1
|
Song P, Duan J, Ding J, Liu J, Fang Z, Xu H, Li Z, Du W, Xu M, Ling Y, He F, Tao K, Wang L. Cellular senescence primes liver fibrosis regression through Notch-EZH2. MedComm (Beijing) 2023; 4:e346. [PMID: 37614965 PMCID: PMC10442476 DOI: 10.1002/mco2.346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/27/2023] [Accepted: 07/12/2023] [Indexed: 08/25/2023] Open
Abstract
Cellular senescence plays a pivotal role in wound healing. At the initiation of liver fibrosis regression, accumulated senescent cells were detected and genes of senescence were upregulated. Flow cytometry combined with single-cell RNA sequencing analyses revealed that most of senescent cells were liver nonparenchymal cells. Removing senescent cells by dasatinib and quercetin (DQ), alleviated hepatic cellular senescence, impeded fibrosis regression, and disrupted liver sinusoids. Clearance of senescent cells not only decreased senescent macrophages but also shrank the proportion of anti-inflammatory M2 macrophages through apoptotic pathway. Subsequently, macrophages were depleted by clodronate, which diminished hepatic senescent cells and impaired fibrosis regression. Mechanistically, the change of the epigenetic regulator enhancer of zeste homolog2 (EZH2) accompanied with the emergence of hepatic senescent cells while liver fibrosis regressed. Blocking EZH2 signaling by EPZ6438 reduced hepatic senescent cells and macrophages, decelerating liver fibrosis regression. Moreover, the promoter region of EZH2 was transcriptionally suppressed by Notch-Hes1 (hairy and enhancer of split 1) signaling. Disruption of Notch in macrophages using Lyz2 (lysozyme 2) Cre-RBP-J (recombination signal binding protein Jκ) f/f transgenic mice, enhanced hepatic cellular senescence, and facilitated fibrosis regression by upregulating EZH2 and blocking EZH2 abrogated the above effects caused by Notch deficiency. Ultimately, adopting Notch inhibitor Ly3039478 or exosome-mediated RBP-J decoy oligodeoxynucleotides accelerated liver fibrosis regression by augmenting hepatic cellular senescence.
Collapse
Affiliation(s)
- Ping Song
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Juan‐Li Duan
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Jian Ding
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Jing‐Jing Liu
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Zhi‐Qiang Fang
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Hao Xu
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Zhi‐Wen Li
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Wei Du
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Ming Xu
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Yu‐Wei Ling
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Fei He
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Kai‐Shan Tao
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| | - Lin Wang
- Department of Hepatobiliary SurgeryXi‐Jing HospitalFourth Military Medical UniversityXi'anChina
| |
Collapse
|
2
|
Shen J, Yang H, Qiao X, Chen Y, Zheng L, Lin J, Lang J, Yu Q, Wang Z. The E3 ubiquitin ligase TRIM17 promotes gastric cancer survival and progression via controlling BAX stability and antagonizing apoptosis. Cell Death Differ 2023; 30:2322-2335. [PMID: 37697039 PMCID: PMC10589321 DOI: 10.1038/s41418-023-01221-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 08/20/2023] [Accepted: 08/31/2023] [Indexed: 09/13/2023] Open
Abstract
Tripartite motif 17 (TRIM17) belongs to a subfamily of the RING-type E3 ubiquitin ligases, and regulates several cellular processes and pathological conditions including cancer. However, its potential function in gastric cancer (GC) remains obscure. Here, we have found TRIM17 mRNA and protein levels are both upregulated in human GC compared with normal specimens, and TRIM17 upregulation indicates poor survival for GC patients. Functionally, TRIM17 was found to act as an oncogene by promoting the proliferation and survival of GC cell lines AGS and HGC-27. Mechanistically, TRIM17 acts to interact with BAX and promote its ubiquitination and proteasomal degradation, leading to a deficiency in BAX-dependent apoptosis in GC cells in the absence and presence of apoptosis stimuli. Moreover, TRIM17 and BAX expression levels are inversely correlated in human GC specimens. Our data thus suggest TRIM17 contributes to gastric cancer survival through regulating BAX protein stability and antagonizing apoptosis, which provides a promising therapeutic target for GC treatment and a biomarker for prognosis.
Collapse
Affiliation(s)
- Jiajia Shen
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hang Yang
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xinran Qiao
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Chen
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Liyun Zheng
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyu Lin
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyu Lang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Yu
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Zhen Wang
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
3
|
Qiao X, Lin J, Shen J, Chen Y, Zheng L, Ren H, Zhao X, Yang H, Li P, Wang Z. FBXO28 suppresses liver cancer invasion and metastasis by promoting PKA-dependent SNAI2 degradation. Oncogene 2023; 42:2878-2891. [PMID: 37596321 PMCID: PMC10516749 DOI: 10.1038/s41388-023-02809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/20/2023]
Abstract
FBXO28 is a member of F-box proteins that are the substrate receptors of SCF (SKP1, CULLIN1, F-box protein) ubiquitin ligase complexes. Despite the implications of its role in cancer, the function of FBXO28 in epithelial-mesenchymal transition (EMT) process and metastasis for cancer remains largely unknown. Here, we report that FBXO28 is a critical negative regulator of migration, invasion and metastasis in human hepatocellular carcinoma (HCC) in vitro and in vivo. FBXO28 expression is upregulated in human epithelial cancer cell lines relative to mesenchymal counterparts. Mechanistically, by directly binding to SNAI2, FBXO28 functions as an E3 ubiquitin ligase that targets the substrate for degradation via ubiquitin proteasome system. Importantly, we establish a cooperative function for PKA in FBXO28-mediated SNAI2 degradation. In clinical HCC specimens, FBXO28 protein levels positively whereas negatively correlate with PKAα and SNAI2 levels, respectively. Low FBXO28 or PRKACA expression is associated with poor prognosis of HCC patients. Together, these findings elucidate the novel function of FBXO28 as a critical inhibitor of EMT and metastasis in cancer and provide a mechanistic rationale for its candidacy as a new prognostic marker and/or therapeutic target in human aggressive HCC.
