1
|
Wang B, Zhang F, Wu X, Ji M. TBK1 is paradoxical in tumor development: a focus on the pathway mediating IFN-I expression. Front Immunol 2024; 15:1433321. [PMID: 39161768 PMCID: PMC11330819 DOI: 10.3389/fimmu.2024.1433321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
TANK-binding kinase 1 (TBK1) is a member of the IKK family and plays a crucial role in the activation of non-canonical NF-κB signaling and type I interferon responses. The aberrant activation of TBK1 contributes to the proliferation and survival of various types of tumor cells, particularly in specific mutational or tumorous contexts. Inhibitors targeting TBK1 are under development and application in both in vivo and in vitro settings, yet their clinical efficacy remains limited. Numerous literatures have shown that TBK1 can exhibit both tumor promoting and tumor inhibiting effects. TBK1 acts as a pivotal node within the innate immune pathway, mediating anti-tumor immunity through the activation of innate immune responses. Facilitating interferon-I (IFN-I) production represents a critical mechanism through which TBK1 bridges these processes. IFN has been shown to exert both beneficial and detrimental effects on tumor progression. Hence, the paradoxical role of TBK1 in tumor development may necessitate acknowledgment in light of its downstream IFN-I signaling cascade. In this paper, we review the signaling pathways mediated by TBK1 in various tumor contexts and summarize the dual roles of TBK1 and the TBK1-IFN pathways in both promoting and inhibiting tumor progression. Additionally, we highlight the significance of the TBK1-IFN pathway in clinical therapy, particularly in the context of immune response. We anticipate further advancements in the development of TBK1 inhibitors as part of novel cancer treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Mei Ji
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
2
|
Miranda A, Shirley CA, Jenkins RW. Emerging roles of TBK1 in cancer immunobiology. Trends Cancer 2024; 10:531-540. [PMID: 38519366 PMCID: PMC11168882 DOI: 10.1016/j.trecan.2024.02.007] [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: 12/29/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/24/2024]
Abstract
TANK-binding kinase 1 (TBK1) is a versatile serine/threonine protein kinase with established roles in innate immunity, metabolism, autophagy, cell death, and inflammation. While best known for its role in regulating innate immunity, TBK1 has emerged as a cancer cell-intrinsic immune evasion gene by virtue of its role in modulating cellular responses to inflammatory signals emanating from the immune system. Beyond its effect on cancer cells, TBK1 appears to regulate lymphoid and myeloid cells in the tumor immune microenvironment. In this review, we detail recent advances in our understanding of the tumor-intrinsic and -extrinsic roles and regulation of TBK1 in tumor immunity.
Collapse
Affiliation(s)
- Alex Miranda
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carl A Shirley
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Russell W Jenkins
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Tian X, Zhang Z, Ding M. TXLNA enhances TBK1 phosphorylation by suppressing PPM1B recruitment. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119550. [PMID: 37506885 DOI: 10.1016/j.bbamcr.2023.119550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
In recent years, there has been a notable increase in cancer incidence and mortality, and immune abnormalities have been closely linked to malignancy development. TANK-binding kinase 1 (TBK1) is a non-classical IκB kinase that regulates interferon and NF-κB signaling pathways and plays a crucial role in innate immunity. Recent studies have shown high expression levels of TBK1 and increased activity in various tumor cells, suggesting its involvement in the development and progression of multiple cancers. Targeting TBK1 for tumor therapy may be a possibility. However, little is known about the abnormal activation and dynamic regulation of TBK1 in cancer. First, we utilized the BioID biotinylation technique combined with TMT-based quantitative proteomics to analyze the TBK1 interacting proteins. Our results revealed that TXLNA interacts with TBK1 and binds to the α-helical scaffold of TBK1. The expression of TXLNA could affect the S172 phosphorylation of TBK1. PPM1B is a phosphatase that can dephosphorylate TBK1 S172, so we used the APEX2 proximity labeling technique combined with TMT-based quantitative proteomics to explore the interacting proteins of PPM1B and search for the regulatory pathway of TXLNA on TBK1 phosphorylation. We found that PPM1B interacts with TXLNA. Based on these results, we further found that TXLNA impairs the binding of PPM1B to TBK1, inhibiting the dephosphorylation of TBK1 and contributing to the abnormal enhancement of TBK1 activity in cancer cells. This study sheds light on the potential mechanism of aberrant activation and dynamic regulation of TBK1 in tumors and provides a potential target for tumor therapy.
