Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Zhejiang Univ Sci B ; 21(4): 341, 2020 04.
Article in English | MEDLINE | ID: mdl-32253843

ABSTRACT

Erratum to: J Zhejiang Univ-Sci B (Biomed & Biotechnol) 2019 2019 20(10):816-827. https://doi.org/10.1631/jzus.B1900071. The original version of this article unfortunately contained a mistake. In p.823, Figs. 8c and 8d were in-correct, and the obvious pathological changes were mistakenly placed in the picture. The correct versions should be as follows.

2.
Acta Trop ; 207: 105458, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32243879

ABSTRACT

Mastitis is the inflammation of mammary glands which causes huge economic loss in dairy cows. Inflammation, any tissue injury and pathogens in cow udder activate Toll-like Receptors (TLRs). Staphylococcus aureus (S. aureus) is the major cause of mastitis. In mastitis, activated TLRs initiate the NF-κB/MAPKs pathways which further trigger the gene expression associated with mastitis followed by innate immune response. In this study, pathogenic-induced gene expression profile of pro-inflammatory cytokines in mammary gland tissues, was investigated in mastitis. The Hematoxylin and Eosin (H & E) results indicated severe histopathological changes in infected tissues. Western blot results suggested the over expressions of TLR2/TLR4 with NF-κB/MAPKs pathways activation in infected tissues. qRT-PCR results revealed the gene expression associated with TLR2/TLR4-mediated NF-κB/MAPKs pathways in infected tissues in comparison with non-infected. Statistical analysis of mRNA and relative protein expression levels indicated the up-regulation of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) in infected tissues rather than non-infected tissues. These results suggested that the up-regulation of gene expression levels implicated the underlying regulatory pathways for proper immune function in mammary glands. In conclusion, our study might give new insights for investigation and better understanding of mammary gland pathophysiology and TLRs and NF-κB/MAPKs-mediated gene expression of pro-inflammatory cytokines.


Subject(s)
Cytokines/genetics , Mastitis, Bovine/immunology , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Toll-Like Receptors/physiology , Animals , Cattle , Female , Up-Regulation
3.
Microb Pathog ; 143: 104109, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32171710

ABSTRACT

Acute lung injury (ALI) is considered as an uncontrolled inflammatory response that can leads to acute respiratory distress syndrome (ARDS), which limits the therapeutic strategies. Ginsenosides Rb1 (Rb1), an active ingredient obtained from Panax ginseng, possesses a broad range of pharmacological and medicinal properties, comprising the anti-inflammatory, anti-oxidant, and anti-tumor activities. Therefore, the purpose of the present study was to investigate the protective effects of Rb1 against S. aureus-induced (ALI) through regulation of Nuclear factor erythroid 2-related factor 2 (Nrf2) and mitochondrial-mediated apoptotic pathways in mice (in-vivo), and RAW264.7 cells (in-vitro). For that purpose, forty Kunming mice were randomly assigned into four treatment groups; (1) Control group (phosphate buffer saline (PBS); (2) S. aureus group; (3) S. aureus + Rb1 (20 mg/kg) group; and (4) Rb1 (20 mg/kg) group. The 20 µg/mL dose of Rb1 was used in RAW264.7 cells. In the present study, we found that Rb1 treatment reduced ALI-induced oxidative stress via suppressing the accumulation of malondialdehyde (MDA) and myeloperoxidase (MPO) and increase the antioxidant enzyme activities of superoxidase dismutase 1 (SOD1), Catalase (CAT), and glutathione peroxidase 1 (Gpx1). Similarly, Rb1 markedly increased messenger RNA (mRNA) expression of antioxidant genes (SOD1, CAT and Gpx1) in comparison with ALI group. The histopathological results showed that Rb1 treatment ameliorated ALI-induced hemorrhages, hyperemia, perivascular edema and neutrophilic infiltration in the lungs of mice. Furthermore, Rb1 enhanced the antioxidant defense system through activating the Nrf2 signaling pathway. Our findings showed that Rb1 treated group significantly up-regulated mRNA and protein expression of Nrf2 and its downstream associated genes down-regulated by ALI in vivo and in vitro. Moreover, ALI significantly increased the both mRNA and protein expression of mitochondrial-apoptosis-related genes (Bax, caspase-3, caspase-9, cytochrome c and p53), while decreased the Bcl-2. In addition, Rb1 therapy significantly reversed the mRNA and protein expression of these mitochondrial-apoptosis-related genes, as compared to the ALI group in vivo and in vitro. Taken together, Rb1 alleviates ALI-induced oxidative injury and apoptosis by modulating the Nrf2 and mitochondrial signaling pathways in the lungs of mice.


