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
91 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 ]
Page:
-
Research Paper Volume 14, Issue 4 pp 1627-1650
The telomere-mitochondrial axis of aging in newborns
Relevance score: 4.1488914Charlotte Van Der Stukken, Tim S. Nawrot, Rossella Alfano, Congrong Wang, Sabine A.S. Langie, Michelle Plusquin, Bram G. Janssen, Dries S. Martens
Keywords: telomere length, mitochondrial DNA content, p53, PGC-1α, aging
Published in Aging on February 15, 2022
-
Research Paper Volume 13, Issue 18 pp 22242-22255
miR-21 regulates ischemic neuronal injury via the p53/Bcl-2/Bax signaling pathway
Relevance score: 4.1488914Honglin Yan, Wenxian Huang, Jie Rao, Jingping Yuan
Keywords: cerebral ischemia, neuronal injury, miR-21, p53, Bcl-2/Bax
Published in Aging on September 22, 2021
-
Research Paper Volume 13, Issue 18 pp 22109-22119
LHX9, a p53-binding protein, inhibits the progression of glioma by suppressing glycolysis
Relevance score: 5.3065157Xiangying Luo, Jianwei Ge, Tao Chen, Jinfang Liu, Ziyuan Liu, Changlong Bi, Song Lan
Keywords: LHX9, p53, glioma, glycolysis, PGK1
Published in Aging on September 17, 2021
-
Research Paper Volume 13, Issue 18 pp 21914-21940
Metformin-induced chemosensitization to cisplatin depends on P53 status and is inhibited by Jarid1b overexpression in non-small cell lung cancer cells
Relevance score: 5.649303Tharcisio Citrangulo Tortelli, Rodrigo Esaki Tamura, Mara de Souza Junqueira, Janio da Silva Mororó, Silvina Odete Bustos, Renato Jose Mendonça Natalino, Shonagh Russell, Laurent Désaubry, Bryan Eric Strauss, Roger Chammas
Keywords: metformin, cisplatin, Jarid1b, p53, NSCLC
Published in Aging on September 16, 2021
-
Research Paper Volume 13, Issue 17 pp 21587-21598
FOXN3 inhibits cell proliferation and invasion via modulating the AKT/MDM2/p53 axis in human glioma
Relevance score: 4.423868Chaojia Wang, Hanjun Tu, Ling Yang, Chunming Ma, Juntao Hu, Jie Luo, Hui Wang
Keywords: FOXN3, cell proliferation, cell migration, glioma, AKT/MDM2/p53 pathway
Published in Aging on September 12, 2021
-
Research Paper
Local anesthetic levobupivacaine induces ferroptosis and inhibits progression by up-regulating p53 in non-small cell lung cancer
Relevance score: 5.649303Minghua Meng, Meiling Huang, Cuicui Liu, Jiming Wang, Weihua Ren, Shaomin Cui, Jianhua Gu, Jinlan Xie, Bin Ma, Guangzhu Yang, Shuncheng He
Keywords: NSCLC, progression, levobupivacaine, ferroptosis, p53
Published in Aging on June 27, 2021
-
Priority Research Paper Volume 13, Issue 10 pp 13380-13392
Cdkn1a transcript variant 2 is a marker of aging and cellular senescence
Relevance score: 4.8441844José Alberto López-Domínguez, Sandra Rodríguez-López, Ulises Ahumada-Castro, Pierre-Yves Desprez, Maria Konovalenko, Remi-Martin Laberge, César Cárdenas, José Manuel Villalba, Judith Campisi
Keywords: p21, p53, mouse dermal fibroblast, ionizing radiation, doxorubicin
Published in Aging on May 25, 2021
-
Research Paper Volume 13, Issue 9 pp 12537-12551
Silencing long noncoding RNA NEAT1 alleviates acute liver failure via the EZH2-mediated microRNA-139/PUMA axis
Relevance score: 2.9862788Qiang Wang, Shu Liu, Huan Wang, Lian Liu, Sheng Zhang, Yingzi Ming, Yujun Zhao, Ke Cheng