Collapse
Affiliation(s)
- Xinran Qiao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingyu Lin
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiajia Shen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Chen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyun Zheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hangjiang Ren
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoli Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hang Yang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Pengyu Li
- Qilu Hospital of Shan Dong University, Jinan, Shandong Province, China
| | - Zhen Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
4
|
Kong SH, Ma L, Yuan Q, Liu X, Han Y, Xiang W, Liu DX, Zhang Y, Lu J. Inhibition of EZH2 alleviates SAHA-induced senescence-associated secretion phenotype in small cell lung cancer cells. Cell Death Discov 2023; 9:289. [PMID: 37543653 PMCID: PMC10404275 DOI: 10.1038/s41420-023-01591-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/10/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023] Open
Abstract
Chemotherapy has been widely used in small cell lung cancer (SCLC) treatment in the past decades. However, SCLC is easy to recur after chemotherapy. The senescence of cancer cells during chemotherapy is one of the effective therapeutic strategies to inhibit the progression of cancer. Nevertheless, the senescence-associated secretion phenotype (SASP) promotes chronic inflammation of the cancer microenvironment and further accelerates the progression of tumors. Therefore, inducing the senescence of cancer cells and inhibiting the production of SASP factors during anticancer treatment have become effective therapeutic strategies to improve the anticancer effect of drugs. Here we reported that SCLC cells treated with an FDA-approved HDAC inhibitor SAHA underwent senescence and displayed remarkable SASP. In particular, SAHA promoted the formation of cytoplasmic chromatin fragments (CCFs) in SCLC cells. The increased CCFs in SAHA-treated SCLC cells were related to nuclear porin Tpr, which activated the cGAS-STING pathway, and promoted the secretion of SASP in cancer cells. Inhibition of EZH2 suppressed the increase of CCFs in SAHA-treated SCLC cells, weakened the production of SASP, and increased the antiproliferative effect of SAHA. Overall, our work affords new insight into the secretion of SASP in SCLC and establishes a foundation for constructing a new therapeutic strategy for SCLC patients.
Collapse
Affiliation(s)
- Sun-Hyok Kong
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
- School of Life Science, University of Science, Pyongyang, 999091, Democratic People's Republic of Korea
| | - Lie Ma
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, 130024, China
| | - Qingxia Yuan
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
| | - Xiangxiang Liu
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, 130024, China
| | - Yu Han
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
| | - Weifang Xiang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
| | - Dong-Xu Liu
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, 1010, New Zealand
| | - Yu Zhang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China.
| | - Jun Lu
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, 130024, China.
| |
Collapse
|
5
|
Kim MK, Shin HS, Shin MH, Kim H, Lee DH, Chung JH. Dual role of enhancer of zeste homolog 2 in the regulation of ultraviolet radiation-induced matrix metalloproteinase-1 and type I procollagen expression in human dermal fibroblasts. Matrix Biol 2023; 119:112-124. [PMID: 37031807 DOI: 10.1016/j.matbio.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/13/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Abnormalities in the extracellular matrix (ECM) caused by ultraviolet (UV) radiation are mediated by epigenetic mechanisms. Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that is implicated in inflammation, immune regulation, and senescence. However, its role in controlling UV-induced ECM alterations in the skin remains elusive. Here, we investigated the role of EZH2 in UV-induced expression of matrix metalloproteinase (MMP)-1 and type I procollagen. We found that UV induced EZH2 expression in human skin in vivo and in human dermal fibroblasts (HDFs). EZH2 knockdown reduced the expression and promoter activity of MMP-1 and increased those of type I procollagen, whereas EZH2 overexpression had the opposite effects. Mechanistically, EZH2 increased NF-κB activity, and p65 and p50 expression and promoter activity. Intriguingly, chromatin immunoprecipitation assays revealed that the EZH2/p65/p50 complex was recruited and bound to the MMP-1 promoter after UV irradiation, independent of its histone methyltransferase activity. In contrast, EZH2-induced DNA methyltransferase 1 (DNMT1) formed a complex with EZH2 and enhanced the enrichment of H3K27me3 on the COL1A2 promoter following UV irradiation. These findings indicate that EZH2 plays a dual role in regulating MMP-1 and type I procollagen expression and improve our understanding of how this epigenetic mechanism contributes to UV-induced skin responses and photoaging. This study shows that inhibiting EZH2 is a potential anti-aging strategy for preventing UV-induced skin aging by reducing MMP-1 expression and inducing type I procollagen expression.