Subject(s)
Acute Lung Injury/drug therapy , Apoptosis/drug effects , Ginsenosides/therapeutic use , Oxidative Stress/drug effects , Staphylococcal Infections/complications , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Ginsenosides/chemistry , Mice , Panax/chemistry , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction
4.
Food Funct ; 10(10): 6543-6555, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31545328

ABSTRACT

Mastitis, a major disease affecting dairy cows, is most commonly caused by Staphylococcus aureus (S. aureus). Selenium (Se) can activate pivotal proteins in immune responses and regulate the immune system, and microRNA-155 (miR-155) is a key transcriptional regulator for inflammation-related diseases. We constructed the model of mouse mastitis in vivo and primary mouse mammary epithelial cells (MMECs) in vitro, which were induced by S. aureus. Se content of the mammary was estimated using an atomic fluorescence spectrophotometer. Histopathological analysis was performed via hematoxylin and eosin (H&E) staining. The mmu-miR-155-5p mimic was transfected in MMECs, and viability was determined through the MTT assay. Transfected efficiency was evaluated by qPCR and fluorescence staining. Cytokines including TNF-α, IL-1ß, IL-10 and TLRs were detected with qPCR. In addition, western blotting was used to evaluate the expression of the NF-κB and MAPKs signaling pathways. The results demonstrated that a Se-supplemented diet improved the content of Se in mammary tissues. Histopathological studies indicated that the mammary glands were protected in the Se-supplemented group after S. aureus infection. Se-supplementation suppressed the production of MPO, mmu-miR-155, TNF-α, IL-1ß, and TLR2 and significantly inhibited the phosphorylation of NF-κB and MAPKs in vivo and in vitro. All the data indicated that mmu-miR-155 played a pro-inflammatory role in our study, and Se-supplementation could suppress the expression of mmu-miR-155 to inhibit inflammation in S. aureus-induced mastitis in mice.


Subject(s)
Cattle Diseases/drug therapy , Mastitis/drug therapy , MicroRNAs/genetics , Selenium/administration & dosage , Staphylococcal Infections/veterinary , Animals , Cattle , Cattle Diseases/genetics , Cattle Diseases/immunology , Cattle Diseases/microbiology , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression Regulation , Mastitis/genetics , Mastitis/immunology , Mastitis/microbiology , Mice , MicroRNAs/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology
5.
J Zhejiang Univ Sci B ; 20(10): 816-827, 2019.
Article in English | MEDLINE | ID: mdl-31489801

ABSTRACT

Catalpol is the main active ingredient of an extract from Radix rehmanniae, which in a previous study showed a protective effect against various types of tissue injury. However, a protective effect of catalpol on uterine inflammation has not been reported. In this study, to investigate the protective mechanism of catalpol on lipopolysaccharide (LPS)-induced bovine endometrial epithelial cells (bEECs) and mouse endometritis, in vitro and in vivo inflammation models were established. The Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling pathway and its downstream inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qRT-PCR), western blot (WB), and immunofluorescence techniques. The results from ELISA and qRT-PCR showed that catalpol dose-dependently reduced the expression of pro-inflammatory cytokines such as tumor necrosis factor α (TNF-α), interleukin (IL)-1ß, and IL-6, and chemokines such as C-X-C motif chemokine ligand 8 (CXCL8) and CXCL5, both in bEECs and in uterine tissue. From the experimental results of WB, qRT-PCR, and immunofluorescence, the expression of TLR4 and the phosphorylation of NF-κB p65 were markedly inhibited by catalpol compared with the LPS group. The inflammatory damage to the mouse uterus caused by LPS was greatly reduced and was accompanied by a decline in myeloperoxidase (MPO) activity. The results of this study suggest that catalpol can exert an anti-inflammatory impact on LPS-induced bEECs and mouse endometritis by inhibiting inflammation and activation of the TLR4/NF-κB signaling pathway.


