Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Virol ; 93(9)2019 05 01.
Article in English | MEDLINE | ID: mdl-30787154

ABSTRACT

Oncogenic virus replication often leads to genomic instability, causing DNA damage and inducing the DNA damage response (DDR) pathway. The DDR pathway is a cellular pathway that senses DNA damage and regulates the cell cycle to maintain genomic stability. Therefore, the DDR pathway is critical for the viral lifecycle and tumorigenesis. Marek's disease virus (MDV), an alphaherpesvirus that causes lymphoma in chickens, has been shown to induce DNA damage in infected cells. However, the interaction between MDV and the host DDR is unclear. In this study, we observed that MDV infection causes DNA strand breakage in chicken fibroblast (CEF) cells along with an increase in the DNA damage markers p53 and p21. Interestingly, we showed that phosphorylation of STAT3 was increased during MDV infection, concomitantly with a decrease of Chk1 phosphorylation. In addition, we found that MDV infection was enhanced by VE-821, an ATR-specific inhibitor, but attenuated by hydroxyurea, an ATR activator. Moreover, inhibition of STAT3 phosphorylation by Stattic eliminates the ability of MDV to inhibit Chk1 phosphorylation. Finally, we showed that MDV replication was decreased by Stattic treatment. Taken together, these results suggest that MDV disables the ATR-Chk1 pathway through STAT3 activation to benefit its replication.IMPORTANCE MDV is used as a biomedical model to study virus-induced lymphoma due to the similar genomic structures and physiological characteristics of MDV and human herpesviruses. Upon infection, MDV induces DNA damage, which may activate the DDR pathway. The DDR pathway has a dual impact on viruses because it manipulates repair and recombination factors to facilitate viral replication and also initiates antiviral action by regulating other signaling pathways. Many DNA viruses evolve to manipulate the DDR pathway to promote virus replication. In this study, we identified a mechanism used by MDV to inhibit ATR-Chk1 pathways. ATR is a cellular kinase that responds to broken single-stranded DNA, which has been less studied in MDV infection. Our results suggest that MDV infection activates STAT3 to disable the ATR-Chk1 pathway, which is conducive to viral replication. This finding provides new insight into the role of STAT3 in interrupting the ATR-Chk1 pathway during MDV replication.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Avian Proteins/metabolism , Checkpoint Kinase 1/metabolism , Mardivirus/physiology , Marek Disease/metabolism , STAT3 Transcription Factor/metabolism , Virus Replication/physiology , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Avian Proteins/genetics , Cell Line , Checkpoint Kinase 1/genetics , Chickens , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Marek Disease/genetics , Marek Disease/pathology , Pyrazines/pharmacology , STAT3 Transcription Factor/genetics , Sulfones/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Virus Replication/drug effects
2.
BMC Vet Res ; 14(1): 122, 2018 Apr 03.
Article in English | MEDLINE | ID: mdl-29615025

ABSTRACT

BACKGROUND: Marek's disease virus (MDV) resides in the genus Mardivirus in the family Herpesviridae. MDV is a highly contagious virus that can cause neurological lesions, lymphocytic proliferation, immune suppression, and death in avian species, including Galliformes (chickens, quails, partridges, and pheasants), Strigiformes (owls), Anseriformes (ducks, geese, and swans), and Falconiformes (kestrels). CASE PRESENTATION: In 2015, two red-crowned cranes died in Nanjing (Jiangsu, China). It was determined that the birds were infected with Marek's disease virus by histopathological examination, polymerase chain reaction (PCR), gene sequencing and sequence analysis of tissue samples from two cranes. Gross lesions included diffuse nodules in the skin, muscle, liver, spleen, kidney, gizzard and heart, along with liver enlargement and gizzard mucosa hemorrhage. Histopathological assay showed that infiltrative lymphocytes and mitotic figures existed in liver and heart. The presence of MDV was confirmed by PCR. The sequence analysis of the Meq gene showed 100% identity with Md5, while the VP22 gene showed the highest homology with CVI988. Furthermore, the phylogenetic analysis of the VP22 and Meq genes suggested that the MDV (from cranes) belongs to MDV serotype 1. CONCLUSION: We describe the first molecular detection of Marek's disease in red-crowned cranes based on the findings previously described. To our knowledge, this is also the first molecular identification of Marek's disease virus in the order Gruiformes and represents detection of a novel MDV strain.


