1
|
Dai JH, Xu ZH, Li QL, Huang J, Niu Z, Zhang CH, Hu S, Sun R, Li YC. TRIM14-NF-κB pathway in the anterior cingulate cortex modulates comorbid depressive symptoms in chronic pain. Mol Pain 2025; 21:17448069251335503. [PMID: 40200729 PMCID: PMC12035169 DOI: 10.1177/17448069251335503] [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: 01/02/2025] [Revised: 03/22/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
Depression is commonly observed in individuals suffering from chronic pain, but the exact molecular mechanisms behind these symptoms are still not fully understood. This study highlights the important role of the TRIM14-NF-κB pathway in the anterior cingulate cortex (ACC) in regulating comorbid depressive symptoms associated with chronic pain. Our results show that the CFA model induces both chronic pain and depression-like behaviors in mice, with significant activation of the ACC brain regions. Specifically, the protein expression of TRIM14 was notably elevated in the ACC of CFA mice. Furthermore, reducing TRIM14 expression alleviated both chronic pain and depression-like behaviors in these mice. In addition, we also discovered that NF-κB may act as a downstream target of TRIM14, as silencing TRIM14 expression led to a reduction in the levels of phosphorylated NF-κB. Notably, inhibiting NF-κB produced similar improvements in chronic pain and depression-like behaviors, mirroring the effects observed with TRIM14 knockdown. In summary, our findings emphasize the critical role of the TRIM14-NF-κB pathway in regulating chronic pain and depression-like behaviors in the CFA mouse model. These insights provide a foundation for further exploration of the molecular mechanisms underlying chronic pain and depression, and may guide the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jia-Hao Dai
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Zhen-Hua Xu
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Qiu-Lan Li
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jie Huang
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Zheng Niu
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Chen-Hao Zhang
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Shufen Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Ren Sun
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yong-Chang Li
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
2
|
Li X, Zhou F, Niu K, Wang Y, Shi Y, Li Y, Gao X, Zhao W, Chen T, Zhang Y. Emerging discoveries on the role of TRIM14: from diseases to immune regulation. Cell Death Discov 2024; 10:513. [PMID: 39719450 DOI: 10.1038/s41420-024-02276-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024] Open
Abstract
TRIM14 is an important member of the TRIM family and is widely expressed in a variety of tissues. Like other members of the TRIM family, TRIM14 is also involved in ubiquitination modifications. TRIM14 was initially reported as an interferon-stimulated gene (ISG). In recent years, many studies have focused on the regulatory role of TRIM14 in signaling pathways such as the PI3K/Akt, NF-κB, and cGAS/STING pathways and revealed its mechanism of action in a variety of pathophysiological processes, and the regulation of TRIM14 has attracted the interest of many researchers as a new direction for the treatment of various diseases. However, there are no reviews on the role of TRIM14 in diseases. In this paper, we will describe the structure of TRIM14, review its role in cancer, cardiovascular disease, cervical spondylosis, inflammation and antiviral immunity, and provide an outlook on future research directions.
Collapse
Affiliation(s)
- Xinhao Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Feilong Zhou
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kaiyi Niu
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yizhu Wang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yanlong Shi
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yunxin Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xin Gao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Weijie Zhao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tianyi Chen
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
3
|
Wei S, Ai M, Zhan Y, Yu J, Xie T, Hu Q, Fang Y, Huang X, Li Y. TRIM14 suppressed the progression of NSCLC via hexosamine biosynthesis pathway. Carcinogenesis 2024; 45:324-336. [PMID: 38267812 DOI: 10.1093/carcin/bgae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 01/26/2024] Open
Abstract
Tripartite Motif 14 (TRIM14) is an oncoprotein that belongs to the E3 ligase TRIM family, which is involved in the progression of various tumors except for non-small cell lung carcinoma (NSCLC). However, little is currently known regarding the function and related mechanisms of TRIM14 in NSCLC. Here, we found that the TRIM14 protein was downregulated in lung adenocarcinoma tissues compared with the adjacent tissues, which can suppress tumor cell proliferation and migration both in vitro and in vivo. Moreover, TRIM14 can directly bind to glutamine fructose-6-phosphate amidotransferase 1 (GFAT1), which in turn results in the degradation of GFAT1 and reduced O-glycosylation levels. GFAT1 is a key enzyme in the rate-limiting step of the hexosamine biosynthetic pathway (HBP). Replenishment of N-acetyl-d-glucosamine can successfully reverse the inhibitory effect of TRIM14 on the NSCLC cell growth and migration as expected. Collectively, our data revealed that TRIM14 suppressed NSCLC cell proliferation and migration through ubiquitination and degradation of GFAT1, providing a new regulatory role for TRIM14 on HBP.
Collapse
Affiliation(s)
- Sisi Wei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Meiling Ai
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- Department of Pharmacy, Jiangxi Maternal and Child Health Hospital, Nanchang 330006, China
| | - Yuan Zhan
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Jieqing Yu
- Department of Otorhinolaryngology Head and Neck Surgery, Jiangxi Otorhinolaryngology Head and Neck Surgery Institute, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Tao Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Qinghua Hu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yang Fang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yong Li
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| |
Collapse
|
4
|
Hou H, Chen Y, Feng X, Xu G, Yan M. Tripartite motif‑containing 14 may aggravate cardiac hypertrophy via the AKT signalling pathway in neonatal rat cardiomyocytes and transgenic mice. Mol Med Rep 2023; 28:173. [PMID: 37503784 PMCID: PMC10433706 DOI: 10.3892/mmr.2023.13060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/25/2023] [Indexed: 07/29/2023] Open
Abstract
Tripartite motif‑containing 14 (TRIM14) is an E3 ubiquitin ligase that primarily participates in the natural immune response and in tumour development via ubiquitination. However, the role of TRIM14 in cardiac hypertrophy is not currently clear. The present study examined the role of TRIM14 in cardiac hypertrophy and its potential molecular mechanism. TRIM14 was overexpressed in neonatal rat cardiomyocytes using adenovirus and cardiomyocyte hypertrophy was induced using phenylephrine (PE). Cardiomyocyte hypertrophy was assessed by measuring cardiomyocyte surface area and markers of hypertrophy. In addition, TRIM14‑transgenic (TRIM14‑TG) mice were created and cardiac hypertrophy was induced using transverse aortic constriction (TAC). Cardiac function, heart weight‑to‑body weight ratio (HW/BW), cardiomyocyte cross‑sectional area, cardiac fibrosis and hypertrophic markers were further examined. The expression of AKT signalling pathway‑related proteins was detected. TRIM14 overexpression in cardiomyocytes promoted PE‑induced increases in cardiomyocyte surface area and hypertrophic markers. TRIM14‑TG mice developed worse cardiac function, greater HW/BW, cross‑sectional area and cardiac fibrosis, and higher levels of hypertrophic markers in response to TAC. TRIM14 overexpression also increased the phosphorylation levels of AKT, GSK‑3β, mTOR and p70S6K in vivo and in vitro. To the best our knowledge, the present study was the first to reveal that overexpression of TRIM14 aggravated cardiac hypertrophy in vivo and in vitro, which may be related to activation of the AKT signalling pathway.
Collapse
Affiliation(s)
- Hongwei Hou
- Department of Cardiology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Yan Chen
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Xiuyuan Feng
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Guang Xu
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Min Yan
- Department of General Practice, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
5
|
Salem ML, Atia I, Elmashad NM. Higher cytotoxic activities of CD8 + T cells and natural killer cells from peripheral blood of early diagnosed lung cancer patients. BMC Immunol 2023; 24:24. [PMID: 37580655 PMCID: PMC10426146 DOI: 10.1186/s12865-023-00553-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/27/2023] [Indexed: 08/16/2023] Open
Abstract
INTRODUCTION Cytotoxic (CD8+) and natural killer (NK) cells play critical roles in anti-tumor immunity. Dysfunction in these cells is considered as one of the extrinsic mechanisms for tumor relapse. AIM We aimed in this study to assess cytotoxic activities of CD8 + T and NK cells in the peripheral blood from lung cancer patients before and after induction of chemotherapy. SUBJECTS AND METHODS Healthy (n = 5) volunteers and lung cancer patients (n = 15:5 before, 5 during, and 5 after induction of chemotherapy) were recruited. Flow cytometry was used to analyze the numbers of CD8 + T cells, NK and CD56+T cells and their intracellular expression of granzyme B (GzB) in fresh peripheral blood mononuclear cells (PBMCs) and after 72 h of their culture in vitro and stimulation with 5 µg/ml Concanavalin A (Con A) and 50ng/ml IL-2). In addition, the plasma levels of inflammatory cytokines were measured using luminex. RESULTS After culture, significant increases in the number of GzB expressing cells gated on CD3+, CD4+, CD8 + and NKCD8 + T cells in the PBMCs from lung cancer patients before induction of chemotherapy as compared to control individuals as well as patients during and after induction of chemotherapy. Serum levels of IL-1 and CXCL8 in patients before induction of chemotherapy showed 37- and 40-fold increases, respectively, as compared to control individuals. Both GzB expression and cytokines levels in patients during and after chemotherapy were similar. CONCLUSION Polyclonal stimulation of PBMCs can restore the cytolytic activities of cytotoxic CD8 and NK cells from lung cancer patients even after chemotherapy.
Collapse
Affiliation(s)
- Mohamed Labib Salem
- Zoology Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
- Center of Excellence in Cancer Research, New Tanta University Teaching Hospital, Tanta, Egypt.
| | - Ismail Atia
- Center of Excellence in Cancer Research, New Tanta University Teaching Hospital, Tanta, Egypt
- Zoology Department, Faculty of Science, Al-Azhar University, Assuit, Egypt
| | - Nehal M Elmashad
- Oncology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
6
|
Lou Y, Lu J, Zhang Y, Gu P, Wang H, Qian F, Zhou W, Zhang W, Zhong H, Han B. The centromere-associated protein CENPU promotes cell proliferation, migration, and invasiveness in lung adenocarcinoma. Cancer Lett 2022; 532:215599. [PMID: 35176420 DOI: 10.1016/j.canlet.2022.215599] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/19/2022] [Accepted: 02/13/2022] [Indexed: 12/01/2022]
Abstract
CENPU, encoding an important factor involved in kinetochore assembly during mitosis, is associated with shorter survival rates in lung adenocarcinoma (LUAD) patients. CENPU promotes growth rates and invasive behavior of LUAD cells; however, its mechanism of action in LUAD progression remains to be elucidated. CENPU mRNA and protein expression were elevated in LUAD tumors, and high CENPU gene expression was associated with inferior survival prognosis in LUAD patients. CENPU knockdown inhibited LUAD cell proliferation, clone formation, migration, invasion, and epithelial-mesenchymal transition (EMT) in addition to inducing cell cycle arrest and apoptosis in vitro and reduced LUAD xenograft tumor growth in vivo. Furthermore, we identified CENPU-regulated genes significantly enriched for proliferation and apoptosis pathways, and identified HSP Family Member C10 (DNAJC10) as putative effector of CENPU. CENPU knockdown produced DNAJC10 protein downregulation, and DNAJC10 overexpression partially rescued the phenotypic effects of CENPU knockdown in LUAD cells. Moreover, CENPU's coiled-coil domain was essential for CENPU's phenotypic effects in LUAD cells. In conclusion, the kinetochore component CENPU plays a critical role in LUAD cell proliferation and invasiveness. Targeting CENPU-DNAJC10 axis may inhibit LUAD tumor cell proliferation and metastasis.
Collapse
Affiliation(s)
- Yuqing Lou
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Lu
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanwei Zhang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Gu
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huimin Wang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfei Qian
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wensheng Zhou
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Hua Zhong
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Baohui Han
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
7
|
Liu B, Zhou F, Liu H, Wang Y, Wang J, Ren F, Xu S. Knockdown of LINC00511 decreased cisplatin resistance in non-small cell lung cancer by elevating miR-625 level to suppress the expression of leucine rich repeat containing eight volume-regulated anion channel subunit E. Hum Exp Toxicol 2022; 41:9603271221089000. [PMID: 35363093 DOI: 10.1177/09603271221089000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND LINC00511 has been reported as a biomarker related to the prognosis of non-small cell lung cancer (NSCLC), but the molecular mechanism and exact functions of LINC00511 in chemoresistance of NSCLC remain to be elucidated. METHODS RT-qPCR was used to evaluate the mRNA expression of LINC00511, miR-625, and leucine rich repeat containing 8 volume-regulated anion channel subunit E (LRRC8E). Western blotting detected the protein levels of Ki-67, MMP-9, cleaved-caspase-3. The interaction between miR-625 and LINC00511 or LRRC8E was verified by luciferase reporter assays. CCK-8, TUNEL, and Transwell assays were used to evaluate IC50 value, proliferation, migration, and invasion of NSCLC cells. RESULTS In our study, it was discovered that the levels of LINC00511 and LRRC8E were increased, while miR-625 expression was decreased in NSCLC tissues, DDP-resistant NSCLC cells, and non-resistant NSCLC cells. LINC00511 depletion significantly curbed cell growth, IC50 value, and metastasis in DDP-resistant NSCLC cells. In addition, the influence of LINC00511 deficiency on the DDP resistance in NSCLC was overturned by suppressing miR-625. Furthermore, LRRC8E overexpression abolished the promotive effect of miR-625 abundance on the DDP sensitivity in DDP-resistant NSCLC cells. CONCLUSION Our results demonstrated that LINC00511 increased DDP resistance in NSCLC by suppressing miR-625 to upregulate LRRC8E.
Collapse
Affiliation(s)
- Benkun Liu
- Department of Thoracic Surgery, 91631Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Fucheng Zhou
- Department of Thoracic Surgery, 91631Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - He Liu
- Department of Thoracic Surgery, 91631Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanbo Wang
- Department of Thoracic Surgery, 91631Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Junfeng Wang
- Department of Thoracic Surgery, 91631Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Fenghai Ren
- Department of Thoracic Surgery, 91631Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Shidong Xu
- Department of Thoracic Surgery, 91631Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
8
|
Liu W, Xia L, Xia Z, Chen L. Comprehensive Analysis of Innate Immunophenotyping Based on Immune Score Predicting Immune Alterations and Prognosis in Breast Cancer Patients. Genes (Basel) 2021; 13:88. [PMID: 35052427 PMCID: PMC8774675 DOI: 10.3390/genes13010088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is the most common cancer, with the highest mortality rate and the most diagnosed cancer type in women worldwide. To identify the effect innate immune checkpoint for breast cancer immunotherapy, the innate immune prognostic biomarkers were selected through the ICI score model and the risk model in breast cancer patients. Moreover, the reliability and accuracy of the ICI score model and the risk model were further examined through the analysis of breast cancer prognosis and immune cell infiltration. The pan cancer analysis further confirmed and selected CXCL9 as the key innate immune checkpoint for breast cancer immunotherapy and identified three small molecular drugs for target CXCL9 through molecular docking analysis. In summary, CXCL9 significantly correlated with the prognostic of breast cancer and immune cell infiltration and could be innate immune checkpoint for breast cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Liming Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing 210023, China; (W.L.); (L.X.); (Z.X.)
| |
Collapse
|
9
|
Huang Y, Xiao Y, Zhang X, Huang X, Li Y. The Emerging Roles of Tripartite Motif Proteins (TRIMs) in Acute Lung Injury. J Immunol Res 2021; 2021:1007126. [PMID: 34712740 PMCID: PMC8548118 DOI: 10.1155/2021/1007126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/09/2021] [Indexed: 11/21/2022] Open
Abstract
Acute lung injury (ALI) is an inflammatory disorder of the lung that causes high mortality and lacks any pharmacological intervention. Ubiquitination plays a critical role in the pathogenesis of ALI as it regulates the alveolocapillary barrier and the inflammatory response. Tripartite motif (TRIM) proteins are one of the subfamilies of the RING-type E3 ubiquitin ligases, which contains more than 80 distinct members in humans involved in a broad range of biological processes including antivirus innate immunity, development, and tumorigenesis. Recently, some studies have shown that several members of TRIM family proteins play important regulatory roles in inflammation and ALI. Herein, we integrate emerging evidence regarding the roles of TRIMs in ALI. Articles were selected from the searches of PubMed database that had the terms "acute lung injury," "ubiquitin ligases," "tripartite motif protein," "inflammation," and "ubiquitination" using both MeSH terms and keywords. Better understanding of these mechanisms may ultimately lead to novel therapeutic approaches by targeting TRIMs for ALI treatment.