Collapse
Affiliation(s)
- Min-Kyoung Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hye Sun Shin
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea; Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Mi Hee Shin
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Haesoo Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea; Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea; Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea; Institute on Aging, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Yang Y, Liu K, Liu M, Zhang H, Guo M. EZH2: Its regulation and roles in immune disturbance of SLE. Front Pharmacol 2022; 13:1002741. [DOI: 10.3389/fphar.2022.1002741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is related to immune homeostasis imbalance. Epigenetic mechanisms have played a significant role in breaking immune tolerance. Enhancer of zeste homolog 2 (EZH2), the specific methylation transferase of lysine at position 27 of histone 3, is currently found to participate in the pathogenesis of SLE through affecting multiple components of the immune system. This review mainly expounds the mechanisms underlying EZH2-mediated disruption of immune homeostasis in SLE patients, hoping to provide new ideas in the pathogenesis of SLE and new targets for future treatment.
Collapse
|
7
|
Ji Y, Xu X, Long C, Wang J, Ding L, Zheng Z, Wu H, Yang L, Tao L, Gao F. SMYD2 aggravates gastrointestinal stromal tumor via upregulation of EZH2 and downregulation of TET1. Cell Death Dis 2022; 8:274. [PMID: 35668081 PMCID: PMC9170715 DOI: 10.1038/s41420-022-01038-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022]
Abstract
SMYD2, as an oncogene, has been involved in multiple types of cancer, but the potential role of SMYD2 in gastrointestinal stromal tumors (GIST) remains enigmatic and requires further investigation. Hence, this study was conducted with the main objective of analyzing the effect of SMYD2 on GIST. GIST and adjacent normal tissues were collected from 46 patients with GIST where the expression of EZH2, SMYD2, and TET1 was determined, followed by the analysis of their interactions. The functional role of SMYD2 in cell biological functions was determined using a loss-of-function assay in GIST-T1 cells. Nude mouse xenograft experiments were performed to verify the role of the SMYD2/EZH2/TET1 axis in GIST in vivo. EZH2 was upregulated in GIST tissues and cell lines, which was positively correlated with SMYD2 expression and inversely correlated with TET1 expression in GIST tissues. EZH2 silencing due to SMYD2 inhibition reduced GIST-T1 cell proliferation and accelerated cell senescence. EZH2 repressed TET1 expression by promoting H3K27me3 methylation in the TET1 promoter region. TET1 inhibition reversed the effect of EZH2 silencing on the biological functions of GIST-T1 cells. In vivo data further revealed the promoting effect of SMYD2 on the progression of GIST by regulating the EZH2/TET1 axis. Overall, this study demonstrates that SMYD2 can increase EZH2 expression while suppressing TET1 expression, thus accelerating GIST, and creating new treatment opportunities for GIST.
Collapse
Affiliation(s)
- Yong Ji
- Department of General Gastrointestinal Surgery, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Xiaofeng Xu
- Department of Clinical Laboratory, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Cong Long
- Department of Clinical Laboratory, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Jianjiang Wang
- Department of General Surgery, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Li Ding
- Department of Clinical Laboratory, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Zhizhong Zheng
- Department of Clinical Laboratory, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Huiping Wu
- Department of Science and Education, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Liu Yang
- Department of Clinical Laboratory, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Lan Tao
- Central Laboratory, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China
| | - Feng Gao
- Department of General Surgery, Jingjiang People's Hospital, 214500, Jingjiang, P.R. China.
| |
Collapse
|
8
|
Li H, Zhao X, Zheng L, Wang X, Lin S, Shen J, Ren H, Li Y, Qiu Q, Wang Z. Bruceine A protects against diabetic kidney disease via inhibiting galectin-1. Kidney Int 2022; 102:521-535. [DOI: 10.1016/j.kint.2022.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 03/13/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
|
9
|
Wang R, Sun L, Xia S, Wu H, Ma Y, Zhan S, Zhang G, Zhang X, Shi T, Chen W. B7-H3 suppresses doxorubicin-induced senescence-like growth arrest in colorectal cancer through the AKT/TM4SF1/SIRT1 pathway. Cell Death Dis 2021; 12:453. [PMID: 33958586 PMCID: PMC8102521 DOI: 10.1038/s41419-021-03736-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022]
Abstract
Emerging evidence suggests that cellular senescence induced by chemotherapy has been recognized as a new weapon for cancer therapy. This study aimed to research novel functions of B7-H3 in cellular senescence induced by a low dose of doxorubicin (DOX) in colorectal cancer (CRC). Here, our results demonstrated that B7-H3 knockdown promoted, while B7-H3 overexpression inhibited, DOX-induced cellular senescence. B7-H3 knockdown dramatically enhanced the growth arrest of CRC cells after low-dose DOX treatment, but B7-H3 overexpression had the opposite effect. By RNA-seq analysis and western blot, we showed that B7-H3 prevented cellular senescence and growth arrest through the AKT/TM4SF1/SIRT1 pathway. Blocking the AKT/TM4SF1/SIRT1 pathway dramatically reversed B7-H3-induced resistance to cellular senescence. More importantly, B7-H3 inhibited DOX-induced cellular senescence of CRC cells in vivo. Therefore, targeting B7-H3 or the B7-H3/AKT/TM4SF1/SIRT1 pathway might be a new strategy for promoting cellular senescence-like growth arrest during drug treatment in CRC.
Collapse
Affiliation(s)
- Ruoqin Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China
- Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Linqing Sun
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Suhua Xia
- Department of Oncology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Hongya Wu
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Yanchao Ma
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Shenghua Zhan
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China.
- Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China.
| | - Weichang Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China.
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China.
- Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China.
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China.
| |
Collapse
|
10
|
Jiang Y, Xiang C, Zhong F, Zhang Y, Wang L, Zhao Y, Wang J, Ding C, Jin L, He F, Wang H. Histone H3K27 methyltransferase EZH2 and demethylase JMJD3 regulate hepatic stellate cells activation and liver fibrosis. Am J Cancer Res 2021; 11:361-378. [PMID: 33391480 PMCID: PMC7681085 DOI: 10.7150/thno.46360] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: As the central hallmark of liver fibrosis, transdifferentiation of hepatic stellate cells (HSCs), the predominant contributor to fibrogenic hepatic myofibroblast responsible for extracellular matrix (ECM) deposition, is characterized with transcriptional and epigenetic remodeling. We aimed to characterize the roles of H3K27 methyltransferase EZH2 and demethylase JMJD3 and identify their effective pathways and novel target genes in HSCs activation and liver fibrosis. Methods: In primary HSCs, we analyzed effects of pharmacological inhibitions and genetic manipulations of EZH2 and JMJD3 on HSCs activation. In HSCs cell lines, we evaluated effects of EZH2 inhibition by DZNep on proliferation, cell cycling, senescence and apoptosis. In CCl4 and BDL murine models of liver fibrosis, we assessed in vivo effects of DZNep administration and Ezh2 silencing. We profiled rat primary HSCs transcriptomes with RNA-seq, screened the pathways and genes associated with DZNep treatment, analyzed EZH2 and JMJD3 regulation towards target genes by ChIP-qPCR. Results: EZH2 inhibition by DZNep resulted in retarded growth, lowered cell viability, cell cycle arrest in S and G2 phases, strengthened senescence, and enhanced apoptosis of HSCs, decreased hepatic collagen deposition and rescued the elevated serum ALT and AST activities of diseased mice, and downregulated cellular and hepatic expressions of H3K27me3, EZH2, α-SMA and COL1A. Ezh2 silencing by RNA interference in vitro and in vivo showed similar effects. JMJD3 inhibition by GSK-J4 and overexpression of wild-type but not mutant Jmjd3 enhanced or repressed HSCs activation respectively. EZH2 inhibition by DZNep transcriptionally inactivated TGF-β1 pathway, cell cycle pathways and vast ECM components in primary HSCs. EZH2 inhibition decreased H3K27me3 recruitment at target genes encoding TGF-β1 pseudoreceptor BAMBI, anti-inflammatory cytokine IL10 and cell cycle regulators CDKN1A, GADD45A and GADD45B, and increased their expressions, while Jmjd3 overexpression manifested alike effects. Conclusions: EZH2 and JMJD3 antagonistically modulate HSCs activation. The therapeutic effects of DZNep as epigenetic drug in liver fibrosis are associated with the regulation of EZH2 towards direct target genes encoding TGF-β1 pseudoreceptor BAMBI, anti-inflammatory cytokine IL10 and cell cycle regulators CDKN1A, GADD45A and GADD45B, which are also regulated by JMJD3. Our present study provides new mechanistic insight into the epigenetic modulation of EZH2 and JMJD3 in HSCs biology and hepatic fibrogenesis.
Collapse
|
11
|
The role of histone methylation in the development of digestive cancers: a potential direction for cancer management. Signal Transduct Target Ther 2020; 5:143. [PMID: 32747629 PMCID: PMC7398912 DOI: 10.1038/s41392-020-00252-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/22/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023] Open
Abstract
Digestive cancers are the leading cause of cancer-related death worldwide and have high risks of morbidity and mortality. Histone methylation, which is mediated mainly by lysine methyltransferases, lysine demethylases, and protein arginine methyltransferases, has emerged as an essential mechanism regulating pathological processes in digestive cancers. Under certain conditions, aberrant expression of these modifiers leads to abnormal histone methylation or demethylation in the corresponding cancer-related genes, which contributes to different processes and phenotypes, such as carcinogenesis, proliferation, metabolic reprogramming, epithelial–mesenchymal transition, invasion, and migration, during digestive cancer development. In this review, we focus on the association between histone methylation regulation and the development of digestive cancers, including gastric cancer, liver cancer, pancreatic cancer, and colorectal cancer, as well as on its clinical application prospects, aiming to provide a new perspective on the management of digestive cancers.