Subject(s)
Birds/virology , Herpesvirus 2, Gallid , Marek Disease/diagnosis , Animals , Animals, Wild/virology , China , Herpesvirus 2, Gallid/genetics , Marek Disease/pathology , Marek Disease/virology , Phylogeny , Polymerase Chain Reaction/veterinary , Sequence Analysis, DNA/veterinary
3.
Membranes (Basel) ; 10(9)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854258

ABSTRACT

Three new phenylacetylene monomers having one or two carbamate groups were synthesized and polymerized by using (Rh(norbornadiene)Cl)2 as an initiator. The resulting polymers had very high average molecular weights (Mw) of 1.4-4.8 × 106, with different solubility and membrane-forming abilities. The polymer having two carbamate groups and no hydroxy groups in the monomer unit showed the best solubility and membrane-forming ability among the three polymers. In addition, the oxygen permeability coefficient of the membrane was more than 135 times higher than that of a polymer having no carbamate groups and two hydroxy groups in the monomer unit with maintaining similar oxygen permselectivity. A better performance in membrane-forming ability and oxygen permeability may be caused by a more extended and flexible cis-transoid conformation and lower polarity. On the other hand, the other two new polymers having one carbamate group and two hydroxy groups in the monomer unit showed lower performances in membrane-forming abilities and oxygen permeabilities. It may be caused by a very tight cis-cisoid conformation, which was maintained by intramolecular hydrogen bonds.

4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(9): 1255-9, 2016 Sep.
Article in Zh | MEDLINE | ID: mdl-27609583

ABSTRACT

Objective To prepare monoclonal antibodies (mAbs) against chicken cell cycle checkpoint kinase 2 (cChk2). Methods The cChk2 gene was amplified by reverse transcription PCR (RT-PCR) and subcloned into the prokaryotic expression vector pGEX-4T-3. After induced by IPTG, cChk2 was expressed in BL21 (DE3) E.coli cells and analyzed by SDS-PAGE to determine its soluability. BALB/c mice were immunized with cChk2 protein peritoneally. Indirect immunofluorescence assay (IFA) and Western blotting were used to detect anti-serum; if the detection result was positive, IFA and limited dilution was performed to screen hybridoma clones that produced antibodies against cChk2. Results cChk2 was mainly expressed in inclusion bodies. The anti-sera were able to recognize Chk2. Nine positive hybridoma clones were obtained and identified as 1F4, 2D9, 2G1, 3D9, 3E3, 4B5, 4E2, 5C9 and 5F7. Conclusion The study has prepared mAbs against cChk2 with a good specificity and a high titer.


Subject(s)
Antibodies, Monoclonal/analysis , Checkpoint Kinase 2/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Blotting, Western , Checkpoint Kinase 2/genetics , Chickens , Cloning, Molecular , Escherichia coli/genetics , Immunization , Mice , Mice, Inbred BALB C
5.
Genome Announc ; 1(4)2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23950112

ABSTRACT

In 2000, three Newcastle disease virus (NDV) strains were isolated from outbreaks of infection in layers, ducklings, and geese in the same region of China during the same time period. Here, we report their complete genome sequences, which belong to the NDV genotype VIId. This discovery might provide clues as to the evolution of the NDVs of different avian origins.

6.
J Virol ; 81(5): 2401-17, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17151125

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) infection significantly increases the risk of Kaposi's sarcoma (KS) occurrence in individuals infected with Kaposi's sarcoma-associated herpesvirus (KSHV). KSHV infection appears to be necessary but not sufficient for KS development without other cofactors. However, factors that facilitate KSHV to cause KS have not been well defined. Previously, we determined that human herpesvirus 6 was one of the cofactors that activated lytic cycle replication of KSHV. Here, we demonstrate that the Tat protein of HIV-1 is a potentially important factor in the pathogenesis of KS, as determined by production of lytic phase mRNA transcripts and viral proteins in BCBL-1 cells. Mechanistic studies showed ectopic expression of Tat induced the production of human interleukin-6 (huIL-6) and its receptor (huIL-6Ra) and activated STAT3 signaling. Neutralization of huIL-6 or huIL-6R or inhibition of STAT3 signaling enhanced the replication. In addition, IL-4/STAT6 signaling also partially contributed to Tat-induced KSHV replication. These findings suggest that Tat may participate in KS pathogenesis by inducing KSHV replication and increasing KSHV viral load. These data also suggest that JAK/STAT signaling may be of therapeutic value in AIDS-related KS patients.


Subject(s)
Gene Products, tat/physiology , HIV-1/physiology , Herpesvirus 8, Human/physiology , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Animals , Base Sequence , Callithrix , Cell Line , DNA Primers/genetics , Gene Expression , Gene Products, tat/genetics , Genes, tat , HIV Infections/complications , HIV Infections/virology , HIV-1/genetics , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Interleukin-4/genetics , Interleukin-6/genetics , Mice , NIH 3T3 Cells , Receptors, Interleukin-6/genetics , STAT6 Transcription Factor/genetics , Sarcoma, Kaposi/etiology , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Signal Transduction , Virus Replication/genetics , Virus Replication/physiology , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL