and J, as well as unclassified Pestivirus originated from non-ruminant animals. Whether this putative B-cell epitope is implicated in pestivirus pathogenesis or evolution needs further investigations once large numbers of isolates are available in the future.


Assuntos
Técnicas de Visualização da Superfície Celular , Vírus da Diarreia Viral Bovina Tipo 1/imunologia , Vírus da Diarreia Viral Bovina Tipo 2/imunologia , Mapeamento de Epitopos , Epitopos de Linfócito B/imunologia , Biblioteca de Peptídeos , Proteínas do Core Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Bovinos , Vírus da Diarreia Viral Bovina Tipo 1/genética , Vírus da Diarreia Viral Bovina Tipo 1/patogenicidade , Vírus da Diarreia Viral Bovina Tipo 2/genética , Vírus da Diarreia Viral Bovina Tipo 2/patogenicidade , Cães , Epitopos de Linfócito B/administração & dosagem , Epitopos de Linfócito B/genética , Feminino , Imunização , Imunogenicidade da Vacina , Células Madin Darby de Rim Canino , Camundongos Endogâmicos BALB C , Mutação , Proteínas do Core Viral/administração & dosagem , Proteínas do Core Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
13.
Virulence ; 12(1): 1345-1361, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34008469

RESUMO

High mobility group box 1 (HMGB1), a ubiquitous DNA-binding protein, can be released into extracellular space and function as a strong proinflammatory cytokine, which plays critical roles in the pathogenesis of various inflammatory diseases. Here, we showed that BoHV-1 productive infection in MDBK cells at later stage significantly increases HMGB1 mRNA expression and the protein release, but decreases the steady-state protein levels. Virus infection increases accumulation of HMGB1 protein in both nucleus and mitochondria, and relocalizes nuclear HMGB1 to assemble in highlighted foci via a confocal microscope assay. Interestingly, ß-catenin-specific inhibitor iCRT14 is able to increase HMGB1 transcription and the protein release, and subcellular translocation in virus-infected cells. HMGB1-specific inhibitor, glycyrrhizin, could differentially affect virus gene transcription such as, the viral regulatory protein bICP0, bICP4 and bICP22, as well as glycoprotein gD. In summary, our data provides a novel mechanism that ß-catenin signaling may regulate inflammatory response via affecting HMGB1 signaling.


Assuntos
Proteína HMGB1 , Infecções por Herpesviridae , Herpesvirus Bovino 1 , beta Catenina , Animais , Bovinos , Técnicas de Cultura de Células , Linhagem Celular , Proteína HMGB1/genética , Transdução de Sinais , Proteínas Virais , beta Catenina/genética
14.
J Vet Sci ; 21(5): e72, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33016019

RESUMO

BACKGROUND: Fenbendazole, a dewormer drug, is used widely in the clinical treatment of parasite infections in animals. Recent studies have shown that fenbendazole has substantial effects on tumor growth, immune responses, and inflammatory responses, suggesting that fenbendazole is a pluripotent drug. Nevertheless, the antiviral effects have not been reported. Fenbendazole can disrupt microtubules, which are essential for multiple viruses infections, suggesting that fenbendazole might have antiviral effects. OBJECTIVES: This study examined whether fenbendazole could inhibit bovine herpesvirus 1 (BoHV-1) productive infection in cell cultures. METHODS: The effects of fenbendazole on viral production, transcription of the immediate early (IE) genes, viron-associated protein expression, and the cellular signaling PLC-γ1/Akt pathway were assessed using distinct methods. RESULTS: Fenbendazole could inhibit BoHV-1 productive infections significantly in MDBK cells in a dose-dependent manner. A time-of-addition assay indicated that fenbendazole affected both the early and late stages in the virus replication cycles. The transcription of IE genes, including BoHV-1 infected cell protein 0 (bICP0), bCP4, and bICP22, as well as the synthesis of viron-associated proteins, were disrupted differentially by the fenbendazole treatment. The treatment did not affect the cellular signaling pathway of PLC-γ1/Akt, a known cascade playing important roles in virus infection. CONCLUSIONS: Overall, fenbendazole has antiviral effects on BoHV-1 replication.