Collapse
Affiliation(s)
- Yingjie Huang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yue Xiao
- The First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Xuekang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Yong Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
10
|
Mohammadi A, Pour Abbasi MS, Khorrami S, Khodamoradi S, Mohammadi Goldar Z, Ebrahimzadeh F. The TRIM proteins in cancer: from expression to emerging regulatory mechanisms. Clin Transl Oncol 2021; 24:460-470. [PMID: 34643877 DOI: 10.1007/s12094-021-02715-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022]
Abstract
New clinical evidence suggests that dysregulation of the ubiquitin-mediated destruction of tumor suppressors or oncogene products is probably engaged in the etiology of leukemia and carcinoma. The superfamily of tripartite motif (TRIM)-containing protein family is among the biggest recognized single protein RING finger E3 ubiquitin ligases that are considered vital carcinogenesis regulators, which is not shocking since TRIM proteins are engaged in various biological processes, including cell growth, development, and differentiation; hence, TRIM proteins' alterations may influence apoptosis, cell proliferation, and transcriptional regulation. In this review article, the various mechanisms through which TRIM proteins exert their role in the most prevalent malignancies including lung, prostate, colorectal, liver, breast, brain cancer, and leukemia are summarized.
Collapse
Affiliation(s)
- A Mohammadi
- Department of Genetics Islamic, Azad University of Marand, Marand, Iran
| | | | - S Khorrami
- Tehran University of Medical Sciences, Tehran, Iran
| | - S Khodamoradi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Z Mohammadi Goldar
- Department of Medicine, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - F Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Wang WJ, Yuan Y, Zhang D, Liu P, Liu F. miR-671-5p repressed progression of papillary thyroid carcinoma via TRIM14. Kaohsiung J Med Sci 2021; 37:983-990. [PMID: 34292652 DOI: 10.1002/kjm2.12424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/20/2021] [Accepted: 06/21/2021] [Indexed: 11/11/2022] Open
Abstract
The pivotal role of dysregulated miRNAs in development of papillary thyroid carcinoma has been emphasized in recent research. miR-671-5p was previously documented to function as a tumor suppressor. However, the role and mechanism of miR-671-5p in progression of papillary thyroid carcinoma remain to be further studied. Data from functional assays indicated that forced expression of miR-671-5p decreased cell viability, repressed cell proliferation, migration, and invasion in papillary thyroid carcinoma cells. In vivo study showed that miR-671-5p overexpression inhibited tumor growth, downregulated Ki67, and decreased tumor volume and weight. Tripartite motif containing 14 (TRIM14) was verified as downstream target of miR-671-5p. The expression of TRIM14 was suppressed by miR-671-5p in papillary thyroid carcinoma. Overexpression of TRIM14 increased cell viability, and promoted the proliferation, migration, and invasion of papillary thyroid carcinoma. Moreover, TRIM14 counteracted the suppressive effect of miR-671-5p overexpression on papillary thyroid carcinoma cell growth. In conclusion, miR-671-5p repressed progression of papillary thyroid carcinoma through downregulation of TRIM14, providing a promising target for therapy of papillary thyroid carcinoma.
Collapse
Affiliation(s)
- Wan-Ju Wang
- Department of General Surgery, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan City, China
| | - Yuan Yuan
- Department of Thyroid and Breast Surgery, Wuhan No. 1 Hospital, Wuhan City, China
| | - Dong Zhang
- Department of General Surgery, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan City, China
| | - Piao Liu
- Department of General Surgery, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan City, China
| | - Fang Liu
- Department of General Surgery, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan City, China
| |
Collapse
|
12
|
Grieve S, Ding K, Moore J, Finniss M, Ray A, Lees M, Hossain F, Murugesan A, Agar J, Acar C, Taylor J, Shepherd FA, Reiman T. Immunohistochemical validation study of 15-gene biomarker panel predictive of benefit from adjuvant chemotherapy in resected non-small-cell lung cancer: analysis of JBR.10. ESMO Open 2021; 5:S2059-7029(20)30069-7. [PMID: 32220948 PMCID: PMC7174014 DOI: 10.1136/esmoopen-2020-000679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
Objective There are no validated approaches to predict benefit from adjuvant chemotherapy for resected patients with non-small-cell lung cancer (NSCLC). The aim of this study was to translate a 15-gene mRNA expression profile published by Zhu et al, shown to be prognostic and predictive of benefit, into a readily applicable immunohistochemistry (IHC) panel. Methods For seven of the genes in the gene expression profile (GEP) for which suitable commercial antibodies were available, we semiquantitatively assessed the IHC expression and prognostic significance for 173 patients treated at the Saint John Regional Hospital (SJRH). Cut-offs for high and low expression were defined for each marker and applied to IHC scores from 291 of the 482 patients in JBR.10, including patients on both the adjuvant chemotherapy and observation arms. The prognostic and predictive value of these markers on overall survival (OS) or recurrence-free survival (RFS) was assessed by Cox regression models. Results In the SJRH cohort, in 62 patients with resected stage II–III NSCLC, the prognostic significance of IHC assays for four proteins were concordant with Zhu’s GEP results. Low FOSL2 (OS, HR=0.15; p=0.0001; RFS, HR=0.14; p<0.0001) and high STMN2 (RFS, HR=2.501; p=0.0197) were adverse prognostic factors. Low ATP1B1 and low TRIM14 expression trended toward worse OS and RFS. Validation of these markers with JBR.10 patients failed to show prognostic significance either individually or in combined risk classifications. Additionally, the interaction between these markers and chemotherapy treatment in predicting OS (FOSL2, p=0.52; STMN2 p=0.14; ATP1B1, p=0.33; TRIM14, p=0.81) or RFS (FOSL2, p=0.63; STMN2, p=0.12; ATP1B1, p=0.66; TRIM14, p=0.57) did not reach significance, individually or in combination panels. Conclusions Zhu’s GEP could not be translated into an IHC panel predictive of benefit from adjuvant chemotherapy. Future predictive biomarker analysis in the adjuvant NSCLC setting may need to focus on novel therapies.
