1
|
Chen X, Wu W, Jeong JH, Rokavec M, Wei R, Feng S, Schroth W, Brauch H, Zhong S, Luo JL. Cytokines-activated nuclear IKKα-FAT10 pathway induces breast cancer tamoxifen-resistance. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1413-1426. [PMID: 38565741 DOI: 10.1007/s11427-023-2460-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/26/2023] [Indexed: 04/04/2024]
Abstract
Endocrine therapy that blocks estrogen signaling is the most effective treatment for patients with estrogen receptor positive (ER+) breast cancer. However, the efficacy of agents such as tamoxifen (Tam) is often compromised by the development of resistance. Here we report that cytokines-activated nuclear IKKα confers Tam resistance to ER+ breast cancer by inducing the expression of FAT10, and that the expression of FAT10 and nuclear IKKα in primary ER+ human breast cancer was correlated with lymphotoxin β (LTB) expression and significantly associated with relapse and metastasis in patients treated with adjuvant mono-Tam. IKKα activation or enforced FAT10 expression promotes Tam-resistance while loss of IKKα or FAT10 augments Tam sensitivity. The induction of FAT10 by IKKα is mediated by the transcription factor Pax5, and coordinated via an IKKα-p53-miR-23a circuit in which activation of IKKα attenuates p53-directed repression of FAT10. Thus, our findings establish IKKα-to-FAT10 pathway as a new therapeutic target for the treatment of Tam-resistant ER+ breast cancer.
Collapse
Affiliation(s)
- Xueyan Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Weilin Wu
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Ji-Hak Jeong
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Matjaz Rokavec
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Rui Wei
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shaolong Feng
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA
| | - Werner Schroth
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, 70376, Germany
- iFIT Cluster of Excellence, University of Tübingen, Tübingen, 72074, Germany
| | - Hiltrud Brauch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, 70376, Germany
- iFIT Cluster of Excellence, University of Tübingen, Tübingen, 72074, Germany
| | - Shangwei Zhong
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA.
- The Cancer Research Institute and the Second Affiliated Hospital, Henyang Medical School, University of South China, Hengyang, 421001, China.
| | - Jun-Li Luo
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, 33458, USA.
- The Cancer Research Institute and the Second Affiliated Hospital, Henyang Medical School, University of South China, Hengyang, 421001, China.
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China.
| |
Collapse
|
2
|
Targeting Inflammatory Signaling in Prostate Cancer Castration Resistance. J Clin Med 2021; 10:jcm10215000. [PMID: 34768524 PMCID: PMC8584457 DOI: 10.3390/jcm10215000] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/04/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
Although castration-resistant prostate cancer (CRPC) as a whole, by its name, refers to the tumors that relapse and/or regrow independently of androgen after androgen deprivation therapy (ADT), untreated tumor, even in early-stage primary prostate cancer (PCa), contains androgen-independent (AI) PCa cells. The transformation of androgen-dependent (AD) PCa to AI PCa under ADT is a forced evolutionary process, in which the small group of AI PCa cells that exist in primary tumors has the unique opportunity to proliferate and expand selectively and dominantly, while some AD PCa cells that have escaped from ADT-induced death acquire the capability to survive in an androgen-depleted environment. The adaptation and reprogramming of both PCa cells and the tumor microenvironment (TME) under ADT make PCa much stronger than primary tumors so that, currently, there are no effective therapeutic methods available for the treatment of CRPC. Many mechanisms have been found to be related to the emergence and maintenance of PCa castration resistance; in this review, we focus on the role of inflammatory signaling in both PCa cells and the TME for the emergence and maintenance of CRPC and summarize the recent advances of therapeutic strategies that target inflammatory signaling for the treatment of CRPC.
