Search
To search the journal, enter a term in the search bar. If you'd like to find specific authors, titles, or abstracts, use the advanced search to the right.
Search Results
5 results found. Results per page: [ 20 ][ 40 ][ 60 ][ 80 ][ 100 ][ 200 ][ 300 ]
Sort by: [ Publication Date ][ Score ]
Year of publication: [ 2025 ][ 2024 ][ 2023 ][ 2022 ][ 2021 ][ 2020 ][ 2019 ][ 2018 ][ 2017 ][ 2016 ][ 2015 ][ 2014 ][ 2013 ][ 2012 ][ 2011 ][ 2010 ][ 2009 ][ Any ]
-
Research Paper Volume 13, Issue 17 pp 21142-21154
Serglycin promotes proliferation, migration, and invasion via the JAK/STAT signaling pathway in osteosarcoma
Relevance score: 15.457615Bin Lv, Guangyu Gao, Yuhong Guo, Zhiping Zhang, Renfeng Liu, Zhengzai Dai, Cheng Ju, Yiping Liang, Xiaofeng Tang, Min Tang, Xiao-Bin Lv
Keywords: SRGN, osteosarcoma, GEO, bioinformatics analysis, JAK/STAT
Published in Aging on September 7, 2021
-
Research Paper Volume 13, Issue 4 pp 5185-5196
A short deletion in the DNA-binding domain of STAT3 suppresses growth and progression of colon cancer cells
Relevance score: 12.610607Yi-Jia Xiong, Dong-Yang Liu, Rong-Rong Shen, Yong Xiong
Keywords: STAT3, gain of function, JAK/STAT pathway, colon cancer
Published in Aging on February 1, 2021
-
Research Paper Volume 12, Issue 11 pp 10896-10911
SPTBN1 suppresses the progression of epithelial ovarian cancer via SOCS3-mediated blockade of the JAK/STAT3 signaling pathway
Relevance score: 13.416022Mo Chen, Jia Zeng, Shuyi Chen, Jiajia Li, Huijie Wu, Xuhui Dong, Yuan Lei, Xiuling Zhi, Liangqing Yao
Keywords: SPTBN1, epithelial ovarian cancer, SOCS3, JAK/STAT pathway, EMT
Published in Aging on June 8, 2020
SPTBN1 is closely related to the progression of epithelial ovarian cancer. (A) Analysis of TCGA data. The expression of SPTBN1 was significantly decreased as EOC staging increased. NS: stage 1 vs stage 2, 3, and 4; *P<0.05 vs stage 3 and 4; #P<0.05 vs stage 3. (B, C) The progression-free survival (PFS) and overall survival (OS) rates were retrieved from the TCGA dataset (probe 214856_at) and compared between the low-SPTBN1 and high-SPTBN1 ovarian cancer patients. The low-SPTBN1 group had shorter PFS than the high-SPTBN1 group. (D) Expression of SPTBN1 in 8 ovarian cancer cell lines. The expression of SPTBN1 in high-metastasis HO8910-PM cells was lower than that in conventional HO8910 cells, suggesting that the decrease in SPTBN1 expression may be correlated with the increased incidence of malignancy of epithelial ovarian cancer.
SPTBN1 inhibits the growth of epithelial ovarian cancer cells in vitro and in vivo. (A, B) The protein level of SPTBN1 was detected by western blot. (C, D) The mRNA level of SPTBN1 was analyzed by real-time PCR. SPTBN1 was overexpressed in HO8910 cells but decreased in A2780 cells after transfection with the SPTBN1-V5 or SPTBN1-sh plasmid, respectively. *P < 0.05 vs Con-V5, **P < 0.01 vs Con-sh, n=3. (E, F) Cell viability was evaluated by CCK8 assay. **P < 0.01 vs Con-V5, *P < 0.05 vs Con-sh, n=3. (G) Cell proliferation was assessed by colony formation assay. Loss of SPTBN1 promotes the proliferation of A2780 and HO8910 cells. *P < 0.05 vs Con-sh, n=3; (H–J) Mouse xenograft tumors derived from A2780 cells. Loss of SPTBN1 promotes the growth of epithelial ovarian cancer cells in vivo. (H) Images of mice with tumors (upper) and harvested tumors for each treatment group (lower). (I) Tumor growth curves. (J) Tumor weight at sacrifice. *P < 0.05; **P < 0.01 vs LV-Con.
