Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Vet Res ; 49(1): 15, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29439710

ABSTRACT

Porcine circovirus type 2 (PCV2) is recognized as the causative agent of porcine circovirus-associated diseases. PCV2 replication could be promoted by low doses of ochratoxin A (OTA) as in our previous study and selenium has been shown to attenuate PCV2 replication. However, the underlying mechanism remains unclear. The aim of the study was to investigate the effects of selenomethionine (SeMet), the major component of organic selenium, on OTA-induced PCV2 replication promotion and its potential mechanism. The present study demonstrates that OTA could promote PCV2 replication as measured by cap protein expression, viral titer, viral DNA copies and the number of infected cells. In addition, OTA could activate autophagy as indicated by up-regulated light chain 3 (LC3)-II and autophagy-related protein 5 expressions and autophagosome formation. Further, OTA could down-regulate p-AKT and p-mTOR expressions and OTA-induced autophagy was inhibited when insulin was applied. SeMet at 2, 4 and 6 µM had significant inhibiting effects against OTA-induced PCV2 replication promotion. Furthermore, SeMet could attenuate OTA-induced autophagy and up-regulate OTA-induced p-AKT and p-mTOR expression inhibition. Rapamycin, an inhibitor of AKT/mTOR, could reverse the effects of SeMet on OTA-induced autophagy and the PCV2 replication promotion. In conclusion, SeMet could block OTA-induced PCV2 replication promotion by inhibiting autophagy by activating the AKT/mTOR pathway. Therefore, SeMet supplementation could be an effective prophylactic strategy against PCV2 infections and autophagy may be a potential marker to develop novel anti-PCV2 drugs.


Subject(s)
Autophagy/drug effects , Circovirus/physiology , Selenomethionine/metabolism , Signal Transduction/drug effects , Virus Replication/drug effects , Animals , Cell Line , Ochratoxins/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Swine , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
2.
Cell Death Dis ; 8(6): e2909, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28661479

ABSTRACT

Ochratoxin A (OTA) is a mycotoxin produced by Aspergillus and Penicillium. Porcine circovirus type 2 (PCV2) is recognized as the causative agent of porcine circovirus-associated diseases. Recently, we reported that low doses of OTA promoted PCV2 replication in vitro and in vivo, but the underlying mechanism needed further investigation. The present studies further confirmed OTA-induced PCV2 replication promotion as measured by cap protein expression, viral titer, viral DNA copies and the number of infected cells. Our studies also showed that OTA induced autophagy in PK-15 cells, as assessed by the markedly increased expression of microtubule-associated protein 1 light chain 3 (LC3)-II, autophagy-related protein 5 (ATG5), and Beclin-1 and the accumulation of green fluorescent protein (GFP)-LC3 dots. OTA induced complete autophagic flux, which was detected by monitoring p62 degradation and LC3-II turnover using immunoblotting. Inhibition of autophagy by 3-methylademine (3-MA) and chloroquine (CQ) significantly attenuated OTA-induced PCV2 replication promotion. The observed phenomenon was further confirmed by the knock-down of ATG5 or Beclin-1 by specific siRNA. Further studies showed that N-acetyl-L-cysteine (NAC), an ROS scavenger could block autophagy induced by OTA, indicating that ROS may be involved in the regulation of OTA-induced autophagy. Furthermore, we observed significant increases in OTA concentrations in lung, spleen, kidney, liver and inguinal lymph nodes (ILN) and bronchial lymph nodes (BLN) of pigs fed 75 and 150 µg/kg OTA compared with controls in vivo. Administration of 75 µg/kg OTA significantly increased PCV2 replication and autophagy in the lung, spleen, kidney and BLN of pigs. Taken together, it could be concluded that OTA-induced autophagy in vitro and in vivo promotes PCV2 replication.


Subject(s)
Circoviridae Infections/genetics , Circovirus/genetics , Ochratoxins/administration & dosage , Virus Replication/drug effects , Animals , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Beclin-1/genetics , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circovirus/drug effects , Circovirus/pathogenicity , DNA, Viral/drug effects , Gene Expression Regulation, Viral/genetics , Microtubule-Associated Proteins , Swine , Tissue Distribution
3.
Sci Rep ; 7: 40440, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28071725