Collapse
Affiliation(s)
- Stacy Grieve
- Department of Biology, University of New Brunswick Saint John, Saint John, New Brunswick, Canada
| | - Keyue Ding
- Canadian Cancer Trials Group, Kingston, Ontario, Canada
| | - Jonathan Moore
- Department of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Mathew Finniss
- Department of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Ayush Ray
- Department of Biology, University of New Brunswick Saint John, Saint John, New Brunswick, Canada
| | - Miranda Lees
- Department of Biology, University of New Brunswick Saint John, Saint John, New Brunswick, Canada
| | - Faisal Hossain
- Department of Biology, University of New Brunswick Saint John, Saint John, New Brunswick, Canada
| | - Alli Murugesan
- Department of Biology, University of New Brunswick Saint John, Saint John, New Brunswick, Canada.,Department of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Jane Agar
- Department of Pathology, Saint John Regional Hospital, Saint John, New Brunswick, Canada
| | - Cenk Acar
- Department of Pathology, Saint John Regional Hospital, Saint John, New Brunswick, Canada
| | - James Taylor
- Canadian Cancer Trials Group, Kingston, Ontario, Canada
| | - Frances A Shepherd
- Department of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Tony Reiman
- Department of Biology, University of New Brunswick Saint John, Saint John, New Brunswick, Canada .,Department of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| |
Collapse
|
13
|
Chen J, Huang L, Quan J, Xiang D. TRIM14 regulates melanoma malignancy via PTEN/PI3K/AKT and STAT3 pathways. Aging (Albany NY) 2021; 13:13225-13238. [PMID: 33982666 PMCID: PMC8148494 DOI: 10.18632/aging.203003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/14/2021] [Indexed: 12/14/2022]
Abstract
Melanoma is one of the most aggressive cancers with poor overall survival. To date, there are still few effective methods for the treatment of melanoma. TRIM14 was previously reported to be an important oncogene in many tumors. Nevertheless, the roles of TRIM14 in melanoma remain unknown. In this study, we found that TRIM14 was abnormally upregulated in melanoma cell lines. Knockdown of TRIM14 suppressed melanoma cell proliferation, migration, invasion, epithelial-mesenchymal transition, and melanin synthesis. Overexpression of TRIM14 had opposite effects on the cellular functions of melanoma cell lines. Further study revealed that TRIM14 knockdown increased PTEN protein levels, which in turn inactivated AKT and STAT3 pathways. Moreover, blocking AKT or STAT3 pathway with a specific inhibitor could partially reverse the promotion of melanoma malignancy mediated by TRIM14 overexpression. In addition, in vivo assay also supported the above findings. These results indicated that TRIM14 might be a promising target for melanoma treatment.
Collapse
Affiliation(s)
- Jiangyan Chen
- Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing, China
| | - Lin Huang
- Department of Dermatology, Jiangjin Central Hospital of Chongqing, Chongqing, China
| | - Jin Quan
- Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing, China
| | - Debing Xiang
- Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing, China
| |
Collapse
|
14
|
LncRNA MSC-AS1 Promotes Colorectal Cancer Progression by Regulating miR-325/TRIM14 Axis. JOURNAL OF ONCOLOGY 2021; 2021:9954214. [PMID: 34054957 PMCID: PMC8131164 DOI: 10.1155/2021/9954214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023]
Abstract
Background LncRNA MSC-AS1 has been reported to be a tumor promoter in hepatocellular carcinoma. However, the function of MSC-AS1 in colorectal cancer (CRC) has not been elucidated. It is designed to study the expression level of MSC-AS1 and investigate its biological effect on the progression of CRC. Methods The expression patterns of MSC-AS1, miR-325, and TRIM14 were explored by RT-qPCR in CRC tissues and cells. The protein expression of TRIM14 was tested by Western blot assay. The association between MSC-AS1 expression and clinicopathological data was analyzed by chi-squared test. CCK-8 assay, colony formation, and Transwell assay were used to investigate the effect of MSC-AS1 on cell growth, invasion, and migration in CRC cells. The correlations among MSC-AS1, miR-325, and TRIM14 were analyzed by Pearson's correlation coefficient analysis. Results We found that MSC-AS1 and TRIM14 were upregulated in CRC tissues, while miR-325 was downregulated in CRC tissues. Functional experiments demonstrated that MSC-AS1 knockdown inhibited cell proliferation, migration, and invasion abilities in CRC cells. Additionally, miR-325 was proved to be a target miRNA of MSC-AS1, and TRIM14 might be a downstream gene of miR-325. Besides that, MSC-AS1 counteracted the inhibitory effect of miR-325 on the cell progression and TRIM14 expression. Conclusion Our results indicated that MSC-AS1 facilitated CRC progression by sponging miR-325 to upregulate TRIM14 expression. We suggested that MSC-AS1 might be a potential lncRNA-target for CRC therapy.
Collapse
|
15
|
Zhao G, Liu C, Wen X, Luan G, Xie L, Guo X. The translational values of TRIM family in pan-cancers: From functions and mechanisms to clinics. Pharmacol Ther 2021; 227:107881. [PMID: 33930453 DOI: 10.1016/j.pharmthera.2021.107881] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023]
Abstract
Cancer is the second leading cause of human death across the world. Tripartite motif (TRIM) family, with E3 ubiquitin ligase activities in majority of its members, is reported to be involved in multiple cellular processes and signaling pathways. TRIM proteins have critical effects in the regulation of biological behaviors of cancer cells. Here, we discussed the current understanding of the molecular mechanism of TRIM proteins regulation of cancer cells. We also comprehensively reviewed published studies on TRIM family members as oncogenes or tumor suppressors in the oncogenesis, development, and progression of a variety of types of human cancers. Finally, we highlighted that certain TRIM family members are potential molecular biomarkers for cancer diagnosis and prognosis, and potential therapeutic targets.
Collapse
Affiliation(s)
- Guo Zhao
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Chuan Liu
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Xin Wen
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Gan Luan
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Longxiang Xie
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiangqian Guo
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| |
Collapse
|
16
|
Xie X, Wang F, Li X. Inhibition of TRIM14 protects cerebral ischemia/reperfusion injury through regulating NF-κB/NLRP3 pathway-mediated inflammation and apoptosis. J Recept Signal Transduct Res 2021; 42:197-205. [PMID: 33691569 DOI: 10.1080/10799893.2021.1887218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Many proteins in tripartite motif (TRIM) family have been reported to play an important role in cerebral ischemia/reperfusion (I/R) injury. This study was designed to investigate the effect of TRIM14 on the cerebral I/R injury in rats. METHODS The rat model was constructed through inserting thread into the middle cerebral artery. The expression of TRIM14 was measured by qRT-PCR, immunoblotting, and immunofluorescence. The hippocampal sections were stained with 2,3,5-triphenyltetrazolium chloride (TTC) to determine infarct volume and used for measuring the neurologic deficit score and brain water content. The H&E staining was used for immunohistochemical (IHC) staining. The number of apoptotic cells was measured by fluorescence microscopy. The levels of IL-6, IL-1β, and TNFα were detected by qRT-PCR and ELISA. The swimming speed, latency time, and number of platform crossings were measured by the water maze test. RESULTS TRIM14 was significantly enhanced in rats with cerebral I/R injury compared to Sham rats, showing its highest level at 24 h after I/R. TRIM14 inhibition reduced ischemic brain injury, suppressed neuron apoptosis, suppressed inflammation, and improved cognitive dysfunction in rats with cerebral I/R injury. TRIM14 inhibition also suppressed the activation of NF-κB/NLRP3 pathway in rats with cerebral I/R injury. CONCLUSION In conclusion, the expression of TRIM14 was increased in rats with cerebral I/R injury, the protective effect of TRIM14 inhibitor on cerebral I/R injury in rats depends on its anti-apoptotic and anti-inflammatory effect. The underlying mechanism was, at least partially, through regulating NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Xianlong Xie
- Department of Geriatrics, Wuhan No. 1 Hospital, Wuhan, China
| | - Fan Wang
- Department of Geriatrics, Wuhan No. 1 Hospital, Wuhan, China
| | - Xiujuan Li
- Department of Geriatrics, Wuhan No. 1 Hospital, Wuhan, China
| |
Collapse
|
17
|
Wang X, Veeraraghavan J, Liu CC, Cao X, Qin L, Kim JA, Tan Y, Loo SK, Hu Y, Lin L, Lee S, Shea MJ, Mitchell T, Li S, Ellis MJ, Hilsenbeck SG, Schiff R, Wang XS. Therapeutic Targeting of Nemo-like Kinase in Primary and Acquired Endocrine-resistant Breast Cancer. Clin Cancer Res 2021; 27:2648-2662. [PMID: 33542078 DOI: 10.1158/1078-0432.ccr-20-2961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/29/2020] [Accepted: 02/01/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Endocrine resistance remains a major clinical challenge in estrogen receptor (ER)-positive breast cancer. Despite the encouraging results from clinical trials for the drugs targeting known survival signaling, relapse is still inevitable. There is an unmet need to discover new drug targets in the unknown escape pathways. Here, we report Nemo-like kinase (NLK) as a new actionable kinase target that endows previously uncharacterized survival signaling in endocrine-resistant breast cancer. EXPERIMENTAL DESIGN The effects of NLK inhibition on the viability of endocrine-resistant breast cancer cell lines were examined by MTS assay. The effect of VX-702 on NLK activity was verified by kinase assay. The modulation of ER and its coactivator, SRC-3, by NLK was examined by immunoprecipitation, kinase assay, luciferase assay, and RNA sequencing. The therapeutic effects of VX-702 and everolimus were tested on cell line- and patient-derived xenograft (PDX) tumor models. RESULTS NLK overexpression endows reduced endocrine responsiveness and is associated with worse outcome of patients treated with tamoxifen. Mechanistically, NLK may function, at least in part, via enhancing the phosphorylation of ERα and its key coactivator, SRC-3, to modulate ERα transcriptional activity. Through interrogation of a kinase profiling database, we uncovered and verified a highly selective dual p38/NLK inhibitor, VX-702. Coadministration of VX-702 with the mTOR inhibitor, everolimus, demonstrated a significant therapeutic effect in cell line-derived xenograft and PDX tumor models of acquired or de novo endocrine resistance. CONCLUSIONS Together, this study reveals the potential of therapeutic modulation of NLK for the management of the endocrine-resistant breast cancers with active NLK signaling.