Collapse
Affiliation(s)
- Xiao Tian
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211199, China
| | - Zhiyuan Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211199, China
| | - Ming Ding
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211199, China.
| |
Collapse
|
4
|
Jiang Q, Guan Y, Zheng J, Lu H. TBK1 promotes thyroid cancer progress by activating the PI3K/Akt/mTOR signaling pathway. Immun Inflamm Dis 2023; 11:e796. [PMID: 36988258 PMCID: PMC10013413 DOI: 10.1002/iid3.796] [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: 09/15/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 03/17/2023] Open
Abstract
INTRODUCTION Thyroid cancer has received increasing attention; however, its detailed pathogenesis and pathological processes remain unclear. We investigated the role of TANK-binding kinase 1 (TBK1) in the progression of thyroid cancer. METHODS The expression of TBK1 in thyroid cancer and normal control tissues was analyzed using real-time quantitative polymerase chain reaction. The function of TBK1 on thyroid cancer cells was detected using MTT, colony formation, wound healing, and Transwell assays. The xenograft assay was carried out to check on the role of TBK1 in thyroid cancer. RESULTS TBK1 was highly expressed in thyroid tumors. High expression of TBK1 raised viability, proliferation, migration, and invasion of thyroid cancer cells. Gene set enrichment analysis revealed that TBK1 activated the phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin pathway. In addition, Myc-associated zinc finger protein (MAZ) was overexpressed in thyroid cancer and transcriptionally activated BK1. MAZ silence reversed the effects of TBK1 overexpression on thyroid cancer progression. Cotransfection with MAZ small-interfering RNA(siRNA) and TBK1 siRNA did not strengthen the inhibitory effect of TBK1 silencing on the thyroid cancer cells. The xenograft tumor assay showed that TBK1 short hairpinRNA inhibited tumor growth. CONCLUSION MAZ silencing inhibited tumor progress of thyroid cancer cells, whereas this inhibitory effect was reversed by TBK1 overexpression.
Collapse
Affiliation(s)
- Qiuli Jiang
- Department of Pathology, Xiamen Branch, Zhongshan HospitalFudan UniversityXiamenFujianP. R. China
| | - Yingying Guan
- Department of Pathology, Xiamen Branch, Zhongshan HospitalFudan UniversityXiamenFujianP. R. China
| | - Jingmei Zheng
- Department of Pathology, Xiamen Branch, Zhongshan HospitalFudan UniversityXiamenFujianP. R. China
| | - Huadong Lu
- Department of Pathology, Xiamen Branch, Zhongshan HospitalFudan UniversityXiamenFujianP. R. China
| |
Collapse
|
5
|
Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat Rev Immunol 2023; 23:90-105. [PMID: 35637393 DOI: 10.1038/s41577-022-00732-1] [Citation(s) in RCA: 208] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 02/04/2023]
Abstract
Great strides have been made in recent years towards understanding the roles of natural killer (NK) cells in immunity to tumours and viruses. NK cells are cytotoxic innate lymphoid cells that produce inflammatory cytokines and chemokines. By lysing transformed or infected cells, they limit tumour growth and viral infections. Whereas T cells recognize peptides presented by MHC molecules, NK cells display receptors that recognize stress-induced autologous proteins on cancer cells. At the same time, their functional activity is inhibited by MHC molecules displayed on such cells. The enormous potential of NK cells for immunotherapy for cancer is illustrated by their broad recognition of stressed cells regardless of neoantigen presentation, and enhanced activity against tumours that have lost expression of MHC class I owing to acquired resistance mechanisms. As a result, many efforts are under way to mobilize endogenous NK cells with therapeutics, or to provide populations of ex vivo-expanded NK cells as a cellular therapy, in some cases by equipping the NK cells with chimeric antigen receptors. Here we consider the key features that underlie why NK cells are emerging as important new additions to the cancer therapeutic arsenal.