Keywords: acute liver failure, long noncoding RNA nuclear enriched abundant transcript 1, microRNA-139, p53-upregulated modulator of apoptosis, methylation
Published in Aging on April 26, 2021
-
Research Paper Volume 13, Issue 8 pp 11774-11785
Inhibition of sestrin 1 alleviates polycystic ovary syndrome by decreasing autophagy
Relevance score: 4.742327Xiaoyuan Xu, Xinli Song, Xiaohua Xu, Yanluan Zheng, Lan Xu, Ling Shen
Keywords: sestrin 1, polycystic ovary syndrome, p53, autophagy
Published in Aging on April 22, 2021
-
Research Paper Volume 13, Issue 5 pp 7180-7189
Neuroprotective effect of hydrogen sulfide against glutamate-induced oxidative stress is mediated via the p53/glutaminase 2 pathway after traumatic brain injury
Relevance score: 4.312628Jianping Sun, Xiaoyu Li, Xiaoyu Gu, Hailong Du, Gengshen Zhang, Jianliang Wu, Feng Wang
Keywords: traumatic brain injury, hydrogen sulfide, glutamate, oxidative stress, p53
Published in Aging on February 26, 2021
-
Research Paper Volume 13, Issue 4 pp 5986-6009
Overexpression of hsa_circ_0002874 promotes resistance of non-small cell lung cancer to paclitaxel by modulating miR-1273f/MDM2/p53 pathway
Relevance score: 5.014036Jianhao Xu, Liwei Ni, Fenglun Zhao, Xiaoxiao Dai, Jialong Tao, Jia Pan, Aiming Shi, Zhu Shen, Cunjin Su, Yusong Zhang
Keywords: hsa_circ_0002874, miR1273f, MDM2, P53, paclitaxel-resistance
Published in Aging on February 17, 2021
-
Research Paper Volume 13, Issue 4 pp 4831-4849
An aging and p53 related marker: HOXA5 promoter methylation negatively correlates with mRNA and protein expression in old age
Relevance score: 4.7651396Laura-Kim Feiner, Sascha Tierling, Sebastian Holländer, Matthias Glanemann, Claudia Rubie
Keywords: ageing, DNA methylation, HOXA5, p53, gene expression
Published in Aging on February 5, 2021
-
Research Paper Volume 12, Issue 23 pp 23647-23667
Identification of the novel Np17 oncogene in human leukemia
Relevance score: 5.2671304Bowen Wu, Yichao Gan, Ying Xu, Zhaoxing Wu, Ganyu Xu, Ping Wang, Chen Wang, Zhipeng Meng, Mengyuan Li, Jiawei Zhang, Haifeng Zhuang, Xuzhao Zhang, Linlin Yang, Jinfan Li, Xiaoxian Gan, Xiaofang Yu, Wendong Huang, Ying Gu, Rongzhen Xu
Keywords: Np17, leukemia, oncogene, P53, Np9
Published in Aging on November 21, 2020
-
Research Paper Volume 12, Issue 21 pp 22112-22121
Autophagy modulates mesenchymal-to-endothelial transition via p53
Relevance score: 4.338167Jie Hu, Shuting Kong, Tiancheng Dong, Zhiwei Lin, Qihao Zhang, Xingxing Chen, Yongsheng Gong, Xiaofang Fan, Mingyu He, Hao Zhou
Keywords: mesenchymal-to-endothelial transition, autophagy, p53, endothelial cells, fibrosis
Published in Aging on November 13, 2020
-
Research Paper Volume 12, Issue 20 pp 20702-20727
Stress-induced p53 drives BAG5 cochaperone expression to control α-synuclein aggregation in Parkinson's disease
Relevance score: 4.338167Huan-Yun Chen, Chin-Hsien Lin, Shu-Chun Teng
Keywords: BAG5, cochaperone, p53, α-synuclein, Parkinson's disease
Published in Aging on October 21, 2020
-
Priority Research Paper Volume 12, Issue 18 pp 17800-17814
p21 can be a barrier to ferroptosis independent of p53
Relevance score: 4.7651396Divya Venkatesh, Brent R. Stockwell, Carol Prives
Keywords: p21, ferroptosis, cancer, p53-independent, organ damage
Published in Aging on September 24, 2020