Collapse
Affiliation(s)
- Xian Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Chia-Chia Liu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Xixi Cao
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Lanfang Qin
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jin-Ah Kim
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Ying Tan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Suet Kee Loo
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Yiheng Hu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Ling Lin
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Sanghoon Lee
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Martin J Shea
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Tamika Mitchell
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Shunqiang Li
- Department of Medicine, Washington University School of Medicine at St Louis, St. Louis, Missouri
| | - Matthew J Ellis
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Susan G Hilsenbeck
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Xiao-Song Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania. .,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
18
|
Zhan W, Zhang S. TRIM proteins in lung cancer: Mechanisms, biomarkers and therapeutic targets. Life Sci 2021; 268:118985. [PMID: 33412211 DOI: 10.1016/j.lfs.2020.118985] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/13/2020] [Accepted: 12/22/2020] [Indexed: 12/24/2022]
Abstract
The tripartite motif (TRIM) family is defined by the presence of a Really Interesting New Gene (RING) domain, one or two B-box motifs and a coiled-coil region. TRIM proteins play key roles in many biological processes, including innate immunity, tumorigenesis, cell differentiation and ontogenetic development. Alterations in TRIM gene and protein levels frequently emerge in a wide range of tumors and affect tumor progression. As canonical E3 ubiquitin ligases, TRIM proteins participate in ubiquitin-dependent proteolysis of prominent components of the p53, NF-κB and PI3K/AKT signaling pathways. The occurrence of ubiquitylation events induced by TRIM proteins sustains internal balance between tumor suppressive and tumor promoting genes. In this review, we summarized the diverse mechanism of TRIM proteins responsible for the most common malignancy, lung cancer. Furthermore, we also discussed recent progress in both the diagnosis and therapeutics of tumors contributed by TRIM proteins.
Collapse
Affiliation(s)
- Weihua Zhan
- Ecology and Health Institute, Hangzhou Vocational & Technical College, Hangzhou 310018, China.
| | - Song Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
19
|
Wu Y, Cheng K, Liang W, Wang X. lncRNA RPPH1 promotes non-small cell lung cancer progression through the miR-326/WNT2B axis. Oncol Lett 2020; 20:105. [PMID: 32831924 PMCID: PMC7439152 DOI: 10.3892/ol.2020.11966] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) serve important regulatory roles in human tumors. The aim of the present study was to examine the role of ribonuclease P RNA component H1 (RPPH1) in non-small cell lung cancer (NSCLC). RPPH1 expression was assessed in datasets from The Cancer Genome Atlas, as well as lung cancer cell lines and patients with NSCLC. RPPH1 was significantly upregulated in NSCLC cell lines, compared with a normal lung epithelial cell line. Moreover, high RPPH1 expression was associated with poor overall survival and disease progression. RPPH1 was knocked down in A549 and H1299 cells using short hairpin (sh) RNA constructs, and the expressions of target genes and proteins were determined by reverse transcription-quantitative PCR and western blotting. Cell invasion potential was also determined using Transwell Matrigel assays. Compared with the negative control, RPPH1 silencing significantly reduced the number of invading cells, increased E-cadherin expression and reduced vimentin protein expression. Cell resistance to cisplatin/cis-diamminedichloridoplatinum (CDDP) was also evaluated using Cell Counting Kit-8 and colony formation assays. RPPH1 overexpression increased the resistance of A549 and H1299 cells to CDDP. Moreover, the potential interactions between RPPH1, microRNA (miR)-326 and Wnt family member 2B (WNT2B) were investigated using luciferase reporter assays and co-transfection experiments. MiR-326 expression was directly inhibited by RPPH1. In A549 cells co-transfected with shRPPH1 and miR-326 inhibitor, the invading cell number significantly increased compared with cells transfected with shRPPH1 alone. In addition, E-cadherin expression levels were reduced, and vimentin was upregulated. MiR-326 overexpression partially reduced the resistance of A549 cells to CDDP induced by RPPH1 overexpression. WNT2B expression was directly suppressed using miR-326. A549 cells co-transfected with a miR-326 mimic and a WNT2B overexpression vector demonstrated increased invasion potential, reduced E-cadherin and increased vimentin protein expression levels, compared with cells transfected with the mimic alone. miR-326 overexpression reduced CDDP resistance in A549 cells. However, co-transfection with WNT2B partially enhanced CDDP resistance, compared with the mimic alone. In conclusion, RPPH1 promoted NSCLC progression and lung cancer cell resistance to CDDP through miR-326 and WNT2B.
Collapse
Affiliation(s)
- Yuying Wu
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Kewei Cheng
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Wenjun Liang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Xiaohua Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
20
|
Li L, Lin L, Veeraraghavan J, Hu Y, Wang X, Lee S, Tan Y, Schiff R, Wang XS. Therapeutic role of recurrent ESR1-CCDC170 gene fusions in breast cancer endocrine resistance. Breast Cancer Res 2020; 22:84. [PMID: 32771039 PMCID: PMC7414578 DOI: 10.1186/s13058-020-01325-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 07/27/2020] [Indexed: 01/07/2023] Open
Abstract
Background Endocrine therapy is the most common treatment for estrogen receptor (ER)-positive breast cancer, but its effectiveness is limited by high rates of primary and acquired resistance. There are likely many genetic causes, and recent studies suggest the important role of ESR1 mutations and fusions in endocrine resistance. Previously, we reported a recurrent ESR1 fusion called ESR1-CCDC170 in 6–8% of the luminal B breast cancers that has a worse clinical outcome after endocrine therapy. Despite being the most frequent ESR1 fusion, its functional role in endocrine resistance has not been studied in vivo, and the engaged mechanism and therapeutic relevance remain uncharacterized. Methods The endocrine sensitivities of HCC1428 or T47D breast cancer cells following genetic perturbations of ESR1-CCDC170 were assessed using clonogenic assays and/or xenograft mouse models. The underlying mechanisms were investigated by reverse phase protein array, western blotting, immunoprecipitation, and bimolecular fluorescence complementation assays. The sensitivity of ESR1-CCDC170 expressing breast cancer cells to concomitant treatments of tamoxifen and HER/SRC inhibitors was assessed by clonogenic assays. Results Our results suggested that different ESR1-CCDC170 fusions endow different levels of reduced endocrine sensitivity in vivo, resulting in significant survival disadvantages. Further investigation revealed a novel mechanism that ESR1-CCDC170 binds to HER2/HER3/SRC and activates SRC/PI3K/AKT signaling. Silencing of ESR1-CCDC170 in the fusion-positive cell line, HCC1428, downregulates HER2/HER3, represses pSRC/pAKT, and improves endocrine sensitivity. More important, breast cancer cells expressing ectopic or endogenous ESR1-CCDC170 are highly sensitive to treatment regimens combining endocrine agents with the HER2 inhibitor lapatinib and/or the SRC inhibitor dasatinib. Conclusion ESR1-CCDC170 may endow breast cancer cell survival under endocrine therapy via maintaining/activating HER2/HER3/SRC/AKT signaling which implies a potential therapeutic strategy for managing these fusion positive tumors.