Collapse
|
6
|
Liu H, Li H, Zhang J, Meng Q, Ma L. Correlation of TBK1, AR, and other serum cancer-related biomarkers in breast cancer patients: An observational study. Medicine (Baltimore) 2022; 101:e29996. [PMID: 35984205 PMCID: PMC9387973 DOI: 10.1097/md.0000000000029996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Breast cancer (BC) ranks first for incidence and mortality in gynecological malignant tumors. This study aims to investigate the diagnostic value of Tank-binding kinase 1 (TBK1) and its correlation with androgen receptor (AR) and other serum cancer-related biomarkers in BC patient. The present observational study included 451 female BC patients and 451 healthy controls. Serum levels of TBK1, AR and other cancer-related biomarkers were detected in all the patients and healthy controls. Patients' demographic data and clinical data including age, body mass index (BMI), tumor node Metastasis (TNM), pathological type, tumor size and lymph node metastasis were collected. The follow-up lasted for 5 years. The deceased group had higher rate of patients with TNM III~IV, lymph node metastasis or tumor diameter >2. Deceased group had much higher rate of patients with negative ER and positive Ki67. Besides, increased TBK1 was found in BC patients with positive correlation with AR, CA15-3, CA125, CEA, and CA19-9. Serum TBK1 was associated with the clinic outcomes of BC patients and those with high TBK1 had lower 5-year survival rate. Moreover, cutoff value of 13.95 ng/mL TBK1 showed AUC of 0.981 (93.6% for sensitivity and 86.3% for specificity) for diagnosing BC, and cutoff value of 22.65 ng/mL TBK1 had AUC of 0.996 (97.7% for sensitivity and 96.3% for specificity) for diagnosing the death of BC patients. Serum TBK1 was positively correlated with AR and other serum cancer-related biomarkers. In addition, high TBK1 predicted the poor prognosis and might be used for the diagnosis of BC.
Collapse
Affiliation(s)
- HanCheng Liu
- Department of Breast Surgery, Affiliated Hospital of ChengDe Medical College, Chengde City, Hebei, 067000, China
| | - HuiMing Li
- Department of Breast Surgery, Affiliated Hospital of ChengDe Medical College, Chengde City, Hebei, 067000, China
| | - Jie Zhang
- Department of Breast Surgery, Affiliated Hospital of ChengDe Medical College, Chengde City, Hebei, 067000, China
| | - QingLai Meng
- Department of Breast Surgery, Affiliated Hospital of ChengDe Medical College, Chengde City, Hebei, 067000, China
| | - LiHui Ma
- Department of Breast Surgery, Affiliated Hospital of ChengDe Medical College, Chengde City, Hebei, 067000, China
- * Correspondence: LiHui Ma, Department of Breast Surgery, Affiliated Hospital of ChengDe Medical College, Feng Yingzi town, Shuangqiao, Chengde City, Hebei, 067000, China (e-mail: )
| |
Collapse
|
7
|
BACH1 Expression Is Promoted by Tank Binding Kinase 1 (TBK1) in Pancreatic Cancer Cells to Increase Iron and Reduce the Expression of E-Cadherin. Antioxidants (Basel) 2022; 11:antiox11081460. [PMID: 36009179 PMCID: PMC9405201 DOI: 10.3390/antiox11081460] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 02/01/2023] Open
Abstract
BTB and CNC homology 1 (BACH1) represses the expression of genes involved in the metabolism of iron, heme and reactive oxygen species and promotes metastasis of various cancers including pancreatic ductal adenocarcinoma (PDAC). However, it is not clear how BACH1 is regulated in PDAC cells. Knockdown of Tank binding kinase 1 (TBK1) led to reductions of BACH1 mRNA and protein amounts in AsPC−1 human PDAC cells. Gene expression analysis of PDAC cells with knockdown of TBK1 or BACH1 suggested the involvement of TBK1 and BACH1 in the regulation of iron homeostasis. Ferritin mRNA and proteins were both increased upon BACH1 knockdown in AsPC−1 cells. Flow cytometry analysis showed that AsPC−1 cells with BACH1 knockout or knockdown contained lower labile iron than control cells, suggesting that BACH1 increased labile iron by repressing the expression of ferritin genes. We further found that the expression of E-cadherin was upregulated upon the chelation of intracellular iron content. These results suggest that the TBK1-BACH1 pathway promotes cancer cell metastasis by increasing labile iron within cells.