Regulation of ferroptosis by p53 is highly context specific. (A) The indicated cell lines were categorized based on the relative amount of cell death observed in response to 24 hours treatment with erastin in a 6-well format. After 24 hours of treatment, the sensitive cell lines had an EC50 of less than 2 μM of erastin, while the moderately sensitive cell lines had an EC50 that was greater than 2 μM, but lesser than 10 μM of erastin. In the resistant cell lines, erastin did not achieve 50% killing at this time point. (B, C) Viability of isogenic cell lines with wild-type (WT) p53 or no p53 (KO) in (B) HCT-116 and (C) RKO when treated with indicated doses of either doxorubicin (left panel) or IKE (right panel) for 24 hours. (D) Immunoblot showing p53 and p21 protein levels in HCT-116 and RKO cells. Multiple replicates of the wild-type and p53 KO cell lines cultured in separate dishes were used. Actin was used as a loading control. The data in (B, C) represent the mean ± SE for two of four independent experiments. The viability data have been normalized to that of the DMSO control.
p21 protein is differentially regulated between cells that are sensitive and resistant in response to ferroptosis. (A–E) Impact of treatment with erastin/IKE on the protein levels of p21 and p53. (A) HT-1080 cells, (B) SK-HEP1 cells and (C) U2OS cells were treated for 16 hours whereas (D) H1299 cells and (E) HCT116 cells (+/+ and -/- isogenic lines with respect to p53 status) were treated for 48 hours.
p21 mRNA is upregulated in both ferroptosis-sensitive and ferroptosis-resistant cells after treatment with IKE. (A–C) Left panels: Impact of IKE treatment on the mRNA levels of p21. (A) HT-1080 cells were treated for 16 hours while (B, C) H1299 and HCT-116 cells were treated for 48 hours. mRNA levels of ptgs2 and chac1 were measured in (A, B) as markers of ferroptosis. Right panels: the corresponding protein levels in the cells used in the left panels are shown. The data in left panels of (A–C) represent the mean ± SE for three biological replicates with two technical replicates each.
Altering p21 protein levels changes the sensitivity of cells to ferroptosis. (A, B) HCT116 (A) or H1299 (B) cells were transfected with two different siRNAs (#1, #2) directed against p21 mRNA for 24 hours, and then treated with erastin or IKE as indicated for an additional 48 hours. As a control, cells were transfected with luciferase siRNA a (siCtrl/C). The right panels in A and B show the corresponding changes in p21 protein levels. (C) HCT-116 cells and HCT116 p21 (-/-) cells were treated with increasing doses of either erastin (left panel) or IKE (right panel) for 48 hours. (D) Left panel: Viability of HT-1080 cells that were transfected with the indicated plasmids expressing p21 variants or an empty vector and then treated with either DMSO or IKE for 48 hours. The panel on the right shows the corresponding immunoblot detecting p21 protein levels. The data in (A, B) represent the mean ± SE for two of three independent experiments, in (C) represent the mean ± SE for two out of four independent experiments, in (D) represent the mean ± SE for three independent experiments. The viability data have been normalized to the DMSO control in (A–C) and to their respective untreated control in (D).