Collapse
Affiliation(s)
- Li Li
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ling Lin
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yiheng Hu
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xian Wang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sanghoon Lee
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, 15206, USA
| | - Ying Tan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiao-Song Wang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA. .,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA. .,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA. .,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, 15206, USA.
| |
Collapse
|
21
|
Huang X, Li Y, Li X, Fan D, Xin HB, Fu M. TRIM14 promotes endothelial activation via activating NF-κB signaling pathway. J Mol Cell Biol 2020; 12:176-189. [PMID: 31070748 PMCID: PMC7181718 DOI: 10.1093/jmcb/mjz040] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 04/18/2019] [Accepted: 04/28/2019] [Indexed: 12/12/2022] Open
Abstract
Endothelial activation by proinflammatory cytokines is closely associated to the pathogenesis of atherosclerosis and other vascular diseases; however, the molecular mechanisms controlling endothelial activation are not fully understood. Here we identify TRIM14 as a new positive regulator of endothelial activation via activating NF-κB signal pathway. TRIM14 is highly expressed in human vascular endothelial cells (ECs) and markedly induced by inflammatory stimuli such as TNF-α, IL-1β, and LPS. Overexpression of TRIM14 significantly increased the expression of adhesion molecules such as VCAM-1, ICAM-1, E-selectin, and cytokines such as CCL2, IL-8, CXCL-1, and TNF-α in activated ECs and by which it facilitated monocyte adhesion to ECs. Conversely, knockdown of TRIM14 has opposite effect on endothelial activation. Upon TNF-α stimulation, TRIM14 is recruited to IKK complex via directly binding to NEMO and promotes the phosphorylation of IκBα and p65, which is dependent on its K63-linked ubiquitination. Meanwhile, p65 can directly bind to the promoter regions of human TRIM14 gene and control its mRNA transcription. Finally, TRIM14 protein level is significantly upregulated in mouse and human atheroma compared to normal arteries. Taken together, these results indicate that TRIM14-NF-κB forms a positive feedback loop to enhance EC activation and TRIM14 may be a potential therapeutic target for vascular inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Xuan Huang
- Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Yong Li
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Xiuzhen Li
- Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Hong-Bo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang 330031, China
| | - Mingui Fu
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| |
Collapse
|
22
|
Wang Y, Bonavida B. A New Linkage between the Tumor Suppressor RKIP and Autophagy: Targeted Therapeutics. Crit Rev Oncog 2019; 23:281-305. [PMID: 30311561 DOI: 10.1615/critrevoncog.2018027211] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The complexities of molecular signaling in cancer cells have been hypothesized to mediate cross-network alterations of oncogenic processes such as uncontrolled cell growth, proliferation, acquisition of epithelial-to-mesenchymal transition (EMT) markers, and resistance to cytotoxic therapies. The two biochemically exclusive processes/proteins examined in the present review are the metastasis suppressor Raf-1 kinase inhibitory protein (RKIP) and the cell-intrinsic system of macroautophagy (hereafter referred to as autophagy). RKIP is poorly expressed in human cancer tissues, and low expression levels are correlated with high incidence of tumor growth, metastasis, poor treatment efficacy, and poor prognoses in cancer patients. By comparison, autophagy is a conserved cytoprotective degradation pathway that has been shown to influence the acquisition of resistance to hypoxia and nutrient depletion as well as the regulation of chemo-immuno-resistance and apoptotic evasion. Evidently, a broad library of cancer-relevant studies exists for RKIP and autophagy, although reports of the interactions between pathways involving RKIP and autophagy have been relatively sparse. To circumvent this limitation, the coordinate regulatory and effector mechanisms were examined for both RKIP and autophagy. Here, we propose three putative pathways that demonstrate the inherent pleiotropism and relevance of RKIP and the microtubule-associated protein 1 light chain 3 (MAP1LC3, LC3) on cell growth, proliferation, senescence, and EMT, among the hallmarks of cancer. Our findings suggest that signaling modules involving p53, signal transducer and activator of transcription 3 (STAT3), nuclear factor-κB (NF-κB), and Snail highlight the novel roles for RKIP in the control of autophagy and vice versa. The suggested potential crosstalk mechanisms are new areas of research in which to further study RKIP and autophagy in cancer models. These should lead to novel prognostic motifs and will provide alternative therapeutic strategies for the treatment of unresponsive aggressive cancer types.
Collapse
Affiliation(s)
- Yuhao Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90025-1747
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747
| |
Collapse
|
23
|
Zhu H, Sun B, Shen Q. TNF-α induces apoptosis of human nucleus pulposus cells via activating the TRIM14/NF-κB signalling pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3004-3012. [PMID: 31322007 DOI: 10.1080/21691401.2019.1643733] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Hao Zhu
- Department of Orthopaedics, The Affiliated Shanghai General Hospital of Nanjing Medical University, Shanghai, China
- Department of Orthopaedics, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| | - Bao Sun
- Department of Orthopaedics, The Affiliated Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Qiang Shen
- Department of Orthopaedics, The Affiliated Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| |
Collapse
|
24
|
Somatic Alteration Burden Involving Non-Cancer Genes Predicts Prognosis in Early-Stage Non-Small Cell Lung Cancer. Cancers (Basel) 2019; 11:cancers11071009. [PMID: 31330989 PMCID: PMC6678704 DOI: 10.3390/cancers11071009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023] Open
Abstract
The burden of somatic mutations and neoantigens has been associated with improved survival in cancer treated with immunotherapies, especially non-small cell lung cancer (NSCLC). However, there is uncertainty about their effect on outcome in early-stage untreated cases. We posited that the burden of mutations in a specific set of genes may also contribute to the prognosis of early NSCLC patients. From a small cohort of 36 NSCLC cases, we were able to identify somatic mutations and copy number alterations in 865 genes that contributed to patient overall survival. Simply, the number of altered genes (NAG) among these 865 genes was associated with longer disease-free survival (hazard ratio (HR) = 0.153, p = 1.48 × 10-4). The gene expression signature distinguishing patients with high/low NAG was also prognostic in three independent datasets. Patients with a high NAG could be further stratified based on the presence of immunogenic mutations, revealing a further subgroup of stage I NSCLC with even better prognosis (85% with >5 years survival), and associated with cytotoxic T-cell expression. Importantly, 95% of the highly-altered genes lacked direct relation to cancer, but were implicated in pathways regulating cell proliferation, motility and immune response.
Collapse
|
25
|
Shen W, Jin Z, Tong X, Wang H, Zhuang L, Lu X, Wu S. TRIM14 promotes cell proliferation and inhibits apoptosis by suppressing PTEN in colorectal cancer. Cancer Manag Res 2019; 11:5725-5735. [PMID: 31296997 PMCID: PMC6598940 DOI: 10.2147/cmar.s210782] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
Background Colorectal cancer (CRC) is among the most frequent and lethal malignancies worldwide. Although great advances have been made in the treatment of CRC, prognosis remains poor. Our previous study indicated that tripartite motif-containing 14 (TRIM14) was upregulated in CRC samples. Methods In the current study, the association between TRIM14 and CRC was investigated. Protein expression was determined by Western blotting and immunohistochemistry. Further, the biological roles of TRIM14 in CRC cell proliferation and apoptosis were explored both in vitro and in vivo. Results We observed that increased TRIM14 expression in CRC tissues was closely related with aggressive clinicopathological characteristics and poor prognosis. TRIM14 knockdown markedly reduced proliferation and increased apoptosis in HT-29 and SW620 cells, whereas TRIM14 overexpression in LoVo cells displayed opposite results. Xenograft experiments using HT-29 cells confirmed suppression of tumor growth and induction of apoptosis upon TRIM14 knockdown in vivo. Furthermore, downregulation of TRIM14 inhibited the AKT pathway, as indicated by reduced levels of phosphorylated AKT, Bcl-2 and Cyclin D1, and elevated levels of phosphatase and
tensin homology (PTEN) and p27. In addition, TRIM14 colocalized with PTEN in the cytoplasm and induced PTEN ubiquitination. Moreover, PTEN overexpression significantly inhibited pro-proliferative effects of TRIM14, indicating an involvement of PTEN/AKT signaling in mediating TRIM14 functions. Conclusions The present data demonstrate that TRIM14 overexpression promotes CRC cell proliferation, suggesting TRIM14 as an attractive therapeutic target for CRC.