Collapse
|
3
|
Wang X, Teng F, Lu J, Mu D, Zhang J, Yu J. Expression and prognostic role of IKBKE and TBK1 in stage I non-small cell lung cancer. Cancer Manag Res 2019; 11:6593-6602. [PMID: 31406474 PMCID: PMC6642623 DOI: 10.2147/cmar.s204924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/27/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The inhibitors of nuclear factor kappa-B kinase subunit epsilon (IKBKE) and TANK-binding kinase 1 (TBK1) are important members of the nonclassical IKK family that share the kinase domain. They are important oncogenes for activation of several signaling pathways in several tumors. This study aims to explore the expression of IKBKE and TBK1 and their prognostic role in stage I non-small cell lung cancer (NSCLC). PATIENTS AND METHODS A total of 142 surgically resected stage I NSCLC patients were enrolled and immunohistochemistry of IKBKE and TBK1 was performed. RESULTS IKBKE and TBK1 were expressed in 121 (85.2%) and 114 (80.3%) of stage I NSCLC patients respectively. IKBKE expression was significantly associated with TBK1 expression (P=0.004). Furthermore, multivariate regression analyses showed there was a significant relationship between patients with risk factors, the recurrence pattern of metastasis and IKBKE+/TBK1+ co-expression (P=0.032 and P=0.022, respectively). In Kaplan-Meier survival curve analyses, the IKBKE+/TBK1+ co-expression subgroup was significantly associated with poor overall survival (P=0.014). CONCLUSIONS This is the first study to investigate the relationship between IKBKE and TBK1 expression and clinicopathologic characteristics in stage I NSCLC patients. IKBKE+/TBK1+ co-expression was significantly obvious in patients with risk factors and with recurrence pattern of distant metastasis. Furthermore, IKBKE+/TBK1+ is also an effective prognostic predictor for poor overall survival.
Collapse
Affiliation(s)
- Xin Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei430060, People’s Republic of China
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong250117, People’s Republic of China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong250117, People’s Republic of China
| | - Jie Lu
- Department of Neurosurgery, Shandong Province Qianfoshan Hospital of Shandong University, Jinan, Shandong250014, People’s Republic of China
| | - Dianbin Mu
- Department of Pathology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong250117, People’s Republic of China
| | - Jianbo Zhang
- Department of Pathology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong250117, People’s Republic of China
| | - Jinming Yu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei430060, People’s Republic of China
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong250117, People’s Republic of China
| |
Collapse
|
4
|
Wang W, Wang Z, Chen S, Zang X, Miao J. Interleukin-1β/nuclear factor-κB signaling promotes osteosarcoma cell growth through the microRNA-181b/phosphatase and tensin homolog axis. J Cell Biochem 2018; 120:1763-1772. [PMID: 30977354 DOI: 10.1002/jcb.27477] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/19/2018] [Indexed: 02/06/2023]
Abstract
So far, microRNA has attracted plenty of interest due to its role in tumorigenesis. Reportedly, miR-181b may be involved in the tumorigenesis of osteosarcoma (OS). In the current study, we attempted to investigate the detailed function and mechanism of miR-181b in OS carcinogenesis. Herein, miR-181a, miR-181b, miR-181c, and miR-181d expressions in OS tissues were higher than that in nontumor tissue samples as examined real-time polymerase chain reaction. Via direct targeting, miR-181b negatively regulated the expression of phosphatase and tensin homolog (PTEN), a well-known tumor suppressor. Furthermore, a small interfering RNA strategy was used to find that interleukin (IL)-1B and nuclear factor-κB (NF-κB) regulate miR-181b and PTEN expression. Consequently, the repression of PTEN by miR-181b promotes OS cell proliferation. In summary, our data support a critical role for NF-κB-dependent upregulation of miR-181b, which further inhibited PTEN expression and promoted the cell proliferation of OS cell lines. The above findings represent a new pathway for the repression of PTEN and the promotion of cell proliferation upon IL-1β induction.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhengguang Wang
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shijie Chen
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaofang Zang
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jinglei Miao
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
5
|
Wang H, Zhao S, Chen B, Fu C, Dang Y, Fang P, Wang J, Wang N, Liu L. Repression of the expression of PPP3CC by ZEB1 confers activation of NF-κB and contributes to invasion and growth in glioma cells. Jpn J Clin Oncol 2018; 48:175-183. [PMID: 29294030 DOI: 10.1093/jjco/hyx182] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/11/2017] [Indexed: 11/12/2022] Open
Abstract
Background Gliomas are highly malignant brain tumors. Aberrant activation of NF-κB plays a crucial role in tumor progression. Method ELISA assay was used to detect NF-κB activity in glimoas cells with different treatments. PPP3CC expression was evaluated by qRT-PCR and western blot assay. Kaplan-Meier analysis estimated the overall survival rates according to the protein level of PPP3CC. Transwell and MTS assay were performed to determine cell invasion and growth. Chromatin immunoprecipitation combined with luciferase reporter assays illustrated the transcriptional regulation of PPP3CC. Results We showed that PPP3CC decrease was responsible for constitutive activation of NF-κB in gliomas. Restored PPP3CC expression inhibited activation of NF-κB. PPP3CC was frequently decreased in gliomas and that repression of the expression of PPP3CC correlated glioma progression. The ectopic expression of PPP3CC inhibited the invasive potential of glioma cells, and inhibited glioma cells proliferation in vitro and growth in vivo. Additionally, the expression of Zinc finger E-box-binding homeobox 1(ZEB1) was increased in gliomas and was negatively correlated with clinical outcomes of glioma patients. ZEB1 inversely correlated with the expression of PPP3CC. ZEB1 was also confirmed to physically bind to the PPP3CC promoter. ZEB1 knockdown resulted in an increase in the expression of PPP3CC and elevation of PPP3CC promoter activity in glioma cells. Conclusion These findings indicated that the down-regulation of PPP3CC by ZEB1 resulted in activation of NF-κB is a critical oncogenic event in gliomas.
Collapse
Affiliation(s)
- Hongquan Wang
- Department of Neurosurgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine
| | - Shuli Zhao
- Department of Pharmacy, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Hubei,PR China
| | - Bo Chen
- Department of Neurosurgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine
| | - Chuhua Fu
- Department of Neurosurgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine
| | - Yanwei Dang
- Department of Neurosurgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine
| | - Peihai Fang
- Department of Neurosurgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine
| | - Jun Wang
- Department of Neurosurgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine
| | - Ning Wang
- Department of Neurosurgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine
| | - Lijun Liu
- Department of Neurosurgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine
| |
Collapse
|
6
|
Jeong JH, Park SJ, Dickinson SI, Luo JL. A Constitutive Intrinsic Inflammatory Signaling Circuit Composed of miR-196b, Meis2, PPP3CC, and p65 Drives Prostate Cancer Castration Resistance. Mol Cell 2016; 65:154-167. [PMID: 28041912 DOI: 10.1016/j.molcel.2016.11.034] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/26/2016] [Accepted: 11/22/2016] [Indexed: 10/20/2022]
Abstract
Androgen deprivation therapy is the most effective treatment for advanced prostate cancer, but almost all cancer eventually becomes castration resistant, and the underlying mechanisms are largely unknown. Here, we show that an intrinsic constitutively activated feedforward signaling circuit composed of IκBα/NF-κB(p65), miR-196b-3p, Meis2, and PPP3CC is formed during the emergence of castration-resistant prostate cancer (CRPC). This circuit controls the expression of stem cell transcription factors that drives the high tumorigenicity of CRPC cells. Interrupting the circuit by targeting its individual components significantly impairs the tumorigenicity and CRPC development. Notably, constitutive activation of IκBα/NF-κB(p65) in this circuit is not dependent on the activation of traditional IKKβ/NF-κB pathways that are important in normal immune responses. Therefore, our studies present deep insight into the bona fide mechanisms underlying castration resistance and provide the foundation for the development of CRPC therapeutic strategies that would be highly efficient while avoiding indiscriminate IKK/NF-κB inhibition in normal cells.
Collapse
Affiliation(s)
- Ji-Hak Jeong
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Sun-Jin Park
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | | | - Jun-Li Luo
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
7
|
Li J, Huang J, Jeong JH, Park SJ, Wei R, Peng J, Luo Z, Chen YT, Feng Y, Luo JL. Selective TBK1/IKKi dual inhibitors with anticancer potency. Int J Cancer 2013; 134:1972-80. [PMID: 24150799 DOI: 10.1002/ijc.28507] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 08/29/2013] [Accepted: 09/16/2013] [Indexed: 12/25/2022]
Abstract
Increasing evidence suggests that the noncanonical IKKs play critical roles in tumor genesis and development, leading to the notion that noncanonical IKKs may be good targets for cancer therapy. Here, we demonstrate that although TBK1 is not overexpressed or constitutively activated in some tumor cells, targeting IKKi induces the activation of TBK1. Therefore, simultaneously targeting both kinases is necessary to efficiently suppress tumor cell proliferation. We show that three TBK1/IKKi dual inhibitors, which are based on a structurally rigid 2-amino-4-(3'-cyano-4'-pyrrolidine)phenyl-pyrimidine scaffold, potently inhibit cell viability in human breast, prostate and oral cancer cell lines. Treatment with these TBK1/IKKi dual inhibitors significantly impairs tumor development in xenograft and allograft mouse models. The anticancer function of these inhibitors may be partially due to their suppression of TBK1/IKKi-mediated AKT phosphorylation and VEGF expression. Most importantly, these TBK1/IKKi dual inhibitors have drug-like properties including low molecular weight, low cytochrome P450 inhibition and high metabolic stability. Therefore, our studies provide proof of concept for further drug discovery efforts that may lead to novel strategies and new therapeutics for the treatment of human cancer.
Collapse
Affiliation(s)
- Jijia Li
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL
| | | | | | | | | | | | | | | | | | | |
Collapse
|