-
Research Paper Volume 12, Issue 18 pp 17990-18007
SIRT1 activation by minocycline on regulation of microglial polarization homeostasis
Relevance score: 4.742327Ling-Hsuan Wu, Bor-Ren Huang, Sheng-Wei Lai, Chingju Lin, Hsiao-Yun Lin, Liang-Yo Yang, Dah-Yuu Lu
Keywords: microglia, SIRT1, minocycline, microglial polarization, acetyl-p53
Published in Aging on September 23, 2020
-
Research Paper Volume 12, Issue 14 pp 14990-15001
Lysine-specific histone demethylase 1B (LSD2/KDM1B) represses p53 expression to promote proliferation and inhibit apoptosis in colorectal cancer through LSD2-mediated H3K4me2 demethylation
Relevance score: 5.3527546Shaoxin Cai, Jinsi Wang, Wei Zeng, Xuefei Cheng, Lihang Liu, Weihua Li
Keywords: colorectal cancer, proliferation, apoptosis, LSD2, p53
Published in Aging on July 29, 2020
LSD2 expression levels in human colorectal cancer (CRC) tissue and cells. (A) The ONCOMINE database was used to examine mRNA expression of LSD2 in CRC tissue. (B) Real-time Polymerase chain reaction (PCR) to evaluate mRNA expression of LSD2 in clinical CRC specimens relative to normal tissues (n=36). Data were expressed as means ± SEM. (C, D) Western Blot evaluation of LSD2 protein expression in CRC and normal tissue. (E) LSD2 protein expression in normal and CRC tissue was analysed by IHC. (F) Pearson’s χ2 tests to assess the association between colon tissue type and LSD2 expression by IHC(n=36). (G) Western blotting to assess LSD2 expression in CRC cell lines.
LSD2 over-expression promotes CRC cell proliferation. (A) Western blot detection of LSD2 and GAPDH (loading control) protein expression in Caco2 and SW1116 cells with control or LSD2. (B, C) BrdU/PI assay to detect and quantify DNA synthesis in Caco2 and SW1116 cells with control or LSD2. **p < 0.01. (D) Cell Counting Kit-8 (CCK8) assay to measure the viability of Caco2 and SW1116 cells with control or LSD2. (E, F) Colony formation assays and quantification in approximately 500 Caco2 and SW1116 cells with control or LSD2. Data were expressed as means ± SEM, **p < 0.01.
LSD2 down-regulation inhibits CRC cell proliferation. (A) Western blot showing protein expression of LSD2 and GAPDH (loading control) in HCT-116 and LoVo cells with control or sh-LSD2. (B, C) BrdU/PI assay to detect and quantification of DNA synthesis in HCT-116 and LoVo cells with control or sh-LSD2. **p < 0.01. (D) Cell Counting Kit-8 (CCK8) assay to detect the viability of HCT-116 and LoVo cells with control or sh-LSD2. (E, F) Colony formation assays and quantification in approximately 500 HCT-116 and LoVo cells with control or sh-LSD2. Data were expressed as means ± SEM, **p < 0.01.
LSD2 promoted CRC cells proliferation in vivo. (A) Effects of LSD2 down-expression on tumor growth in a xenograft mouse model. Empty vector or sh-LSD2 was transfected into LoVo cells, which were injected in the nude mice (n = 6), and the tumors were obtained at day 35 and was determined by measuring their tumor volume (B) and tumor weight (C). Data were expressed as means ± SEM. (D) The tumor sections were under IHC staining using antibodies against LSD2, p53, p21,ki-67, *P < 0.05.
LSD2 promotes proliferation by inducing G1-S arrest and reducing apoptosis in colorectal cancer (CRC) cells. (A) HCT-116 and LoVo cells stably expressing vector or sh-LSD2#1 or sh- LSD2#2. The percentage of cells in G0/G1, S. or G2/M phases was tested using a subG1 assay and flow cytometry. (B) Apoptotic rates of HCT-116 and LoVo cells stably expressing vector or sh-LSD2#1 or sh-LSD2#2 measured by flow cytometry in annexin V/PI assays. LR, early apoptotic cells; UR, terminal apoptotic cells. Values represent the means ± SD. (C) Quantitative analysis of DNA apoptosis. Data were expressed as means ± SEM. **p < 0.01. (D) Bcl-2, BAX, CL-caspase 3, and CL-caspase 9 protein expression measured by Western blot with GAPDH protein as the loading control.