Collapse
|
8
|
Therapeutic targeting of TANK-binding kinase signaling towards anticancer drug development: Challenges and opportunities. Int J Biol Macromol 2022; 207:1022-1037. [PMID: 35358582 DOI: 10.1016/j.ijbiomac.2022.03.157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/15/2022]
Abstract
TANK-binding kinase 1 (TBK1) plays a fundamental role in regulating the cellular responses and controlling several signaling cascades. It regulates inflammatory, interferon, NF-κB, autophagy, and Akt pathways. Post-translational modifications (PTM) of TBK1 control its action and subsequent cellular signaling. The dysregulation of the TBK1 pathway is correlated to many pathophysiological conditions, including cancer, that implicates the promising therapeutic advantage for targeting TBK1. The present study summarizes current updates on the molecular mechanisms and cancer-inducing roles of TBK1. Designed inhibitors of TBK1 are considered a potential therapeutic agent for several diseases, including cancer. Data from pre-clinical tumor models recommend that the targeting of TBK1 could be an attractive strategy for anti-tumor therapy. This review further highlighted the therapeutic potential of potent and selective TBK1 inhibitors, including Amlexanox, Compound II, BX795, MRT67307, SR8185 AZ13102909, CYT387, GSK8612, BAY985, and Domainex. These inhibitors may be implicated to facilitate therapeutic management of cancer and TBK1-associated diseases in the future.
Collapse
|
9
|
Sun P, Xue Y. Silence of TANK-binding kinase 1 (TBK1) regulates extracellular matrix degradation of chondrocyte in osteoarthritis by janus kinase (JAK)-signal transducer of activators of transcription (STAT) signaling. Bioengineered 2022; 13:1872-1879. [PMID: 35129065 PMCID: PMC8973840 DOI: 10.1080/21655979.2021.2018976] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
TANK-binding kinase 1 (TBK1) was previously reported to be critical for the regulation of osteoclast differentiation. However, its function in osteoarthritis (OA) has not yet been determined. This study aims to reveal the role of TBK1 in the extracellular matrix (ECM) degradation in OA. C57BL/6 J mice were subjected to anterior cruciate ligament transection (ACLT) surgery to establish an OA animal model. ATDC5 cells were treated with IL-1β to construct a cell model of OA. Changes in the expression of TBK1 were analyzed by qRT-PCR, Western blotting, and immunohistochemistry. Safranin O-fast green staining, ELISA, and Western blotting were performed to evaluate the ECM degradation. By searching GSE75181 and GSE6119 datasets, TBK1 was found to be highly expressed in the OA model. Its upregulation was also confirmed in ACLT mice and in a cell model of OA. Silencing of TBK1 reduced cartilage degradation, OARSI score, and serum levels of CTX-II and COMP. Silencing of TBK1 attenuated ECM degradation, as ADAMTS-4, MMP3, and MMP13 were downregulated, whilst SOX9, collagen II, and aggrecan were upregulated. Furthermore, TBK1 activates the JAK/STAT signaling pathway. Transfection of cells with the STAT3 overexpression plasmid blocked the beneficial effects of TBK1 silencing. In conclusion, TBK1 is highly expressed in OA. Silencing of TBK1 inhibited ECM degradation.