Assuntos
Antivirais/farmacologia , Fenbendazol/farmacologia , Herpesvirus Bovino 1/efeitos dos fármacos , Animais , Antinematódeos/farmacologia , Cães , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Células Madin Darby de Rim Canino
15.
Vet Microbiol ; 248: 108804, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32827927

RESUMO

C-Jun, activated by various extracellular signals, is important for cell differentiation, proliferation, apoptosis, and inflammatory responses. We have previously reported that bovine herpesvirus 1 (BoHV-1) infection in MDBK cells stimulates the c-Jun NH2-terminal kinase (JNK)/c-Jun cascade for efficient replication. However, the mechanisms regarding the regulation of c-Jun following BoHV-1 infection remain unknown. In this study, we show that virus infection increases accumulation of p-c-Jun(S73) (phosphorylated c-Jun at Ser73) and p-ß-catenin(S552) in the nucleus, resulting in relocalized nuclear p-c-Jun(S73) to assemble in highlighted punctum via a confocal microscope assay. An association between ß-catenin and c-Jun in the nucleus was readily detected in virus-infected, but not mock-infected cells. Interestingly, ß-catenin was found to be involved in the regulation of c-Jun signaling in virus-infected cells as iCRT14, a ß-catenin-specific inhibitor that can inhibit ß-catenin-dependent transcriptional activity, was able to decrease protein expression and phosphorylation of c-Jun. Furthermore, we suggest that BoHV-1 infection stimulates c-Jun phosphorylation regulated by ß-catenin via both c-Jun NH2-terminal kinase (JNK)-dependent and JNK-independent mechanisms. These data add to our knowledge regarding the regulation of c-Jun following virus infection and further support the important roles of ß-catenin signaling playing in BoHV-1 infection.


Assuntos
Genes jun , Infecções por Herpesviridae/veterinária , Interações Hospedeiro-Patógeno/genética , Transdução de Sinais , beta Catenina/genética , Animais , Bovinos , Linhagem Celular , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Herpesvirus Bovino 1 , Rim/citologia , Fosforilação
16.
Viruses ; 12(9)2020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32846937

RESUMO

Accumulating studies have shown that the epidermal growth factor receptor (EGFR) signaling pathway plays an essential role in mediating cellular entry of numerous viruses. In this study, we report that bovine herpesvirus 1 (BoHV-1) productive infection in both the human lung carcinoma cell line A549 and bovine kidney (MDBK) cells leads to activation of EGFR, as demonstrated by the increased phosphorylation of EGFR at Tyr1068 (Y1068), which in turn plays important roles in virus infection. A time-of-addition assay supported that virus replication at post-entry stages was affected by the EGFR specific inhibitor Gefitinib. Interestingly, both phospholipase C-γ1 (PLC-γ1) and Akt, canonical downstream effectors of EGFR, were activated following virus infection in A549 cells, while Gefitinib could inhibit the activation of PLC-γ1 but not Akt. In addition, virus titers in A549 cells was inhibited by chemical inhibition of PLC-γ1, but not by the inhibition of Akt. However, the Akt specific inhibitor Ly294002 could significantly reduce the virus titer in MDBK cells. Taken together, our data suggest that PLC-γ1 is stimulated in part through EGFR for efficient replication in A549 cells, whereas Akt can be stimulated by virus infection independent of EGFR, and is not essential for virus productive infection, indicating that Akt modulates BoHV-1 replication in a cell type-dependent manner. This study provides novel insights on how BoHV-1 infection activates EGFR signaling transduction to facilitate virus replication.


Assuntos
Receptores ErbB/metabolismo , Herpesvirus Bovino 1/fisiologia , Transdução de Sinais , Replicação Viral , Células A549 , Animais , Bovinos , Linhagem Celular , Receptores ErbB/antagonistas & inibidores , Interações Hospedeiro-Patógeno , Humanos , Fosfolipase C gama/antagonistas & inibidores , Fosfolipase C gama/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
17.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796067