Subject(s)
Endometritis/drug therapy , Inflammation/prevention & control , Iridoid Glucosides/pharmacology , NF-kappa B/physiology , Signal Transduction/drug effects , Toll-Like Receptor 4/physiology , Animals , Cattle , Chemokines/genetics , Cytokines/genetics , Epithelial Cells/drug effects , Female , Iridoid Glucosides/therapeutic use , Lipopolysaccharides/pharmacology , Mice
6.
Microb Pathog ; 136: 103721, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31494298

ABSTRACT

Acute lung Injury (ALI) is the clinical syndrome of parenchymal lung disease, leading to an extremely high mortality. The pathogenesis of ALI is suggested to be a consequence of uncontrolled inflammation. Lipopolysaccharide (LPS)-induced ALI mice model is often used for the mechanism. Studies show that TGF-beta activated kinase 1 (MAP3K7) binding protein 1/2 (TAB2) plays a crucial role in LPS-induced inflammation response. Furthermore, microRNA-142a-3p (miR-142a-3p) has been observed to be involved in inflammation-induced disease. Thus, we investigated the role of miR-142a-3p and TAB2 on LPS-induced ALI, which involved the TLR4/TAB2/NF-κB signaling. ALI and normal lung tissues were collected to access the relative expression of pro-inflammatory cytokines and miR-142a-3p. Histopathological examination and Wet to Dry weight ratios of lung tissues were used to access the establishment of ALI models. Raw264.7 cells were transfected with si-TAB2 or miR-142a-3p mimics to elucidate the role of TAB2 or miR-142a-3p in the inflammatory cascade in ALI. Additionally, the relationship between miR-142a-3p and TAB2 was validated by dual-luciferase report system. Our study discovered that miR-142-3p was up-regulated both in LPS-induced ALI mice model and RAW264.7 cells model. MiR-142a-3p mimics group experienced significant decrease in the secretion of pro-inflammatory cytokines as a result of the inhibition of NF-κB signaling pathway. Bioinformatics database showed that the adaptor protein, TAB2, was critical in this pathway and it is the target gene of miR-142a-3p. Their relation was first confirmed by us via dual-luciferase report system. Results of our study demonstrated that miR-142a-3p exerts as a protective role in LPS-induced ALI through down-regulation of NF-κB signaling pathway.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Adaptor Proteins, Signal Transducing/metabolism , Endotoxins/toxicity , Escherichia coli/pathogenicity , Lipopolysaccharides/toxicity , MicroRNAs/metabolism , Animals , Disease Models, Animal , Mice , Models, Theoretical , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction , Toll-Like Receptor 4/metabolism
7.
Microb Pathog ; 132: 302-312, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31059756

ABSTRACT

Acute lung injury (ALI) is clinically characterized by excessive inflammation leading to acute respiratory distress syndrome (ARDS), having high morbidity and mortality both in human and animals. Ginsenoside Rb1 (Rb1) is a major primary bioactive component extracted by Panax ginseng, which has numerous pharmacological functions such as anti-cancer, anti-inflammatory, and antioxidant. However, the anti-inflammatory effects of Rb1 in Staphylococcus aureus (S. aureus)-induced ALI in mice have not been investigated. The aim of the current study was to determine the anti-inflammatory influence of Rb1 on S. aureus-induced ALI in mice, and to explore its possible underlying principle mechanisms in RAW 264.7 macrophage cells. The results of physical morphology, histopathological variation and wet-to-dry weight ratio of lungs revealed that Rb1 significantly attenuated S. aureus-induced lung injury. Furthermore, qPCR results displayed that Rb1 inhibited IL-1ß, IL-6 and TNF-α production both in vivo and in vitro. The activation of Toll-like receptor 2 (TLR2) by S. aureus was inhibited by application of Rb1 as confirmed by results of immunofluorescence assay. The expression of NF-kB and MAPK signaling proteins revealed that Rb1 significantly attenuated the phosphorylation of p65, ERK, as well as JNK. Altogether, the results of this experiment presented that Rb1 has ability to protect S. aureus-induced ALI in mice by attenuating TLR-2-mediated NF-kB and MAPK signaling pathways. Consequently, Rb-1 might be a potential medicine in the treatment of S. aureus-induced lung inflammation.