Collapse
Affiliation(s)
- Peng Sun
- Department of Orthopaedic Surgery, Tianjin Medical University General Hospital, Tianjin, P.R. China.,Department of Orthopaedic, Affiliated Hospital of Jining Medical University, Jining Shandong, China
| | - Yuan Xue
- Department of Orthopaedic Surgery, Tianjin Medical University General Hospital, Tianjin, P.R. China
| |
Collapse
|
10
|
Wang B, Zhou M, Lin Y, Ma Y, Cao H. TBK1 regulates the induction of innate immune response against GCRV by phosphorylating IRF3 in rare minnow (Gobiocypris rarus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 115:103883. [PMID: 33045274 DOI: 10.1016/j.dci.2020.103883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
Rare minnow (Gobiocypris rarus), a small cyprinid species that is highly sensitive to the grass carp reovirus (GCRV), is regarded as an ideal model to study the mechanisms of innate immunity in fish. In the present study, a TBK1 homologue from rare minnow (GrTBK1) was identified and its roles in defence against viral infection were investigated. Sequence analysis showed that GrTBK1 encoded a 727-amino acid peptide which shared 98% and 72% identity to the black carp (Mylopharyngodon piceus) and human (Homo sapiens) orthologues, respectively. The amino acid sequence analysis demonstrated that GrTBK1 contains a conserved Serine/Threonine protein kinases catalytic domain (S_TKc) at the N-terminus. Furthermore, cellular distribution proved that GrTBK1 was located in the cytoplasm region. Quantitative real-time PCR analysis revealed that GrTBK1 was ubiquitously expressed in all examined organs, but especially highly in liver. Temporal expression analysis in vivo showed that the expression levels of GrTBK1 were obviously up-regulated in response to GCRV infection. Meanwhile, qRT-PCR assay revealed that the levels of S7 RNA, an important segment of GCRV genome, were higher in the liver than in other tissues. This indicates that GrTBK1 might play a crucial role in responses to GCRV infection in fish. In addition, GrTBK1 activated several type I interferon (IFN) promoters and induced the expression of downstream type I IFN-stimulated genes (ISGs). Furthermore, GrTBK1 obviously phosphorylated the interferon regulatory factor 3 (IRF3). Furthermore, overexpression of GrTBK1 remarkably decreased the GCRV proliferation. In summary, we systematically characterized GrTBK1 and illustrated its role in the innate immune response to GCRV infections.
Collapse
Affiliation(s)
- Bing Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Man Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yusheng Lin
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuegang Ma
- Chongqing Fishery Sciences Research Institute, Chongqing, 400020, China
| | - Hong Cao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
11
|
Revach OY, Liu S, Jenkins RW. Targeting TANK-binding kinase 1 (TBK1) in cancer. Expert Opin Ther Targets 2020; 24:1065-1078. [PMID: 32962465 PMCID: PMC7644630 DOI: 10.1080/14728222.2020.1826929] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/18/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION TANK-binding kinase 1 (TBK1) is a Ser/Thr kinase with a central role in coordinating the cellular response to invading pathogens and regulating key inflammatory signaling cascades. While intact TBK1 signaling is required for successful anti-viral signaling, dysregulated TBK1 signaling has been linked to a variety of pathophysiologic conditions, including cancer. Several lines of evidence support a role for TBK1 in cancer pathogenesis, but the specific roles and regulation of TBK1 remain incompletely understood. A key challenge is the diversity of cellular processes that are regulated by TBK1, including inflammation, cell cycle, autophagy, energy homeostasis, and cell death. Nevertheless, evidence from pre-clinical cancer models suggests that targeting TBK1 may be an effective strategy for anti-cancer therapy in specific settings. AREAS COVERED This review provides an overview of the roles and regulation of TBK1 with a focus on cancer pathogenesis and drug targeting of TBK1 as an anti-cancer strategy. Relevant literature was derived from a PubMed search encompassing studies from 1999 to 2020. EXPERT OPINION TBK1 is emerging as a potential target for anti-cancer therapy. Inhibition of TBK1 alone may be insufficient to restrain the growth of most cancers; hence, combination strategies will likely be necessary. Improved understanding of tumor-intrinsic and tumor-extrinsic TBK1 signaling will inform novel therapeutic strategies.