RESUMO

Neurotropic Alphaherpesvirinae subfamily members such as bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) establish and maintain lifelong latent infections in neurons. Following infection of ocular, oral, or nasal cavities, sensory neurons within trigeminal ganglia (TG) are an important site for latency. Certain external stressors can trigger reactivation from latency, in part because activation of the glucocorticoid receptor (GR) stimulates productive infection and promoters that drive expression of key viral transcriptional regulators. The Akt serine/threonine protein kinase family is linked to maintaining latency. For example, Akt3 is detected in more TG neurons during BoHV-1 latency than in reactivation and uninfected calves. Furthermore, Akt signaling correlates with maintaining HSV-1 latency in certain neuronal models of latency. Finally, an active Akt protein kinase is crucial for the ability of the HSV-1 latency-associated transcript (LAT) to inhibit apoptosis in neuronal cell lines. Consequently, we hypothesized that viral and/or cellular factors impair stress-induced transcription and reduce the incidence of reactivation triggered by low levels of stress. New studies demonstrate that Akt1 and Akt2, but not Akt3, significantly reduced GR-mediated transactivation of the BoHV-1 immediate early transcription unit 1 (IEtu1) promoter, the HSV-1 infected cell protein 0 (ICP0) promoter, and the mouse mammary tumor virus long terminal repeat (MMTV-LTR). Akt3, but not Akt1 or Akt2, significantly enhanced neurite formation in mouse neuroblastoma cells, which correlates with repairing damaged neurons. These studies suggest that unique biological properties of the three Akt family members promote the maintenance of latency in differentiated neurons.IMPORTANCE External stressful stimuli are known to increase the incidence of reactivation of Alphaherpesvirinae subfamily members. Activation of the glucocorticoid receptor (GR) by the synthetic corticosteroid dexamethasone (DEX) stimulates bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) reactivation. Furthermore, GR and dexamethasone stimulate productive infection and promoters that drive expression of viral transcriptional regulators. These observations lead us to predict that stress-induced transcription is impaired by factors abundantly expressed during latency. Interestingly, activation of the Akt family of serine/threonine protein kinases is linked to maintenance of latency. New studies reveal that Akt1 and Ak2, but not Akt3, impaired GR- and dexamethasone-mediated transactivation of the BoHV-1 immediate early transcription unit 1 and HSV-1 ICP0 promoters. Strikingly, Akt3, but not Akt1 or Akt2, stimulated neurite formation in mouse neuroblastoma cells, a requirement for neurogenesis. These studies provide insight into how Akt family members may promote the maintenance of lifelong latency.


Assuntos
Herpes Simples/imunologia , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Células Receptoras Sensoriais/virologia , Animais , Bovinos , Diferenciação Celular , Linhagem Celular Tumoral , Herpes Simples/genética , Herpes Simples/patologia , Herpes Simples/virologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/imunologia , Camundongos , Neuritos/imunologia , Neuritos/ultraestrutura , Neuritos/virologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/imunologia , Células Receptoras Sensoriais/imunologia , Células Receptoras Sensoriais/patologia , Transdução de Sinais , Ativação Transcricional/imunologia , Gânglio Trigeminal/imunologia , Gânglio Trigeminal/patologia , Gânglio Trigeminal/virologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
18.
PLoS One ; 15(3): e0230870, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32226020

RESUMO

When herpes simplex virus 1 (HSV-1) infection is initiated in the ocular, nasal, or oral cavity, sensory neurons within trigeminal ganglia (TG) become infected. Following a burst of viral transcription in TG neurons, lytic cycle viral genes are suppressed and latency is established. The latency-associated transcript (LAT) is the only viral gene abundantly expressed during latency, and LAT expression is important for the latency-reactivation cycle. Reactivation from latency is required for virus transmission and recurrent disease, including encephalitis. The Wnt/ß-catenin signaling pathway is differentially expressed in TG during the bovine herpesvirus 1 latency-reactivation cycle. Hence, we hypothesized HSV-1 regulates the Wnt/ß-catenin pathway and promotes maintenance of latency because this pathway enhances neuronal survival and axonal repair. New studies revealed ß-catenin was expressed in significantly more TG neurons during latency compared to TG from uninfected mice or mice latently infected with a LAT-/- mutant virus. When TG explants were incubated with media containing dexamethasone to stimulate reactivation, significantly fewer ß-catenin+ TG neurons were detected. Conversely, TG explants from uninfected mice or mice latently infected with a LAT-/- mutant increased the number of ß-catenin+ TG neurons in the presence of DEX relative to samples not treated with DEX. Impairing Wnt signaling with small molecule antagonists reduced virus shedding during explant-induced reactivation. These studies suggested ß-catenin was differentially expressed during the latency-reactivation cycle, in part due to LAT expression.