Collapse
|
12
|
Wu J, Li L, Zhang Y, Zhu J. Decreased miR-124 contributes to the epithelial-mesenchymal transition phenotype formation of lung adenocarcinoma cells via targeting enhancer of zeste homolog 2. Pathol Res Pract 2020; 216:152976. [PMID: 32370988 DOI: 10.1016/j.prp.2020.152976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/18/2020] [Accepted: 04/13/2020] [Indexed: 01/09/2023]
Abstract
INTRODUCTION MiR-124, a tumor suppressor, is involved in regulating various cellular processes. The purpose of this study was to investigate the possible function of miR-124 in LA (lung adenocarcinoma) cells. AIMS MiR-124 expression levels in the 54 pairs of LA tissues (and corresponding non-tumor tissues) obtained at the Sixth People's Hospital of Yancheng City and in LA cells were assessed by qRT-PCR. Colony formation assay, wound healing assay, transwell assays, attachment/detachment, western blotting and immunofluorescence assays were performed to assess the function of miR-124 on proliferation, migration and epithelial-to-mesenchymal (EMT) phenotypes in LA cells in vitro. Enhancer of zeste homolog 2 (EZH2) is identified as a target of miR-124 by bioinformatics analysis and luciferase reporter assays. Rescue assays were applied to verify the relationship between miR-124 and EZH2. RESULTS MiR-124 was down-regulated in LA tissues (compared to adjacent non-tumor tissues), and was down-regulated in 3 out of 4 lung cancer cell lines compared to immortalized, non-tumorigenic bronchial epithelial cells. Forced expression of miR-124 significantly suppressed tumor cell proliferation, migration and inhibited the EMT process. On the contrary, deletion of miR-124 could obviously promote cell proliferation, migration and facilitate the formation of EMT phenotype. Bioinformatics analysis and luciferase reporter assays confirmed that EZH2 was a target gene of miR-124 and was negatively correlated with the level of miR-124 in cancer tissues. CONCLUSION Our current study suggested that miR-124 was a tumor suppressor in LA, and miR-124 was associated with LA cell EMT phenotype formation via targeting EZH2.
Collapse
Affiliation(s)
- Jian Wu
- Department of Medical Oncology, The Sixth People's Hospital of Yancheng City, No. 66 Tingzhong Road, Yancheng, Jiangsu, 224000, China
| | - Lingling Li
- Department of Medical Oncology, The Sixth People's Hospital of Yancheng City, No. 66 Tingzhong Road, Yancheng, Jiangsu, 224000, China
| | - Yi Zhang
- Department of Oncology, Jimin Hospital, Shanghai, 200052, China
| | - Jianjun Zhu
- Department of Medical Oncology, The Sixth People's Hospital of Yancheng City, No. 66 Tingzhong Road, Yancheng, Jiangsu, 224000, China.
| |
Collapse
|
13
|
Li Z, Chen Y, Tang M, Li Y, Zhu WG. Regulation of DNA damage-induced ATM activation by histone modifications. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s42764-019-00004-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
14
|
Wang R, Li L, Duan A, Li Y, Liu X, Miao Q, Gong J, Zhen Y. Crizotinib enhances anti-CD30-LDM induced antitumor efficacy in NPM-ALK positive anaplastic large cell lymphoma. Cancer Lett 2019; 448:84-93. [PMID: 30742941 DOI: 10.1016/j.canlet.2019.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/21/2022]
Abstract
Combining antibody-drug conjugates (ADCs) with targeted small-molecule inhibitors can enhance antitumor effects beyond those attainable with monotherapy. In this study, we investigated the therapeutic combination of a CD30-targeting ADC (anti-CD30-lidamycin [LDM]) with a small-molecule inhibitor (crizotinib) of nucleophosmin-anaplastic lymphoma kinase NPM-ALK in CD30+/ALK+ anaplastic large cell lymphoma (ALCL). In vitro, anti-CD30-LDM showed strong synergistic antiproliferative activity when combined with crizotinib. Furthermore, treatment with anti-CD30-LDM plus crizotinib resulted in a stronger induction of cell apoptosis than monotherapy with either treatment. Western blot analysis revealed that ERK1/2 phosphorylation was increased in response to anti-CD30-LDM-induced DNA damage. Interestingly, the addition of crizotinib inhibited the expression of phosphorylated ERK1/2 and further augmented anti-CD30-LDM-mediated apoptosis, providing a potential synergistic mechanism for DNA-damaging agents combined with NPM-ALK inhibitors. In Karpas299 and SU-DHL-1 xenograft models, anti-CD30-LDM plus crizotinib was more effective in inhibiting tumor growth than either treatment alone. This research demonstrated for the first time that the combination of anti-CD30-LDM and crizotinib exhibits a synergistic inhibitory effect in tumor cells. These results provide scientific support for future clinical evaluations of anti-CD30-LDM, or other DNA-damaging agents, combined with NPM-ALK inhibitors.
Collapse
Affiliation(s)
- Rong Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liang Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Aijun Duan
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yi Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiujun Liu
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qingfang Miao
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Jianhua Gong
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yongsu Zhen
- NHC Key Laboratory of Biotechnology of Antibiotics, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Sane S, Hafner A, Srinivasan R, Masood D, Slunecka JL, Noldner CJ, Hanson AD, Kruisselbrink T, Wang X, Wang Y, Yin J, Rezvani K. UBXN2A enhances CHIP-mediated proteasomal degradation of oncoprotein mortalin-2 in cancer cells. Mol Oncol 2018; 12:1753-1777. [PMID: 30107089 PMCID: PMC6166003 DOI: 10.1002/1878-0261.12372] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 07/12/2018] [Accepted: 08/02/2018] [Indexed: 01/07/2023] Open
Abstract
Overexpression of oncoproteins is a major cause of treatment failure using current chemotherapeutic drugs. Drug-induced degradation of oncoproteins is feasible and can improve clinical outcomes in diverse types of cancers. Mortalin-2 (mot-2) is a dominant oncoprotein in several tumors, including colorectal cancer (CRC). In addition to inactivating the p53 tumor suppressor protein, mot-2 enhances tumor cell invasion and migration. Thus, mot-2 is considered a potential therapeutic target in several cancer types. The current study investigated the biological role of a ubiquitin-like protein called UBXN2A in the regulation of mot-2 turnover. An orthogonal ubiquitin transfer technology followed by immunoprecipitation, in vitro ubiquitination, and Magnetic Beads TUBE2 pull-down experiments revealed that UBXN2A promotes carboxyl terminus of the HSP70-interacting protein (CHIP)-dependent ubiquitination of mot-2. We subsequently showed that UBXN2A increases proteasomal degradation of mot-2. A subcellular compartmentalization experiment revealed that induced UBXN2A decreases the level of mot-2 and its chaperone partner, HSP60. Pharmacological upregulation of UBXN2A using a small molecule, veratridine (VTD), decreases the level of mot-2 in cancer cells. Consistent with the in vitro results, UBXN2A+/- mice exhibited selective elevation of mot-2 in colon tissues. An in vitro Anti-K48 TUBE isolation approach showed that recombinant UBXN2A enhances proteasomal degradation of mot-2 in mouse colon tissues. Finally, we observed enhanced association of CHIP with the UBXN2A-mot-2 complex in tumors in an azoxymethane/dextran sulfate sodium-induced mouse CRC model. The existence of a multiprotein complex containing UBXN2A, CHIP, and mot-2 suggests a synergistic tumor suppressor activity of UBXN2A and CHIP in mot-2-enriched tumors. This finding validates the UBXN2A-CHIP axis as a novel and potential therapeutic target in CRC.