LSD2 inhibits p53 expression via demethylation of H3K4me2 through binding to the p53 promoter. (A, B) Real-time PCR assays to detect p53 mRNA expression for vector, LSD2, ShNC, and Sh-LSD2 in HCT-116 and LoVo cells. (C, D) p53 and H3K4me2 protein expression measured by Western blot for vector, LSD2, and ShNC in HCT-116 and LoVo cells. (E, F) P53 promoter activity in vector, LSD2, ShNC and Sh-LSD2 in HCT-116 and LoVo cells measured by luciferase assay. Data were expressed as means ± SEM. (G) ChIP assay in LoVo cells showing the interaction between LSD2 and H3K4me2 at the p53 promoter. **p < 0.01.
LSD2 regulates colorectal cancer (CRC) cell proliferation and apoptosis through p53. ShRNA mediates p53 knockdown in HCT-116 LSD2-downregulated cell lines, while p53 over-expression in HCT-116 LSD2-upregulated cell lines was observed. (A) Protein expression of p53, p21, pRb and LSD2 measured by Western blot among the CRC cell lines mentioned above. (B) BrdU/PI assays were performed to measure DNA synthesis in the indicated CRC cell lines. (C) Quantitative analysis of DNA synthesis. (D) Western blot showing p53 and LSD2 protein expression in the p53 LSD2 double-downregulated HCT-116 and LoVo cell lines. (E) Flow cytometry detection of the apoptotic rates of the indicated CRC cell lines. (F) Quantitative analysis of DNA apoptosis. Data were expressed as means ± SEM. **p < 0.01.
-
Research Paper Volume 12, Issue 16 pp 16270-16293
CTCF promotes colorectal cancer cell proliferation and chemotherapy resistance to 5-FU via the P53-Hedgehog axis
Relevance score: 4.7651396Qiuhua Lai, Qingyuan Li, Chengcheng He, Yuxin Fang, Simin Lin, Jianqun Cai, Jian Ding, Qian Zhong, Yue Zhang, Changjie Wu, Xinke Wang, Juan He, Yongfeng Liu, Qun Yan, Aimin Li, Side Liu
Keywords: colorectal cancer, CTCF, Hedgehog, P53, chemotherapy resistance
Published in Aging on July 20, 2020
CTCF is significantly upregulated in CRC tissues and acts as a potential oncogenic gene. (A) CTCF is upregulated in all gastrointestinal tumors (GEPIA,
http://gepia.cancer-pku.cn/ ). (B) Kaplan-Meier relapse free survival analysis in CRC patients with high or low expression of CTCF in GSE31598 via online website PROGgeneV2 (http://genomics.jefferson.edu/proggene/ ). (C) GSEA indicated that high expression of CTCF was positively correlated with the cancer related gene set signatures (KEGG_PATHWAYS_IN_CANCER, KEGG_COLORECTAL_CANCER) in CRC patient gene expression profiles (GSE17538, n = 177, and TCGA, n = 465). (D) qRT-PCR analysis of CTCF expression in 77 pairs of CRC patient specimens. (E) Western blot analyses of CTCF in 12 pairs of tumor and match adjacent normal tissues collected from clinical CRC patients. N for Normal, T for Tumor. The above data are presented as mean ± SEM. * P<0.05, **P<0.01, and ***P<0.001.Upregulation of CTCF promotes human CRC cells proliferation. (A) GSEA plot indicated that high expression of CTCF is positively correlated with the cell cycle gene set signatures (KEGG_CELL_CYCLE) in published CRC patient gene expression profiles (GSE17538, n = 177, and TCGA, n = 465). (B, C) qRT-PCR and western blot analyses of CTCF expression level in constructed cell lines (HCT116 and RKO).(D, E) The relative growth rates were measured using CCK8 and colony formation assays and compared between CTCF overexpressed group and Vector group at indicated times in HCT116 and RKO cell lines. (F) Images of EdU staining in both indicated cell lines, and the relative percentage of EdU-positive cells in images of related groups are shown (G). (H, I) Tumor volume and weight were measured and analyzed. (J) The tumor sections were under IHC staining using antibody against Ki-67. The above data are presented as mean ± SEM. * P<0.05, **P<0.01, and ***P<0.001.