ABSTRACT

This study explored the effects of Astragalus polysaccharide (APS) on porcine circovirus type 2 (PCV2) infections and its mechanism in vivo and vitro. First, fifty 2-week-old mice were randomly divided into five groups: a group without PCV2 infection and groups with PCV2 infections at 0, 100, 200 or 400 mg/kg APS treatments. The trial lasted for 28 days. The results showed that APS treatments at 200 and 400 mg/kg reduced the pathological injury of tissues, inhibited PCV2 infection and decreased glucose-regulated protein 78 (GRP78) and GADD153/CHOP gene mRNA and protein expression significantly (P < 0.05). Second, a study on endoplasmic reticulum stress mechanism was carried out in PK15 cells. APS treatments at 15 and 45 µg/mL significantly reduced PCV2 infection and GRP78 mRNA and protein expression (P < 0.05). Tunicamycin supplementation increased GRP78 mRNA and protein expression and significantly attenuated the APS-induced inhibition of PCV2 infection (P < 0.05). Tauroursodeoxycholic acid supplementation decreased GRP78 mRNA and protein expression and significantly inhibited PCV2 infection (P < 0.05). In addition, fifty 2-week-old mice were randomly divided into five groups: Con, PCV2, APS + PCV2, TM + PCV2 and TM + APS + PCV2. The results were similar to those in PK15 cells. Taken together, it could be concluded that APS suppresses PCV2 infection by inhibiting endoplasmic reticulum stress.


Subject(s)
Astragalus Plant/chemistry , Circoviridae Infections/drug therapy , Circoviridae Infections/virology , Circovirus/physiology , Endoplasmic Reticulum Stress/drug effects , Polysaccharides/therapeutic use , Animals , Cell Line , Circoviridae Infections/pathology , Circovirus/drug effects , Endoplasmic Reticulum Chaperone BiP , Mice , Oxidative Stress/drug effects , Phytotherapy , Polysaccharides/pharmacology , Swine , Taurochenodeoxycholic Acid/pharmacology , Tunicamycin/pharmacology , Virus Replication/drug effects
4.
Oncotarget ; 7(15): 20469-85, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26943035

ABSTRACT

Porcine circovirus type 2 (PCV2) is the primary cause of porcine circovirus disease, and ochratoxin A (OTA)-induced oxidative stress promotes PCV2 replication. In humans, selenoprotein S (SelS) has antioxidant ability, but it is unclear whether SelS affects viral infection. Here, we stably transfected PK15 cells with pig pCDNA3.1-SelS to overexpress SelS. Selenium (Se) at 2 or 4 µM and SelS overexpression blocked the OTA-induced increases of PCV2 DNA copy number and infected cell numbers. SelS overexpression also increased glutathione (GSH), NF-E2-related factor 2 (Nrf2) mRNA, and γ-glutamyl-cysteine synthetase mRNA levels; decreased reactive oxygen species (ROS) levels; and inhibited p38 phosphorylation in PCV2-infected PK15 cells, regardless of OTA treatment. Buthionine sulfoximine reversed all of the above SelS-induced changes. siRNA-mediated SelS knockdown decreased Nrf2 mRNA and GSH levels, increased ROS levels, and promoted PCV2 replication in OTA-treated PK15 cells. These data indicate that pig SelS blocks OTA-induced promotion of PCV2 replication by inhibiting the oxidative stress and p38 phosphorylation in PK15 cells.


Subject(s)
Circoviridae Infections/metabolism , Circovirus/pathogenicity , Ochratoxins/toxicity , Oxidative Stress/drug effects , Selenoproteins/metabolism , Swine Diseases/metabolism , Virus Replication/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antioxidants , Apoptosis , Carcinogens/toxicity , Cell Proliferation , Cells, Cultured , Circoviridae Infections/chemically induced , Circoviridae Infections/virology , Circovirus/drug effects , Glutathione/metabolism , NF-E2-Related Factor 2/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Swine , Swine Diseases/chemically induced , Swine Diseases/virology
5.
J Agric Food Chem ; 64(6): 1385-93, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26806088