Collapse
Affiliation(s)
- Sanam Sane
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - Andre Hafner
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - Rekha Srinivasan
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - Daniall Masood
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - John l. Slunecka
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - Collin J. Noldner
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - Alex D. Hanson
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - Taylor Kruisselbrink
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - Xuejun Wang
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| | - Yiyang Wang
- Department of ChemistryCenter for Diagnostics & TherapeuticsGeorgia State UniversityAtlantaGAUSA
| | - Jun Yin
- Department of ChemistryCenter for Diagnostics & TherapeuticsGeorgia State UniversityAtlantaGAUSA
| | - Khosrow Rezvani
- Division of Basic Biomedical SciencesSanford School of MedicineThe University of South DakotaVermillionSDUSA
| |
Collapse
|
16
|
Cao J, Pontes KCS, Heijkants RC, Brouwer NJ, Groenewoud A, Jordanova ES, Marinkovic M, van Duinen S, Teunisse AFAS, Verdijk RM, Snaar‐Jagalska E, Jochemsen AG, Jager MJ. Overexpression of EZH2 in conjunctival melanoma offers a new therapeutic target. J Pathol 2018; 245:433-444. [PMID: 29732557 PMCID: PMC6174981 DOI: 10.1002/path.5094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 03/23/2018] [Accepted: 04/27/2018] [Indexed: 12/22/2022]
Abstract
Malignant melanoma of the conjunctiva (CM) is an uncommon but potentially deadly disorder. Many malignancies show an increased activity of the epigenetic modifier enhancer of zeste homolog 2 (EZH2). We studied whether EZH2 is expressed in CM, and whether it may be a target for therapy in this malignancy. Immunohistochemical analysis showed that EZH2 protein expression was absent in normal conjunctival melanocytes and primary acquired melanosis, while EZH2 was highly expressed in 13 (50%) of 26 primary CM and seven (88%) of eight lymph node metastases. Increased expression was positively associated with tumour thickness (p =0.03). Next, we targeted EZH2 with specific inhibitors (GSK503 and UNC1999) or depleted EZH2 by stable shRNA knockdown in three primary CM cell lines. Both pharmacological and genetic inactivation of EZH2 inhibited cell growth and colony formation and influenced EZH2-mediated gene transcription and cell cycle profile in vitro. The tumour suppressor gene p21/CDKN1A was especially upregulated in CM cells after EZH2 knockdown in CM cells. Additionally, the potency of GSK503 against CM cells was monitored in zebrafish xenografts. GSK503 profoundly attenuated tumour growth in CM xenografts at a well-tolerated concentration. Our results indicate that elevated levels of EZH2 are relevant to CM tumourigenesis and progression, and that EZH2 may become a potential therapeutic target for patients with CM. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jinfeng Cao
- Department of OphthalmologyLeiden University Medical CentreLeidenThe Netherlands
- Department of OphthalmologyThe Second Hospital of Jilin UniversityChangchunPR China
| | - Kelly CS Pontes
- Department of OphthalmologyLeiden University Medical CentreLeidenThe Netherlands
- Department of Molecular Cell Biology, Institute of BiologyLeiden UniversityLeidenThe Netherlands
| | - Renier C Heijkants
- Department of Molecular Cell BiologyLeiden University Medical CentreLeidenThe Netherlands
| | - Niels J Brouwer
- Department of OphthalmologyLeiden University Medical CentreLeidenThe Netherlands
| | - Arwin Groenewoud
- Department of Molecular Cell Biology, Institute of BiologyLeiden UniversityLeidenThe Netherlands
| | - Ekaterina S Jordanova
- Department of PathologyLeiden University Medical CentreLeidenThe Netherlands
- Centre for Gynaecological Oncology Amsterdam (CGOA)VU University Medical CentreAmsterdamThe Netherlands
| | - Marina Marinkovic
- Department of OphthalmologyLeiden University Medical CentreLeidenThe Netherlands
| | - Sjoerd van Duinen
- Department of PathologyLeiden University Medical CentreLeidenThe Netherlands
| | - Amina FAS Teunisse
- Department of Molecular Cell BiologyLeiden University Medical CentreLeidenThe Netherlands
| | - Robert M Verdijk