SPTBN1 inhibits the migration and EMT of epithelial ovarian cancer cells. (A, B) Assessments of cell migration. Overexpression of SPTBN1 inhibits the migration of HO8910 cells (A), while downregulation of SPTBN1 promotes the migration of A2780 cells (B). (C, D) Comparison of EMT-related markers at the protein level (C) and mRNA level (D). * P < 0.05, **P < 0.01 vs Con-sh, n=3.
Positive correlation between the expression of SPTBN1 and SOCS3 in epithelial ovarian cancer patients and cells. (A, B) Positive correlation between SPTBN1 and SOCS3 expression was investigated by GEPIA (gene expression profiling interactive analysis,
http://gepia.cancer-pku.cn ) based on the TCGA dataset (A) and observed by IHC staining of ovarian cancer patient samples (n=11) with 40× magnification (B). (C) The protein level of SOCS3 was decreased, and the phosphorylated STAT3 level was increased when SPTBN1 was downregulated. (D) SPTBN1 regulated SOCS3 expression at the mRNA level. * P < 0.05, ** P < 0.01 vs LV-RFP. (E) ChIP-qPCR was performed in HO8910 cells with anti-SPTBN1 and anti-Smads2/3 antibodies to determine the enrichment of SOCS3 promoter region sequences in the obtained ChIP DNA. *P < 0.05 vs Con-si.Loss of SPTBN1 activates the JAK/STAT3 signaling pathway through downregulation of SOCS3. (A) Assessments of JAK/STAT signaling pathway-associated proteins by western blot after SPTBN1 knockdown cooperated with SOCS3 overexpression in HO8910 cells (left), and SPTBN1 overexpression cooperated with SOCS3 knockdown (right). (B) DNA gel electrophoresis after PCR. Mouse embryonic fibroblasts (MEFs) were cultured from SPTBN1-/- embryos (n=2), SPTBN1+/- embryos (n=3) and wild-type embryos (n=2). The genotypes of MEFs were identified by PCR and DNA gel electrophoresis. Lanes 1 and 2: SPTBN1-/- MEFs; lanes 3, 4, and 5: SPTBN1+/- MEFs; lanes 6 and 7: SPTBN1-/- MEFs. (C) Assessments of EMT and JAK/STAT3 signaling pathway-associated proteins by western blot in SPTBN1-/-, SPTBN1+/-, and SPTBN1+/+ MEFs. Lanes 1 and 2: SPTBN1-/- MEFs; lanes 3, 4, and 5: SPTBN1+/- MEFs; lanes 6 and 7: SPTBN1-/- MEFs. Loss of SPTBN1 can promote EMT, inhibit SOCS3 and activate the JAK/STAT signaling pathway.
SOCS3 overexpression or the JAK2 inhibitor reverses the inhibitory effects of SPTBN1 on cell viability and migration. (A, B) In vitro cell migration assay. * P < 0.05 vs LV-RFP+LV-GFP, #P < 0.05 vs LV-SPTBN1sh+LV-GFP, n=3. (C, D) Comparison of protein (C) and mRNA (D) levels of the EMT-related proteins E-cadherin (E-cad) and Vimentin (Vim). The expression of SOCS3 and E-cadherin was decreased and Vimentin was increased by the loss of SPTBN1, while SOCS3 overexpression reversed the effects of the loss of SPTBN1. *P<0.05 **P<0.01 vs LV-RFP+LV-GFP, ##P<0.01 vs LV-SPTBN1sh+LV-GFP, n=3. (E–H) Cell viability was determined by CCK8 assay. SOCS3 overexpression reversed the enhanced cell viability due to the loss of SPTBN1 in A2780 (E) and HO8910 cells (F). #P<0.05, ##P<0.01 vs LV-RFP+LV-GFP, *P<0.05 vs LV-SPTBN1sh+LV-GFP, n=3. The JAK2 inhibitor Ag490 or tofacitinib (Tofa) inhibited cell viability and reversed the promoting effect of the loss of SPTBN1 in A2780 (G) and HO8910 cells (H). *P<0.05, **P<0.01 vs LV-RFP, #P<0.05 vs LV-SPTBN1sh, n = 3.
-
Research Paper Volume 12, Issue 10 pp 9066-9084
Overexpression of miR-142-5p inhibits the progression of nonalcoholic steatohepatitis by targeting TSLP and inhibiting JAK-STAT signaling pathway
Relevance score: 12.89825Chao Zhou, Pu Wang, Lei Lei, Yi Huang, Yue Wu
Keywords: non-alcoholic steatohepatitis, miR142-5p, TSLP, JAK-STAT signaling pathway
Published in Aging on May 15, 2020
TSLP was up-regulated and miR-142-5p was downregulated in NASH. (A) Heatmap showing differentially expressed genes between health and NASH group. (B) Heatmap showing differentially expressed miRNAs between health and NASH group. The expression fold in NASH groups was calculated compared with the health groups. Log2|FC|>1 and adj. P < 0.05 was considered statistically significant.