ABSTRACT

Selenium (Se) is generally known as an essential micronutrient and antioxidant for humans and animals. Aflatoxin B1 (AFB1) is a frequent contaminant of food and feed, causing immune toxicity and hepatotoxicity. Little has been done about the mechanisms of how Se protects against AFB1-induced immune toxicity. The aim of this present study is to investigate the protective effects of Se against AFB1 and the underlying mechanisms. The primary splenocytes isolated from healthy pigs were stimulated by anti-pig-CD3 monoclonal antibodies and treated by various concentrations of different Se forms and AFB1. The results showed that Se supplementation alleviated the immune toxicity of AFB1 in a dose-dependent manner, as demonstrated by increasing T-cell proliferation and interleukin-2 production. Addition of buthionine sulfoximine abrogated the protective effects of SeMet against AFB1. SeMet enhanced mRNA and protein expression of glutathione peroxidase 1 (GPx1), selenoprotein S (SelS), and thioredoxin reductase 1 without and with AFB1 treatments. Furthermore, knockdown of GPx1 and SelS by GPx1-specific siRNA and SelS-specific siRNA diminished the protective effects of SeMet against AFB1-induced immune toxicity. It is concluded that SeMet diminishes AFB1-induced immune toxicity through increasing antioxidant ability and improving GPx1 and SelS expression in splenocytes. This study suggests that organic selenium may become a promising supplementation to protect humans and animals against the decline in immunity caused by AFB1.


Subject(s)
Aflatoxin B1/toxicity , Glutathione Peroxidase/genetics , Selenium/immunology , Selenoproteins/genetics , Spleen/cytology , Spleen/immunology , Animal Feed/analysis , Animals , Cells, Cultured , Dietary Supplements/analysis , Glutathione Peroxidase/immunology , Selenoproteins/immunology , Spleen/drug effects , Spleen/enzymology , Swine , Glutathione Peroxidase GPX1
6.
Int J Biol Macromol ; 81: 22-30, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26226456

ABSTRACT

Porcine circovirus type 2 (PCV2) is the primary causative agent of porcine circovirus-associated disease (PCVAD). Astragalus polysaccharide (APS), as one kind of biological macromolecule extracted from Astragalus, has antiviral activities. This study was undertaken to explore the effect of APS on PCV2 replication in vitro and the underlying mechanisms. Our results showed that adding APS before PCV2 infection decreased significantly PCV2 DNA copies, the number of infected cells, MDA level, ROS level and NF-κB activation in PK15 cells and increased significantly GSH contents and SOD activity compared to control without APS. Oxidative stress induced by BSO could eliminate the effect of PCV2 replication inhibition by APS. LPS, as a NF-κB activator, could attenuate the effect of PCV2 replication inhibition by APS. BAY 11-7082, as a NF-κB inhibitor, could increase the effect of PCV2 replication inhibition by APS. In conclusion, APS inhibits PCV2 replication by decreasing oxidative stress and the activation of NF-κB signaling pathway, which suggests that APS might be employed for the prevention of PCV2 infection.


Subject(s)
Antiviral Agents/pharmacology , Astragalus Plant/chemistry , Circovirus/drug effects , NF-kappa B/metabolism , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Signal Transduction/drug effects , Virus Replication/drug effects , Animals , Cell Line , Cell Survival/drug effects , Circovirus/genetics , Swine
7.
J Agric Food Chem ; 63(26): 6094-101, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26073049

ABSTRACT

The aim of the present study is to investigate whether aflatoxin B1 (AFB1)-induced immunotoxicity is associated with oxidative stress and the expression of extracellular regulated protein kinases (ERK) 1/2. The primary splenocytes isolated from healthy pigs were activated and proliferated by anti-pig-CD3 monoclonal antibodies (mAb) in the present experiment, which is an antigen-specific stimulant. Results indicated that cell proliferation and interleukin-2 (IL-2) production were significantly suppressed by AFB1 from 4 to 8 µg/mL in a dose-dependent manner compared to the control group. Furthermore, AFB1 significantly increased malondialdehyde (MDA) levels, decreased reduced glutathione (GSH) and total superoxide dismutase levels, and up-regulated p-ERK1/2 expression in the activated splenocytes. N-Acetyl-l-cysteine blocked anti-CD3-induced T-cell suppression by AFB1 through increasing intracellular concentrations of GSH levels, decreasing MDA levels, and down-regulated p-ERK1/2 expression, respectively. Inhibition of the ERK1/2 expression by ERK-specific iRNA attenuated the decrease of T-cell proliferation and IL-2 production induced by AFB1. It was concluded that AFB1 inhibits anti-CD3-induced lymphocyte proliferation and IL-2 production by the oxidative stress mediated ERK1/2 MAPK signaling pathway.


Subject(s)
Aflatoxin B1/toxicity , Antibodies, Monoclonal/immunology , CD3 Complex/immunology , MAP Kinase Signaling System/drug effects , Spleen/cytology , T-Lymphocytes/immunology , Animals , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Spleen/drug effects , Spleen/immunology , Swine , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...