Collapse
Affiliation(s)
- Weidong Shen
- Department of Gastroenterology, Jiangyin Hospital Affiliated to Nantong University, Jiangyin, People's Republic of China
| | - Zhonghai Jin
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, Yiwu, People's Republic of China
| | - Xiuping Tong
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, Yiwu, People's Republic of China
| | - Haiying Wang
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, Yiwu, People's Republic of China
| | - Lilei Zhuang
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, Yiwu, People's Republic of China
| | - Xiaofeng Lu
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, Yiwu, People's Republic of China
| | - Shenbao Wu
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, Yiwu, People's Republic of China
| |
Collapse
|
26
|
Reactive oxygen species and cancer: A complex interaction. Cancer Lett 2019; 452:132-143. [PMID: 30905813 DOI: 10.1016/j.canlet.2019.03.020] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/21/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022]
Abstract
Elevated levels of Reactive Oxygen Species (ROS), increased antioxidant ability and the maintenance of redox homeostasis can cumulatively contribute to tumor progression and metastasis. The sources and the role of ROS in a heterogeneous tumor microenvironment can vary at different stages of tumor: initiation, development, and progression, thus making it a complex subject. In this review, we have summarized the sources of ROS generation in cancer cells, its role in the tumor microenvironment, the possible functions of ROS and its important scavenger systems in tumor progression with special emphasis on solid tumors.
Collapse
|
27
|
Feng S, Cai X, Li Y, Jian X, Zhang L, Li B. Tripartite motif-containing 14 (TRIM14) promotes epithelial-mesenchymal transition via ZEB2 in glioblastoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:57. [PMID: 30728039 PMCID: PMC6364431 DOI: 10.1186/s13046-019-1070-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/30/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Several members of the tripartite motif-containing (TRIM) protein family have been reported to serve as vital regulators of tumorigenesis. Recent studies have demonstrated an oncogenic role of TRIM 14 in multiple human cancers; however, the importance of this protein in glioblastoma remains to be elucidated. METHODS The expression levels of TRIM14 were analyzed in a series of database and were examined in a variety of glioblastoma cell lines. Two independent TRIM14 shRNA were transfected into LN229 and U251 cells, and the effect of TRIM14 depletion was confirmed. Transwell assay and wound healing assay assay were carried out to assess the effect of TRIM14 depletion on glioblastoma cell invasion and migration. Western blotting was performed to screen the downstream gene of TRIM14. The stability analysis and Ubiquitylation assays and Orthotopic xenograft studies were also performed to investigate the role of TRIM14 and the relationship with downstream gene. Human glioblastoma tissues were obtained and immunohistochemical staining were carried out to confirm the clinical significance of TRIM14. RESULTS In this study, we showed that TRIM14 was upregulated in human glioblastoma specimens and cell lines, and correlated with glioblastoma progression and shorter patient survival times. Functional experiments showed that decreased TRIM14 expression reduced glioblastoma cell invasion and migration. Furthermore, we identified that zinc finger E-box binding homeobox 2 (ZEB2), a transcription factor involved in epithelial-mesenchymal transition, is a downstream target of TRIM14. Further investigation revealed that TRIM14 inactivation significantly facilitated ZEB2 ubiquitination and proteasomal degradation, which led to aggressive invasion and migration. Our findings provide insight into the specific biological role of TRIM14 in tumor invasion. CONCLUSIONS Our findings provide insight into the specific biological role of TRIM14 in tumor invasion, and suggest that targeting the TRIM14/ZEB2 axis might be a novel therapeutic approach for blocking glioblastoma.
Collapse
Affiliation(s)
- Shuang Feng
- Department of Encephalopathy, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaomin Cai
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yangyang Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoguang Jian
- Department of Encephalopathy, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Linxin Zhang
- Department of Encephalopathy, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Bin Li
- Department of Encephalopathy, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
28
|
Jin Z, Li H, Hong X, Ying G, Lu X, Zhuang L, Wu S. TRIM14 promotes colorectal cancer cell migration and invasion through the SPHK1/STAT3 pathway. Cancer Cell Int 2018; 18:202. [PMID: 30555277 PMCID: PMC6288942 DOI: 10.1186/s12935-018-0701-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most lethal malignancies. Tripartite Motif Containing 14 (TRIM14) is a member of TRIM family proteins, which are involved in the pathogenesis of various cancers. This study aimed to investigate TRIM14 expression in CRC tissues, and its effects on the migration and invasion of CRC cell lines. Methods TRIM14 mRNA expression was detected by real-time PCR analysis. Cell migration and invasion were measured by Transwell assays. Protein expression was assessed by western blot analysis. Results The expression of TRIM14 was significantly higher in CRC tissues than in matched non-cancerous tissues. TRIM14 knockdown by specific short hairpin RNA (shRNA) attenuated CRC cell migration and invasion, whereas TRIM14 overexpression caused reverse effect. Moreover, TRIM14 positively regulated the protein levels of sphingosine kinase 1 (SPHK1) and phosphorylated STAT3 (p-STAT3), as well as the mRNA and protein expression of matrix metalloproteinase 2, MMP9 and vascular endothelial growth factor, which are transcriptional targets of the STAT3 signaling pathway. Importantly, the blockage of the SPHK1/STAT3 signaling pathway by SKI-II or AG490 could reverse the TRIM14-promoted CRC cell migration and invasion. Conclusions Our results reveal a critical role for TRIM14 in promoting migration and invasion of CRC cells, and suggest TRIM14 may serve as a potential molecular target to prevent CRC metastasis. Electronic supplementary material The online version of this article (10.1186/s12935-018-0701-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhonghai Jin
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, 699 Jiangdong Middle Road, Yiwu, 322000 China
| | - Hongguang Li
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, 699 Jiangdong Middle Road, Yiwu, 322000 China
| | - Xiaofei Hong
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, 699 Jiangdong Middle Road, Yiwu, 322000 China
| | - Guangrong Ying
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, 699 Jiangdong Middle Road, Yiwu, 322000 China
| | - Xiaofeng Lu
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, 699 Jiangdong Middle Road, Yiwu, 322000 China
| | - Lilei Zhuang
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, 699 Jiangdong Middle Road, Yiwu, 322000 China
| | - Shenbao Wu
- Department of Gastroenterology, Yiwu Hospital, Wenzhou Medical University, 699 Jiangdong Middle Road, Yiwu, 322000 China
| |
Collapse
|
29
|
Wang F, Ruan L, Yang J, Zhao Q, Wei W. TRIM14 promotes the migration and invasion of gastric cancer by regulating epithelial‑to‑mesenchymal transition via activation of AKT signaling regulated by miR‑195‑5p. Oncol Rep 2018; 40:3273-3284. [PMID: 30272351 PMCID: PMC6196628 DOI: 10.3892/or.2018.6750] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/21/2018] [Indexed: 02/07/2023] Open
Abstract
Tripartite motif-containing 14 (TRIM14) is a member of the TRIM protein family which has been implicated in several critical processes and is dysregulated in human cancers in a cancer-specific trend. However, its expression and function in human gastric cancer (GC) are still largely unknown. In this study, we confirmed for the first time that TRIM14 mRNA and protein were upregulated in GC tissues and cell lines as determined by qRT-PCR and western blot analysis. Clinical data disclosed that high TRIM14 expression was significantly associated with aggressive prognostic features, including advanced TNM stage and lymph node metastasis. In regards to 5-year survival, TRIM14 served as a potential prognostic marker for GC. Notably, TRIM14 promoted migration, invasion as measured by Transwell and epithelial-to-mesenchymal transition (EMT) as determined by western blot analysis and immunofluorescence (IF) in vitro and in vivo. Moreover, TRIM14 induced protein kinase B (AKT) pathway activation, and inhibition of AKT reversed the TRIM14-induced promotive effects on cell migration, invasion and EMT progression. Furthermore, we demonstrated that TRIM14 expression was regulated by miR-195-5p. miR-195-5p exerted an inhibitory role in GC migration and invasion. Finally, we confirmed that alteration of TRIM14 expression abolished the effects of miR-195-5p on GC cells. Conclusively, our results demonstrated that TRIM14 functions as an oncogene in regulating EMT and metastasis of GC via activating AKT signaling, which was regulated by miR-195-5p, supporting its potential utility as a therapeutic target for GC.