JAK-STAT pathway was actived in NASH patients and verification of the target gene. (A) Dotplot suggest the distributions of some biological pathway gene sets. The size of the circle represents the count value. The gradient changes of color represented adjust P values. (B) KEGG analysis of JAK-STAT pathway in NASH. (C) Expression of TSLP in NASH disease model. *P < 0.05, compared with control group or WT group. (D) Venn diagram showing the overlap between dysregulated miRNAs and miRNAs that target at TSLP. (E) RIP assays of the enrichment of Ago2 on TSLP and miR-142-5p relative to IgG in liver. **P < 0.01, compared with IgG group. (F) The miR-142-5p binding sites on TSLP were predicted by bioinformatics. TSLP wild-type form (TSLP-wt) and mutated form (TSLP-mut) were displayed on the left panel. Dual-luciferase reporter assay was conducted to identify the target relationship between miR-142-5p and TSLP. All data were means ± SD.
The expression of miR-142-5p and TSLP and the function of miR-142-5p to TSLP in the liver of mouse model. (A) The relative expression of miR-142-5p in the liver of different group. (B) Relative mRNA expression of TSLP in the liver of different mouse group. (C) Protein expression of TSLP in the liver of different mouse group; WT group: wild-type mice fed a chow diet (CD); blank group: Ldlr−/− mice fed an high-fat diet (HFD) treated with phosphate buffer saline (PBS); NC group: Ldlr−/− mice fed an HFD treated with control agonis; A142 group: Ldlr−/− mice fed an HFD treated with agonist of miR-142-5p; A142-C group: Ldlr−/− mice fed an HFD treated with miR-142-5p agonist and JAK-STAT signaling pathway activator-colivelin; A142-TSLP group: Ldlr−/− mice fed an HFD treated with miR-142-5p agonist and AAV/TSLP. AAV: adeno-associated virus. n=6. *P < 0.05; **P < 0.01, compared with WT group; #P< 0.05; ##P < 0.01, compared with NC group; ^P < 0.05, compared with A142 group. All data were means ± SD.
The expression of inflammatory factors and JAK-STAT signaling pathway biomarkers in the NASH mouse model. (A) The mRNA expression level of tumor necrosis factor (TNF)-α. (B) The mRNA expression level of interferon (IFN)-β. (C) The mRNA expression level of liver monocyte chemoattractant protein-1 (MCP1). (D) The mRNA expression level of IL-4. (E) The mRNA expression level of IL-6. (F) The expression level of TG in the liver. (G) The protein expression of the biomarkers of JAK-STAT signaling pathway. All data were means ± SD. *P < 0.05, compared with WT group; #P < 0.05, compared with NC group; ^P < 0.05, compared with A142 group.
miR-142-5p reduced liver injury, liver fatty degeneration and liver fibrosis, which could be rescued by JAK-STAT signaling pathway activator or AAV/TSLP. (A) The level of alanine aminotransferase (ALT) secreted into serum. (B) The level of aspartate aminotransferase (AST) secreted into serum. (C, E) H&E staining for liver fatty degeneration. Original magnification ×200. (D, F) The representative images of collagen deposition in Masson’s trichrome. The expression of fibrosis-related genes. Original magnification ×200. (G) The mRNA expression level of Tgfβ and (H) The mRNA expression level of Collagen-1 α2. *P < 0.05, compared with WT group; #P < 0.05, compared with NC group; ^P < 0.05, compared with A142 group. All data were means ± SD.
The effect of molecular interference sequences on the expression of miR-142-5p and TSLP. (A) Relative expression level of miR-142-5p in different groups. (B) The relative mRNA expression of TSLP. (C) The protein expression level of TSLP. WT group: wild-type mice fed a chow diet; blank group: Ldlr−/− mice fed an HFD treated with phosphate buffer saline; NC group: Ldlr−/− mice fed an HFD treated with sh-control; sh-TSLP group: Ldlr−/− mice fed an HFD treated with sh-TSLP; sh-TSLP-C group: Ldlr−/− mice fed an HFD treated with sh-TSLP and colivelin (JAK-STAT signaling pathway activator). Each group contained 6 mice. *P < 0.05, **P < 0.01, compared with WT group; #P < 0.05, compared with NC group. All data were means ± SD.