Inhibition of CTCF represses human CRC cells proliferation. (A) GSEA results showed that “KEGG_DNA_REPLICATION” gene set enriches in the CTCF high expression group and “CYCLIN_D1_KE_V1_UP” gene set enriches in the CTCF low expression group (GSE17538, n = 177, and TCGA, n = 465). (B, C) qRT-PCR and western blot analyses of CTCF expression level in the constructed cell lines (SW480 and RKO). (D–G) Cell reproductive capacity was examined by CCK8, colony formation and EdU staining assays. (H, I) Tumor volume and weight of subcutaneous tumor were measured and analysed. (J) Immunohistochemistry was performed to determine Ki-67 expression. The above data are presented as mean ± SEM. * P<0.05, **P<0.01, and ***P<0.001.
CTCF induces 5-FU-based chemoresistance in CRC. (A, B) Cell growth inhibition rate was measured via CCK8 analysis and compared between different groups with different treatment concentration at indicated time (48 hours). (C) The apoptosis rate of different transfected groups with 5-FU treatment were measured by flow cytometry. (D) Western blot analysis of CTCF in HCT116 and RKO cell lines after 5-FU (10 μM) treatment for 48 hours. (E, F) Western blot analyses of cleaved-PARP, cleaved Capase-3 and ABCG2 in HCT116 and SW480 cell lines. (G) Spearman correlation analyses between relative CTCF and ABCG2 mRNA expression in 13 fresh human CRC specimens. The above data are presented as mean ± SEM. * P<0.05, **P<0.01, and ***P<0.001.
CTCF activates Hedgehog signaling pathway. (A) GSEA plots indicated that “GCNP_SHH_UP_EARLY.V1_UP” and “GCNP_SHH_UP_LATE.V1_UP” and “GCNP_GLI1_UP.V1_UP” gene signatures enrich in the CTCF high expression group and “GCNP_SHH_UP_LATE.V1_DN” gene set enriches in the CTCF low expression group (GSE17538, n = 177, and TCGA, n = 465). (B) Western blot analyses of Key molecules of Hedgehog signaling pathway in different transfected groups with or without the stimulation of Hedgehog signaling pathway inhibitor, GDC-0449 (2 μM). (C) Relative growth rate of different transfected groups with or without the administration of GDC-0449 (2 μM). (D–F) Images of EdU staining of both indicated cell lines with or without the administration of GDC-0449, and the relative percentage of EdU-positive cells in images of related groups is shown. The above data are presented as mean ± SEM. * P<0.05, **P<0.01, and ***P<0.001.
CTCF enhances Hedgehog signaling pathway activation via targeting P53. (A) Venn diagram enrichment analysis of the 5-FU putative target genes. (B) Protein-protein interaction analysis via STRING (
https://string-db.org/ ). (C) GSEA plots indicated that “P53_DN.V1_UP” gene set signature enriches in the CTCF high expression group and “P53_DN.V1_DN” gene set enriches in the CTCF low expression group (GSE17538, n = 177). (D) Schematic view of the P53 gene transcription start site (TSS) with a CTCF-binding site (CBS). (E) qRT-PCR analysis of P53 expression level in constructed cell lines. (F) ChIP-PCR results for CTCF on the CBS in HCT116 cells. (G) P53 luciferase reporter activity was analyzed in SW480 cells. (H) Western blot analysis of Key molecules of Hedgehog signaling pathway and P53 in different transfected groups with or without the stimulation of P53-specific siRNA. (I, J) Nuclear extract assays and western blot analyses of GLI1 in indicated cells. The above data are presented as mean ± SEM. * P<0.05, **P<0.01, and ***P<0.001.CTCF promotes chemoresistance by regulating P53-Hedgehog axis signaling. (A) Apoptosis assays showed the effect of GDC0449 on CTCF-mediated 5-FU stimulated apoptosis of CRC cells. (B) Western blot analysis of the effects of P53 on CTCF-mediated ABCG2 upregulation. (C) The representative images of subcutaneous tumors from different experimental groups are shown. (D, E) Tumor weight and volume analyses showed that JK184 recovered the stimulative cell proliferation caused by upregulated CTCF under stimulation of 5-FU. (F) A hypothetical model illustrating that CTCF transcriptionally represses P53 and activates the Hedgehog signaling pathway to promote proliferation and 5-FU chemotherapy resistance of CRC cells. The above data are presented as mean ± SEM. * P<0.05, **P<0.01, and ***P<0.001.