Collapse
Affiliation(s)
- Or-yam Revach
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Shuming Liu
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Russell W. Jenkins
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Wilcz-Villega E, Carter E, Ironside A, Xu R, Mataloni I, Holdsworth J, Jones W, Moreno Béjar R, Uhlik L, Bentham RB, Godinho SA, Dalli J, Grose R, Szabadkai G, Jones L, Hodivala-Dilke K, Bianchi K. Macrophages induce malignant traits in mammary epithelium via IKKε/TBK1 kinases and the serine biosynthesis pathway. EMBO Mol Med 2020; 12:e10491. [PMID: 31930708 PMCID: PMC7005540 DOI: 10.15252/emmm.201910491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
During obesity, macrophages infiltrate the breast tissue leading to low‐grade chronic inflammation, a factor considered responsible for the higher risk of breast cancer associated with obesity. Here, we formally demonstrate that breast epithelial cells acquire malignant properties when exposed to medium conditioned by macrophages derived from human healthy donors. These effects were mediated by the breast cancer oncogene IKKε and its downstream target—the serine biosynthesis pathway as demonstrated by genetic or pharmacological tools. Furthermore, amlexanox, an FDA‐approved drug targeting IKKε and its homologue TBK1, delayed in vivo tumour formation in a combined genetic mouse model of breast cancer and high‐fat diet‐induced obesity/inflammation. Finally, in human breast cancer tissues, we validated the link between inflammation–IKKε and alteration of cellular metabolism. Altogether, we identified a pathway connecting obesity‐driven inflammation to breast cancer and a potential therapeutic strategy to reduce the risk of breast cancer associated with obesity.
Collapse
Affiliation(s)
- Ewa Wilcz-Villega
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Edward Carter
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Alastair Ironside
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Ruoyan Xu
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Isabella Mataloni
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Julie Holdsworth
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - William Jones
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Rocío Moreno Béjar
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Lukas Uhlik
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Robert B Bentham
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Susana A Godinho
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Jesmond Dalli
- Lipid Mediator Unit, Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Richard Grose
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK.,Francis Crick Institute, London, UK.,Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Kairbaan Hodivala-Dilke
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| | - Katiuscia Bianchi
- Centre for Molecular Oncology, Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, London, UK
| |
Collapse
|
13
|
Arner EN, Du W, Brekken RA. Behind the Wheel of Epithelial Plasticity in KRAS-Driven Cancers. Front Oncol 2019; 9:1049. [PMID: 31681587 PMCID: PMC6798880 DOI: 10.3389/fonc.2019.01049] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular plasticity, a feature associated with epithelial-to-mesenchymal transition (EMT), contributes to tumor cell survival, migration, invasion, and therapy resistance. Phenotypic plasticity of the epithelium is a critical feature in multiple phases of human cancer in an oncogene- and tissue-specific context. Many factors can drive epithelial plasticity, including activating mutations in KRAS, which are found in an estimated 30% of all cancers. In this review, we will introduce cellular plasticity and its effect on cancer progression and therapy resistance and then summarize the drivers of EMT with an emphasis on KRAS effector signaling. Lastly, we will discuss the contribution of cellular plasticity to metastasis and its potential clinical implications. Understanding oncogenic KRAS cellular reprogramming has the potential to reveal novel strategies to control metastasis in KRAS-driven cancers.
Collapse
Affiliation(s)
- Emily N Arner
- Cancer Biology Graduate Program, Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Wenting Du
- Cancer Biology Graduate Program, Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Rolf A Brekken
- Cancer Biology Graduate Program, Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
14
|
Cruz VH, Arner EN, Du W, Bremauntz AE, Brekken RA. Axl-mediated activation of TBK1 drives epithelial plasticity in pancreatic cancer. JCI Insight 2019; 5:126117. [PMID: 30938713 PMCID: PMC6538328 DOI: 10.1172/jci.insight.126117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/27/2019] [Indexed: 01/11/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is characterized by an activating mutation in KRAS. Direct inhibition of KRAS through pharmacological means remains a challenge; however, targeting key KRAS effectors has therapeutic potential. We investigated the contribution of TANK-binding kinase 1 (TBK1), a critical downstream effector of mutant active KRAS, to PDA progression. We report that TBK1 supports the growth and metastasis of KRAS-mutant PDA by driving an epithelial plasticity program in tumor cells that enhances invasive and metastatic capacity. Further, we identify that the receptor tyrosine kinase Axl induces TBK1 activity in a Ras-RalB-dependent manner. These findings demonstrate that TBK1 is central to an Axl-driven epithelial-mesenchymal transition in KRAS-mutant PDA and suggest that interruption of the Axl-TBK1 signaling cascade above or below KRAS has potential therapeutic efficacy in this recalcitrant disease.