- Department of Pathology, Section Ophthalmic PathologyErasmus MC University Medical CentreRotterdamThe Netherlands
| | - Ewa Snaar‐Jagalska
- Department of Molecular Cell Biology, Institute of BiologyLeiden UniversityLeidenThe Netherlands
| | - Aart G Jochemsen
- Department of Molecular Cell BiologyLeiden University Medical CentreLeidenThe Netherlands
| | - Martine J Jager
- Department of OphthalmologyLeiden University Medical CentreLeidenThe Netherlands
| |
Collapse
|
17
|
Liu R, Ma Y, Hu X, Liao Y, Hu X, He B, Sun W. Pioglitazone/metformin adduct regulates insulin secretion and inhibits high glucose‐induced apoptosis via p21‐p53‐MDM2 signaling in INS‐1 cells. J Cell Biochem 2018; 119:5449-5459. [DOI: 10.1002/jcb.26701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/23/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Rong‐Xing Liu
- Department of PharmacologySchool of PharmacyChongqing Medical UniversityChongqingPeople's Republic of China
- Key Laboratory for Biochemistry and Molecular Pharmacology of ChongqingChongqing Medical UniversityChongqingPeople's Republic of China
| | - Yan Ma
- Department of PharmacologySchool of PharmacyChongqing Medical UniversityChongqingPeople's Republic of China
- Key Laboratory for Biochemistry and Molecular Pharmacology of ChongqingChongqing Medical UniversityChongqingPeople's Republic of China
| | - Xue‐Lian Hu
- Department of PharmacochemistrySchool of PharmacyChongqing Medical UniversityChongqingPeople's Republic of China
- Department of PharmacologyXinqiao HospitalChongqingPeople's Republic of China
| | - Yun‐Peng Liao
- Department of PharmacologySchool of PharmacyChongqing Medical UniversityChongqingPeople's Republic of China
- Key Laboratory for Biochemistry and Molecular Pharmacology of ChongqingChongqing Medical UniversityChongqingPeople's Republic of China
| | - Xiang‐Nan Hu
- Department of PharmacochemistrySchool of PharmacyChongqing Medical UniversityChongqingPeople's Republic of China
| | - Bai‐Cheng He
- Department of PharmacologySchool of PharmacyChongqing Medical UniversityChongqingPeople's Republic of China
- Key Laboratory for Biochemistry and Molecular Pharmacology of ChongqingChongqing Medical UniversityChongqingPeople's Republic of China
| | - Wen‐Juan Sun
- Department of PharmacologySchool of PharmacyChongqing Medical UniversityChongqingPeople's Republic of China
- Key Laboratory for Biochemistry and Molecular Pharmacology of ChongqingChongqing Medical UniversityChongqingPeople's Republic of China
| |
Collapse
|
18
|
Shen J, Li P, Shao X, Yang Y, Liu X, Feng M, Yu Q, Hu R, Wang Z. The E3 Ligase RING1 Targets p53 for Degradation and Promotes Cancer Cell Proliferation and Survival. Cancer Res 2017; 78:359-371. [PMID: 29187402 DOI: 10.1158/0008-5472.can-17-1805] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/11/2017] [Accepted: 11/21/2017] [Indexed: 11/16/2022]
Abstract
As a component of the transcriptional repression complex 1 (PRC1), the ring finger protein RING1 participates in the epigenetic regulation in cancer. However, the contributions of RING1 to cancer etiology or development are unknown. In this study, we report that RING1 is a critical negative regulator of p53 homeostasis in human hepatocellular and colorectal carcinomas. RING1 acts as an E3 ubiquitin (Ub) ligase to directly interact with and ubiquitinate p53, resulting in its proteasome-dependent degradation. The RING domain of RING1 was required for its E3 Ub ligase activity. RING1 depletion inhibited the proliferation and survival of the p53 wild-type cancer cells by inducing cell-cycle arrest, apoptosis, and senescence, with only modest effects on p53-deficient cells. Its growth inhibitory effect was partially rescued by p53 silencing, suggesting an important role for the RING1-p53 complex in human cancer. In clinical specimens of hepatocellular carcinoma, RING1 upregulation was evident in association with poor clinical outcomes. Collectively, our results elucidate a novel PRC1-independent function of RING1 and provide a mechanistic rationale for its candidacy as a new prognostic marker and/or therapeutic target in human cancer.Significance: These results elucidate a novel PRC1-independent function of RING1 and provide a mechanistic rationale for its candidacy as a new prognostic marker and/or therapeutic target in human cancer. Cancer Res; 78(2); 359-71. ©2017 AACR.