Subject(s)
Acute Lung Injury/microbiology , Ginsenosides/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Staphylococcus aureus/drug effects , Animals , Cell Survival/drug effects , Disease Models, Animal , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lung/pathology , Male , Mice , Panax/chemistry , Pneumonia , RAW 264.7 Cells/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
8.
Oncotarget ; 8(17): 28481-28493, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28415707

ABSTRACT

Mastitis is a serious and prevalent disease caused by infection by pathogens such as Staphylococcus aureus. We evaluated the anti-inflammatory effects and mechanism of luteolin, a natural flavonoid with a wide range of pharmacological activities, in a mouse model of S. aureus mastitis. We also treated cultured mouse mammary epithelial cells (mMECs) with S. aureus and luteolin. Histopathological changes were examined by H&E staining and the levels of inflammatory cytokine proteins were analyzed using ELISAs. We determined mRNA levels with qPCR and the level of NF-κB and matrix metalloproteinase (MMP) proteins by Western blotting. The observed histopathological changes showed that luteolin protected mammary glands with S. aureus infection from tissue destruction and inflammatory cell infiltration. Luteolin inhibited the expression of TNF-α, IL-1ß, and IL-6, all of which were increased with S. aureus infection of mammary tissues and mMECs. S. aureus-induced TLR2 and TLR4 was suppressed by luteolin, as were levels of IκBα and NF-κB p65 phosphorylation and expression of MMP-2 and MMP-9. Levels of tissue inhibitor of metalloproteinases (TIMP)-1 and TIMP-2 were enhanced. These findings suggest luteolin is a potentially effective new treatment to reduce tissue damage and inflammation from S. aureus-induced mastitis.


Subject(s)
Luteolin/pharmacology , Mastitis/genetics , Mastitis/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , NF-kappa B/metabolism , Staphylococcal Infections/genetics , Staphylococcal Infections/metabolism , Staphylococcus aureus/physiology , Animals , Cell Line , Cell Survival/drug effects , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Luteolin/chemistry , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mastitis/drug therapy , Mastitis/microbiology , Mice , Signal Transduction/drug effects , Staphylococcal Infections/drug therapy , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
9.
Article in English | MEDLINE | ID: mdl-27293467

ABSTRACT

Endometritis is commonly caused by pathogenic microorganisms, including Staphylococcus aureus (S. aureus). Piperine, which is a natural medicine, has shown a variety of biological activities. To explore the effect and mechanism of piperine on S. aureus endometritis, a mouse model of S. aureus endometritis was successfully established in the present study. Histopathological changes were observed with H&E staining, cytokines were analyzed by ELISA, mRNA was analyzed by qPCR, and proteins were detected by western blot. The results showed that piperine could significantly alleviate inflammatory injury in S. aureus endometritis. The qPCR and ELISA results showed that piperine effectively reduced the S. aureus-induced overexpression of TNF-α, IL-1ß, and IL-6 but increased the expression of IL-10. The S. aureus-induced inflammation was related to TLR-2 and TLR-4 because the results showed that their expression was increased in S. aureus infection but then decreased with piperine treatment. To further confirm that piperine caused an anti-inflammatory response by targeting NF-κB and MAPKs, the expression of I-κB, p65, p38, ERK, and JNK was measured. The phosphorylation of I-κB, p65, p38, ERK, and JNK was inhibited by piperine in a dose-dependent manner. All of the results indicated that piperine may be a potential anti-inflammatory drug both in endometritis and in other S. aureus-induced diseases.

10.
Asian Pac J Cancer Prev ; 15(4): 1517-20, 2014.
Article in English | MEDLINE | ID: mdl-24641360

ABSTRACT

AIM: To investigate effects of sulforaphane on the BIU87 cell line and underlying mechanisms involving IGFBP-3. METHODS: Both BIU87 and IGFBP-3-silenced BIU87 cells were treated with sulforaphane. Cell proliferation was detected by MTT assay. Cell cycle and apoptosis were determined via flow cytometry. Quantitative polymerase chain reaction and Western blotting were applied to analyze the expression of IGFBP-3 and NF-κB at both mRNA and protein levels. RESULTS: Sulforaphane (80 µM) treatment could inhibit cell proliferation, inducing apoptosis and cell cycle arrest at G2/M phase. All these effects could be antagonized by IGFBP-3 silencing. Furthermore, sulforaphane (80 µM) could down-regulate NF-κB expression while elevating that of IGFBP-3. CONCLUSIONS: Sulforaphane could suppress the proliferation of BIU87 cells via enhancing IGFBP-3 expression, which negatively regulating the NF-κB signaling pathway.


Subject(s)
Cell Proliferation/drug effects , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Isothiocyanates/pharmacology , Urinary Bladder Neoplasms/drug therapy , Anticarcinogenic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , M Phase Cell Cycle Checkpoints/drug effects , NF-kappa B/biosynthesis , NF-kappa B/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Signal Transduction/drug effects , Sulfoxides
SELECTION OF CITATIONS
SEARCH DETAIL
...