-
Research Paper Volume 12, Issue 11 pp 10473-10487
p53 plays a central role in the development of osteoporosis
Relevance score: 5.0260425Tao Yu, Xiaomeng You, Haichao Zhou, Alex Kang, Wenbao He, Zihua Li, Bing Li, Jiang Xia, Hui Zhu, Youguang Zhao, Guangrong Yu, Yuan Xiong, Yunfeng Yang
Keywords: osteoporosis, osteogenesis, bone mass density, p53
Published in Aging on June 2, 2020
Heat map of the top 60 DEGs in GSE100609 (30 upregulated and 30 downregulated). The GSE100609 dataset, which included gene expression profiles from four healthy individuals and four osteoporotic patients, was obtained from the GEO database. In total, 228 upregulated and 281 downregulated DEGs were identified. Red, upregulation; blue, downregulation.
(A) Enrichment analysis of upregulated genes: hsa05200, cancer pathway; hsa05222, small cell lung cancer pathway; hsa04115, p53 signaling pathway; hsa04310, wnt signaling pathway; hsa04015, rap1 signaling pathway. (B) Enrichment analysis of downregulated genes: hsa04360, axon guidance pathway; hsa05100, bacterial invasion of epithelial cells pathway; hsa05143, African trypanosomiasis pathway; hsa05010, Alzheimer's disease pathway; hsa04020, calcium signaling pathway.
Protein-protein interactions. PPI network constructed using STRING and illustrating the interactions among DEGs.
Degree, betweenness, closeness of hub genes. Within the PPI network, the top three hub genes with highest degree, betweenness and closeness scores are TP53, MAPK1, and CASP3.
GO terms/KEGG pathways associated with top 10 enriched hub genes. Biological process module: GO:0070887, cellular response to chemical stimulus; GO:0031325, positive regulation of cellular metabolic process; GO:0010604, positive regulation of macromolecule metabolic process. Molecular functions module: GO:0019899, enzyme binding; GO:0005102, receptor binding; GO:0044877, macromolecular complex binding. KEGG pathway module: hsa05200, cancer pathway; hsa05205, proteoglycans in cancer pathway; hsa04115, p53 signaling pathway.
Expression of top 50 genes based on degree score from the 6 modules and their positions on chromosome. The hub genes TP53, MAPK1, CASP3, CTNNB1, CCND1, NOTCH1, CDK1, IGF1, ERBB2, and CYCS are highlighted in blue. TP53, MAPK1, and CASP3 are located on chromosomes 17, 22, and 4, respectively.
Downregulating p53 expression may protect against osteoporosis in vitro. (A, B) qRT-PCR and western blot analyses comparing p53 expression between healthy volunteers and osteoporosis patients. (C) Cellular morphology of hMSCs, scale bar, 50 μm. (D, E) hMSCs were treated with PBS, siRNA-NC, or siRNA-p53, after which p53 expression was assessed with qRT-PCR and western blotting. (F, G) qRT-PCR and western blot analysis of osteogenesis-related mRNAs in the three groups. Data are means±SD of triplicate experiments. *p < 0.05, **p < 0.01, ***p < 0.001.
Downregulating p53 expression may protect against osteoporosis in vivo. (A) Cross sections of bone samples from osteoporosis model mice treated for 10 weeks with PBS, siRNA-NC, or siRNA-p53 (n=10 in each group). (B) BV, TV, BV/TV, and BMD values measured in osteoporosis model mice in each treatment group. Data are means±SD of triplicate experiments. *p < 0.05, **p < 0.01, ***p < 0.001.