Assuntos
Regulação da Expressão Gênica , Herpesvirus Humano 1/fisiologia , Neurônios/metabolismo , Neurônios/virologia , Gânglio Trigeminal/citologia , Ativação Viral , beta Catenina/metabolismo , Animais , Feminino , Camundongos , Via de Sinalização Wnt
19.
Oxid Med Cell Longev ; 2019: 4957878, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31687081

RESUMO

Bovine herpesvirus type 1 (BoHV-1) is a significant cofactor for bovine respiratory disease complex (BRDC), the most important inflammatory disease in cattle. BoHV-1 infection in cell cultures induces overproduction of pathogenic reactive oxygen species (ROS) and the depletion of nuclear factor erythroid 2 p45-related factor 2 (Nrf2), a master transcriptional factor regulating a panel of antioxidant and cellular defense genes in response to oxidative stress. In this study, we reported that the virus productive infection in MDBK cells at the later stage significantly decreased the expression levels of heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase-1 (NQO1) proteins, the canonical downstream targets regulated by Nrf2, inhibited Nrf2 acetylation, reduced the accumulation of Nrf2 proteins in the nucleus, and relocalized nuclear Nrf2 proteins to form dot-like staining patterns in confocal microscope assay. The differential expression of Kelch-like ECH associated protein 1 (KEAP1) and DJ-1 proteins as well as the decreased association between KEAP1 and DJ-1 promoted Nrf2 degradation through the ubiquitin proteasome pathway. These data indicated that the BoHV-1 infection may significantly suppress the Nrf2 signaling pathway. Moreover, we found that there was an association between Nrf2 and LaminA/C, H3K9ac, and H3K18ac, and the binding ratios were altered following the virus infection. Taken together, for the first time, we provided evidence showing that BoHV-1 infection inhibited the Nrf2 signaling pathway by complicated mechanisms including promoting Nrf2 degradation, relocalization of nuclear Nrf2, and inhibition of Nrf2 acetylation.


Assuntos
Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/patologia , Herpesvirus Bovino 1/fisiologia , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais , Acetilação , Animais , Bovinos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cromanos/farmacologia , Herpesvirus Bovino 1/efeitos dos fármacos , Laminas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Proteólise/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ubiquitina/metabolismo
20.
Mediators Inflamm ; 2019: 7072917, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31011285

RESUMO

Reactive oxidative species (ROS) are important inflammatory mediators. Electrons escaping from the mitochondrial electron transport chain (ETC) during oxidative phosphorylation (OXPHOS) in the mitochondrial respiratory chain (RC) complexes contribute to ROS production. The cellular antioxidant enzymes are important for maintaining ROS release at the physiological levels. It has been reported that BoHV-1 infection induces overproduction of ROS and oxidative mitochondrial dysfunction in cell cultures. In this study, we found that chemical interruption of RC complexes by TTFA (an inhibitor of RC complex II), NaN3 (an inhibitor of RC complex IV), and oligomycin A (an inhibitor of ATP synthase) consistently decreased virus productive infection, suggesting that the integral processes of RC complexes are important for the virus replication. The virus infection significantly increased the expression of subunit SDHB (succinate dehydrogenase) and MTCO1 (cytochrome c oxidase subunit I), critical components of RC complexes II and IV, respectively. The expression of antioxidant enzymes including superoxide dismutase 1 (SOD1), SOD2, catalase (CAT), and glutathione peroxidase 4 (GPX4) was differentially affected following the virus infection. The protein TFAM (transcription factor A, mitochondrial) stimulated by either nuclear respiratory factor 1 (NRF1) or NRF2 is a key regulator of mitochondrial biogenesis. Interestingly, the virus infection at the late stage (at 16 h after infection) stimulated TFAM expression but decreased the levels of both NRF1 and NRF2, indicating that virus infection activated TFAM signaling independent of either NRF1 or NRF2. Overall, this study provided evidence that BoHV-1 infection altered the expression of molecules associated with RC complexes, antioxidant enzymes, and mitochondrial biogenesis-related signaling NRF1/NRF2/TFAM, which correlated with the previous report that virus infection induces ROS overproduction and mitochondrial dysfunction.


Assuntos
Antioxidantes/metabolismo , Herpesvirus Bovino 1/patogenicidade , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Western Blotting , Linhagem Celular , Cães , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Oligomicinas/farmacologia , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Azida Sódica/farmacologia , Succinato Desidrogenase/metabolismo , Tenoiltrifluoracetona/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...