Collapse
Affiliation(s)
- Feiqian Wang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Litao Ruan
- Department of Ultrasound Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jinru Yang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qiaoling Zhao
- Department of Ultrasound Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wei Wei
- Department of Ultrasound Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
30
|
Expression of the human TRIM14 and its mutant form (P207L) promotes apoptosis in transgenic loaches. Mol Biol Rep 2018; 45:2087-2093. [PMID: 30203243 DOI: 10.1007/s11033-018-4365-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022]
Abstract
The tripartite-motif (TRIM)14 protein, one of the TRIM family members, was shown to participate in the antiviral and antibacterial defence. Besides, it appears to play an essential role in the processes of oncogenesis. In some types of human tumour cells, TRIM14 has been shown to inhibit apoptosis, while in others-the overexpression of TRIM14 promotes apoptosis. However, whether TRIM14 mediates apoptosis in the normal cells remains unknown. In the present study, we investigated the possible participation of the human TRIM14 gene and its mutant form (620C > T) in the induction of apoptosis in the transgenic larvae loach Misgurnus fossilis L. We observed that the expression of both forms of TRIM14 gene was accompanied by the increase of the frequency of pyknotic nuclei in fish embryos compared to control groups. Accordingly, using the TUNEL assay, the enhanced apoptosis was revealed upon expression of both forms of TRIM14 gene. The transcription of proapoptotic genes (bax, tp53, and casp9) was significantly increased in transgenic loaches expressing human wild-type TRIM14, but remained unchanged upon expression of its mutant form. In addition, the transcription of c-myc was upregulated in transgenic loaches expressing both forms. Thus, it can be assumed that during embryonic development TRIM14 has a proapoptotic effect on the cells via the activation of c-myc, tp53, and bax genes. Apparently, the mutant TRIM14 directs apoptosis via c-myc by p53-independent mechanism.
Collapse
|
31
|
TRIM14 promotes chemoresistance in gliomas by activating Wnt/β-catenin signaling via stabilizing Dvl2. Oncogene 2018; 37:5403-5415. [DOI: 10.1038/s41388-018-0344-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/29/2018] [Accepted: 05/11/2018] [Indexed: 01/16/2023]
|
32
|
Liu S, Li S, Hai J, Wang X, Chen T, Quinn MM, Gao P, Zhang Y, Ji H, Cross DAE, Wong KK. Targeting HER2 Aberrations in Non-Small Cell Lung Cancer with Osimertinib. Clin Cancer Res 2018; 24:2594-2604. [PMID: 29298799 DOI: 10.1158/1078-0432.ccr-17-1875] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/27/2017] [Accepted: 12/29/2017] [Indexed: 12/13/2022]
Abstract
Purpose:HER2 (or ERBB2) aberrations, including both amplification and mutations, have been classified as oncogenic drivers that contribute to 2% to 6% of lung adenocarcinomas. HER2 amplification is also an important mechanism for acquired resistance to EGFR tyrosine kinase inhibitors (TKI). However, due to limited preclinical studies and clinical trials, currently there is still no available standard of care for lung cancer patients with HER2 aberrations. To fulfill the clinical need for targeting HER2 in patients with non-small cell lung cancer (NSCLC), we performed a comprehensive preclinical study to evaluate the efficacy of a third-generation TKI, osimertinib (AZD9291).Experimental Design: Three genetically modified mouse models (GEMM) mimicking individual HER2 alterations in NSCLC were generated, and osimertinib was tested for its efficacy against these HER2 aberrations in vivoResults: Osimertinib treatment showed robust efficacy in HER2wt overexpression and EGFR del19/HER2 models, but not in HER2 exon 20 insertion tumors. Interestingly, we further identified that combined treatment with osimertinib and the BET inhibitor JQ1 significantly increased the response rate in HER2-mutant NSCLC, whereas JQ1 single treatment did not show efficacy.Conclusions: Overall, our data indicated robust antitumor efficacy of osimertinib against multiple HER2 aberrations in lung cancer, either as a single agent or in combination with JQ1. Our study provides a strong rationale for future clinical trials using osimertinib either alone or in combination with epigenetic drugs to target aberrant HER2 in patients with NSCLC. Clin Cancer Res; 24(11); 2594-604. ©2018 AACRSee related commentary by Cappuzzo and Landi, p. 2470.
Collapse
Affiliation(s)
- Shengwu Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Shuai Li
- Department of Pathology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Josephine Hai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Xiaoen Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ting Chen
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Max M Quinn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Peng Gao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yanxi Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Darren A E Cross
- AstraZeneca Oncology Innovative Medicines, Alderley Park, Macclesfield, Cheshire, United Kingdom
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York.
| |
Collapse
|
33
|
Hai J, Liu S, Bufe L, Do K, Chen T, Wang X, Ng C, Li S, Tsao MS, Shapiro GI, Wong KK. Synergy of WEE1 and mTOR Inhibition in Mutant KRAS-Driven Lung Cancers. Clin Cancer Res 2017; 23:6993-7005. [PMID: 28821559 DOI: 10.1158/1078-0432.ccr-17-1098] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/06/2017] [Accepted: 08/10/2017] [Indexed: 12/21/2022]
Abstract
Purpose:KRAS-activating mutations are the most common oncogenic driver in non-small cell lung cancer (NSCLC), but efforts to directly target mutant KRAS have proved a formidable challenge. Therefore, multitargeted therapy may offer a plausible strategy to effectively treat KRAS-driven NSCLCs. Here, we evaluate the efficacy and mechanistic rationale for combining mTOR and WEE1 inhibition as a potential therapy for lung cancers harboring KRAS mutations.Experimental Design: We investigated the synergistic effect of combining mTOR and WEE1 inhibitors on cell viability, apoptosis, and DNA damage repair response using a panel of human KRAS-mutant and wild type NSCLC cell lines and patient-derived xenograft cell lines. Murine autochthonous and human transplant models were used to test the therapeutic efficacy and pharmacodynamic effects of dual treatment.Results: We demonstrate that combined inhibition of mTOR and WEE1 induced potent synergistic cytotoxic effects selectively in KRAS-mutant NSCLC cell lines, delayed human tumor xenograft growth and caused tumor regression in a murine lung adenocarcinoma model. Mechanistically, we show that inhibition of mTOR potentiates WEE1 inhibition by abrogating compensatory activation of DNA repair, exacerbating DNA damage in KRAS-mutant NSCLC, and that this effect is due in part to reduction in cyclin D1.Conclusions: These findings demonstrate that compromised DNA repair underlies the observed potent synergy of WEE1 and mTOR inhibition and support clinical evaluation of this dual therapy for patients with KRAS-mutant lung cancers. Clin Cancer Res; 23(22); 6993-7005. ©2017 AACR.
Collapse
Affiliation(s)
- Josephine Hai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Shengwu Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Lauren Bufe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Khanh Do
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ting Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Xiaoen Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Christine Ng
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Shuai Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kwok-Kin Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| |
Collapse
|