Downregulating TSLP inhibited the expression of inflammatory factors and JAK-STAT pathway biomarkers, which could be rescued by JAK-STAT signaling pathway activator. (A) The mRNA expression level of tumor necrosis factor (TNF)-α. (B) The mRNA expression level of interferon (IFN)-β. (C) The mRNA expression level of liver monocyte chemoattractant protein-1 (MCP1). (D) The mRNA expression level of IL-4. (E) The mRNA expression level of IL-6. (F) The expression level of TG in the liver. (G) The protein expression of the biomarkers of JAK-STAT signaling pathway. *P < 0.05; **P < 0.01, compared with WT group; #P < 0.05; ##P < 0.01, compared with NC group; &P < 0.05; &&P < 0.01, compared with sh-TSLP group. All data were means ± SD.
Knockdown TSLP reduced liver injury, liver fatty degeneration and liver fibrosis, which could be rescued by JAK-STAT signaling pathway activator. (A) The level of alanine aminotransferase (ALT) secreted into serum. (B) The level of aspartate aminotransferase (AST) secreted into serum. (C) Liver fatty degeneration was detected by H&E staining; (D) The degree of fibrosis was shown by Masson’s trichrome Original magnification ×200. (E) The mRNA expression level of Tgfβ and (F) The mRNA expression level of Collagen-1α2. *P < 0.05, compared with WT group; #P < 0.05, compared with NC group; &P < 0.05, compared with sh-TSLP group. All data were mean ± SD.
-
Research Paper Volume 9, Issue 11 pp 2411-2435
TNFα-senescence initiates a STAT-dependent positive feedback loop, leading to a sustained interferon signature, DNA damage, and cytokine secretion
Relevance score: 12.064303Renuka Kandhaya-Pillai, Francesc Miro-Mur, Jaume Alijotas-Reig, Tamara Tchkonia, James L. Kirkland, Simo Schwartz
Keywords: senescence, inflammation, DNA-damage, interferon response genes, JAK/STAT pathway
Published in Aging on November 22, 2017
TNFα induces senescence and DNA damage in HUVECs. (A) Long-term growth curve of cells exposed to recombinant human TNFα (5ng/ml). Untreated cells were used as controls. Population doubling and doubling times were calculated based on cell density at confluence. Data represent mean values from 3 independent experiments. (B) The percentage of BrdU-positive cells was determined by FACS analysis in cells untreated or chronically treated with TNFα at the concentration indicated. (C) Western blot analysis of p21, p16, and actin in cells treated with TNFα 5ng/ml for the indicated times. (D) SA-β-gal activity in TNFα (5ng/ml)-treated or control cells for the indicated number of days. (E) Percentages of SA-β-gal-positive cells in control or TNFα-treated cultures. The data represent 2 independent counts of 200 cells from 3 independent experiments. (F) Intracellular ROS levels were monitored by 2',7’-dichlorodihydrofluorescein diacetate staining followed by flow cytometry. Bar graph represents percentage of DCFDA-positive cells treated with TNFα or medium alone. (G) Immunofluorescence detection of γH2AX foci in controls or cells treated with TNFα (5ng/ml) for indicated days. Data in A, B, E, and F represent mean value ± standard deviation (s.d.) from n=3, 2, 3, and 2 independent experiments, respectively.
Top canonical pathways identified by IPA analysis in TNFα-induced senescence. (A) Bar chart represents the top canonical signaling pathways that were influenced during TNFα-induced senescence. p-values were determined using Fisher´s exact test with a threshold value of >0.05. Ratios represent the number of genes that mapped to a specific canonical pathway divided by the total number of genes that make up the respective pathway. (B) Relative mRNA expression of SASP components in cells treated with TNFα at 6, 16, or 26 days compared to untreated cells. (C) Relative mRNA expression of p16 and p21 in cells treated with TNFα (5ng/ml) compared to untreated cells. Results are mean ± standard deviation of n=2 independent experiments.
Activation of the canonical JAK/ STAT pathway. Pathway analysis using IPA reveals canonical activation of STAT1 acts as a central regulator for JAK/ STAT-mediated interferon response genes during TNFα-induced senescence. The red symbols represent up-regulated genes, with the intensity of node color indicating degree of up-regulation and white indicating genes absent from the list. Shape of nodes denotes functions of gene products.