Collapse
Affiliation(s)
- Jiajia Shen
- Department of Biochemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pengyu Li
- Qilu Hospital of Shandong University, Jinan, China
| | - Xuejing Shao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yang Yang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xiujun Liu
- Department of Biochemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Feng
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Biopolis, Singapore
| | - Qiang Yu
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Biopolis, Singapore
| | - Ronggui Hu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
| | - Zhen Wang
- Department of Biochemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
19
|
Liu L, Xu F, Chang CK, He Q, Wu LY, Zhang Z, Li X. MYCN contributes to the malignant characteristics of erythroleukemia through EZH2-mediated epigenetic repression of p21. Cell Death Dis 2017; 8:e3126. [PMID: 29022893 PMCID: PMC5682688 DOI: 10.1038/cddis.2017.526] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 01/18/2023]
Abstract
MYC proto-oncogene family including c-myc and n-myc (MYCN) are critical for normal cell development and tumorigenesis. Overexpression of c-myc causes acute erythroleukemia in vivo. However, the role of MYCN in acute erythroleukemia remains poorly understood. In this study, we found that the patients with erythroleukemia showed higher expression of MYCN than normal controls. In vitro experiments, knockdown of MYCN resulted in decreased cell proliferation, elevated autonomously cell apoptosis and increased P21-mediated cell senescence. On the contrary, overexpression of MYCN obviously promoted cell proliferation, and induced erythroid differentiation block and apoptosis resistance to cytotoxic agent. Further gene microarray and functional analysis revealed that EZH2 is a target of MYCN. Knockdown of MYCN inhibited the expression of EZH2, and then activated p21 expression through removal of H3K27me3 at the p21 promoter. Overexpression of ezh2 could antagonize the p21 activation caused by MYCN knockdown. In addition, Aurora inhibitor MLN8237 inhibited the proliferation of erythroleukemia cells through repression of MYCN/EZH2 axis, whereas it minimally affected the normal hematopoietic cells. In conclusion, MYCN contributes to the malignant characteristics of erythroleukemia through EZH2-meidated epigenetic repression of p21. MYCN may serve as a therapy target for the patients with acute erythroleukemia.
Collapse
Affiliation(s)
- Li Liu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai 200233, China
| | - Feng Xu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai 200233, China
| | - Chun-Kang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai 200233, China
| | - Qi He
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai 200233, China
| | - Ling-Yun Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai 200233, China
| | - Zheng Zhang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai 200233, China
| | - Xiao Li
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai 200233, China
| |
Collapse
|
20
|
Small molecule T63 suppresses osteoporosis by modulating osteoblast differentiation via BMP and WNT signaling pathways. Sci Rep 2017; 7:10397. [PMID: 28871136 PMCID: PMC5583318 DOI: 10.1038/s41598-017-10929-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/17/2017] [Indexed: 12/30/2022] Open
Abstract
Osteoporosis results from the imbalance between bone resorption and bone formation, and restoring the normal balance of bone remodeling is highly desirable for identification of better treatment. In this study, using a cell-based high-throughput screening model representing Runt-related transcription factor 2 (RUNX2) transcriptional activity, we identified a novel small-molecular-weight compound, T63, as an efficient up-regulator of osteogenesis. T63 increased the alkaline phosphatase (ALPL) activity and mineralization as well as gene expression of Alpl and other osteogenic marker genes in mouse osteoblasts and mesenchymal stem cell-like cells. Upon induction of osteoblast differentiation, T63 inhibited adipogenic differentiation in the pluripotent mesenchymal cells. Consistently, T63 up-regulated RUNX2 mRNA and protein levels, and knockdown of RUNX2 reduced the osteogenic role of T63. Mechanistically, T63 activated both BMPs and WNT/β-catenin signaling pathways. Inhibition of either signaling pathway with specific inhibitor suppressed T63-induced RUNX2 expression and the osteogenic phenotypes. Moreover, T63 markedly protected against bone mass loss in the ovariectomized and dexamethasone treated rat osteoporosis model. Collectively, our data demonstrate that T63 could be a promising drug candidate and deserves further development for potential therapeutics in osteoporosis.
Collapse
|
21
|
Huang D, Feng X, Liu Y, Deng Y, Chen H, Chen D, Fang L, Cai Y, Liu H, Wang L, Wang J, Yang Z. AQP9-induced cell cycle arrest is associated with RAS activation and improves chemotherapy treatment efficacy in colorectal cancer. Cell Death Dis 2017. [PMID: 28640255 PMCID: PMC5520935 DOI: 10.1038/cddis.2017.282] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aquaporin-9 (AQP9) expression is associated with arsenic sensitivity in leukemia cells. However, the role of AQP9 in regulating tumor sensitivity to adjuvant chemotherapy in colorectal cancer (CRC) has not been elucidated. In this study, we demonstrated that AQP9 can serve as an independent predictive marker for adjuvant chemotherapy in CRC. Patients with high AQP9 expression had higher rate of disease-free survival (DFS) than those with low AQP9 expression. Upregulation of AQP9 was associated with enhanced chemosensitivity to 5-fluorouracil (5-FU) both in vitro and in vivo. Overexpression of AQP9 resulted in an increased intracellular level of 5-FU in CRC cells, hence leading to a higher percentage of apoptosis after 5-FU treatment. Moreover, AQP9 is positively associated with RAS activation and other downstream signaling molecules in CRC. AQP9 overexpression resulted in p21 upregulation and induced S-phase arrest. Taken together, AQP9 enhances the cytotoxic response to 5-FU in CRC cells by simultaneously inducing S-phase arrest via activation of RAS signaling and facilitating drug uptake. Our results suggest that AQP9 might be a novel predictor for the benefit of 5-FU-based chemotherapy in CRC. The identification of AQP9-induced tumor sensitivity to 5-FU highlights the role of AQP9 in regulating chemosensitivity in CRC.
Collapse
Affiliation(s)
- Dandan Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Xingzhi Feng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Yiting Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Yanhong Deng
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China.,Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Hao Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Daici Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Lekun Fang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Yue Cai
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China.,Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Huanliang Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Lei Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Jianping Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Zihuan Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| |
Collapse
|