Prolonged activation of JAK/ STAT signaling in TNFα-induced senescence. (A) Immunoblot detection of p-Ser727-STAT1 and total STAT1 in cells exposed to TNFα 20ng/ml for the indicated times. (B) SA-β-gal activity in TNFα (20ng/ml)-treated or control cells for 3 or 6 days. (C) Immunoblot detection of p-Ser727-STAT1, total STAT1, p-Tyr705-STAT3, and total STAT3 in cells exposed to TNFα (5ng/ml) for the indicated intervals. (D) Immunoblot detection of pSTAT1, total STAT1, pSTAT3, and total STAT3 in cells stimulated with IL6 (10ng/ml) or IFNγ (1ng/ml) for the indicated intervals. (E) Secretion of IFNγ/IL6 quantified by ELISA in conditioned medium collected in the presence or absence of TNFα. (F) Immunoblot detection of p-Ser727-STAT1, p-Tyr705-STAT3, STAT1, and STAT3 in cells treated with conditioned medium (CM) (cell free-culture supernatants from control and cells stimulated with TNFα for 3 days transferred after 1:4 dilution with fresh culture medium) from TNFα-induced senescent cells or from non-senescent cells for the indicated times
Persistent activation of STAT1/3. Cells were exposed to TNFα (20ng/ml) for 3 days, then washed to remove the residual TNFα and cultured for 3 days in the absence of exogenous TNFα (TNFα-PST). Parallel cultures were exposed to exogenous TNFα throughout the experiment. (A) Levels of p-Ser727-STAT1, p-Tyr705-STAT3, and total STAT3 proteins were quantified by immunoblot. (B) Secretion of IL-6/IFNγ was assessed in culture supernatants from cells treated with TNFα as indicated. (C) Immunodetection of ROS production and γH2AX foci in control or cells treated with TNFα or TNFα-post-stimulated (PST), as indicated. (D) Real-time gene expression of IRF1 and MX1 in cells exposed to TNFα or TNFα-PST for 3 days. Results were normalized to internal control TBP and are shown relative to untreated cells. (E) SA-β-gal activity in TNFα-treated cells for 3 days or in cells treated with TNFα (20ng/ml) for three days, then washed to remove residual TNFα and left untreated for another 3 days. (F) mRNA expression of p21 and p16 quantified by real-time PCR in cells exposed to TNFα or TNFα-PST for 3 days. Data in D and F represent mean value of ± sd from 2 independent experiments.
JAK2 inhibitor decreases TNFα-mediated inflammation, ROS levels, and interferon signature. Cells were exposed to TNFα alone, TNFα in combination with AG490 (30μM), or AG490 alone for 3 days. (A) Immunoblot detection of p-Ser727-STAT1, p-Tyr705-STAT3, and total STAT1 and STAT3. (B) Secretion of IL6 was estimated by ELISA in culture supernatants from cells treated with TNFα alone, or in combination with AG490, or AG490 alone for 3 days. (C) FACS analysis of ROS levels in cells stimulated either with TNFα, TNFα along with AG490, or AG490 alone for 3 days using DCFDA staining 2',7’-dichlorofluorescein (DCF) positive cells were analyzed. Inhibition of STAT signals modulates senescence. (D) Cell cycle analysis using BrdU and 7- aminoactinomycin D (7-AAD) staining in cells exposed to TNFα, TNFα in combination with AG490, or AG490 alone for 3 days. (E) Percentage of SA-β-gal-positive cells. Quantification of SA-β-gal activity in cells stimulated with TNFα 20ng/ml, TNFα in combination with AG490, or AG490 alone for 3 days. The data represent means of 3 independent counts of 200 cells from 2 independent experiments. (F) Effect of AG490 on cell cycle regulatory proteins. Western analysis performed using cells treated with AG490 for 3 days and blotted against anti-p21, CDK2, and NDC80. Actin serves as loading control.
Model of mechanisms involved in TNFα-induced senescence of HUVECs. (A) TNFα activates JAK/STAT and p38 signaling pathways, which mediate increased expression of STAT1/3 phosphorylation. Activation of the JAK pathway leads to persistent phosphorylation of STAT1/3 signaling, which together with ROS, interferon genes, and other SASP components, drives a positive auto-regulatory loop, leading to sustained inflammation and stable senescence. (B) Inhibition of STAT1/3 with the JAK inhibitor AG490 decreased ROS and IL-6 production and decreased expression of interferon response genes. On the other hand, blockade of STAT1/3 expression decreased S phase entry of cells and increased p21 expression, leading to senescence.