Collapse
Affiliation(s)
- Victoria H. Cruz
- Division of Surgical Oncology, Department of Surgery, and the Hamon Center for Therapeutic Oncology Research
| | - Emily N. Arner
- Division of Surgical Oncology, Department of Surgery, and the Hamon Center for Therapeutic Oncology Research
| | - Wenting Du
- Division of Surgical Oncology, Department of Surgery, and the Hamon Center for Therapeutic Oncology Research
| | | | - Rolf A. Brekken
- Division of Surgical Oncology, Department of Surgery, and the Hamon Center for Therapeutic Oncology Research
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
15
|
Zhang Y, Unnithan RVM, Hamidi A, Caja L, Saupe F, Moustakas A, Cedervall J, Olsson AK. TANK-binding kinase 1 is a mediator of platelet-induced EMT in mammary carcinoma cells. FASEB J 2019; 33:7822-7832. [PMID: 30912981 DOI: 10.1096/fj.201801936rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Platelets can promote several stages of the metastatic process and thus contribute to malignant progression. As an example, platelets promote invasive properties of tumor cells by induction of epithelial to mesenchymal transition (EMT). In this study, we show that tumor necrosis factor receptor-associated factor (TRAF) family member-associated NF-κB activator (TANK)-binding kinase 1 (TBK1) is a previously unknown mediator of platelet-induced EMT in mammary carcinoma cells. Coculture of 2 mammary carcinoma cell lines, Ep5 from mice and MCF10A(MII) from humans, with isolated platelets induced morphologic as well as molecular changes characteristic of EMT, which was paralleled with activation of TBK1. TBK1 depletion using small interfering RNA impaired platelet-induced EMT in both Ep5 and MCF10A(MII) cells. Furthermore, platelet-induced activation of the NF-κB subunit p65 was suppressed after TBK1 knockdown, demonstrating that TBK1 mediates platelet-induced NF-κB signaling and EMT. Using an in vivo metastasis assay, we found that depletion of TBK1 from mammary carcinoma cells during in vitro preconditioning with platelets subsequently suppressed the formation of lung metastases in mice. Altogether, these results suggest that TBK1 contributes to tumor invasiveness and may be a driver of metastatic spread in breast cancer.-Zhang, Y., Unnithan, R. V. M., Hamidi, A., Caja, L., Saupe, F., Moustakas, A., Cedervall, J., Olsson, A.-K. TANK-binding kinase 1 is a mediator of platelet-induced EMT in mammary carcinoma cells.
Collapse
Affiliation(s)
- Yanyu Zhang
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Anahita Hamidi
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Laia Caja
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Falk Saupe
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jessica Cedervall
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
Wang S, Li X, Zhang W, Gao Y, Zhang K, Hao Q, Li W, Wang Z, Li M, Zhang W, Zhang Y, Zhang C. Genome-Wide Investigation of Genes Regulated by ERα in Breast Cancer Cells. Molecules 2018; 23:molecules23102543. [PMID: 30301189 PMCID: PMC6222792 DOI: 10.3390/molecules23102543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/26/2018] [Accepted: 10/03/2018] [Indexed: 01/13/2023] Open
Abstract
Estrogen receptor alpha (ERα), which has been detected in over 70% of breast cancer cases, is a driving factor for breast cancer growth. For investigating the underlying genes and networks regulated by ERα in breast cancer, RNA-seq was performed between ERα transgenic MDA-MB-231 cells and wild type MDA-MB-231 cells. A total of 267 differentially expressed genes (DEGs) were identified. Then bioinformatics analyses were performed to illustrate the mechanism of ERα. Besides, by comparison of RNA-seq data obtained from MDA-MB-231 cells and microarray dataset obtained from estrogen (E2) stimulated MCF-7 cells, an overlap of 126 DEGs was screened. The expression level of ERα was negatively associated with metastasis and EMT in breast cancer. We further verified that ERα might inhibit metastasis by regulating of VCL and TNFRSF12A, and suppress EMT by the regulating of JUNB and ID3. And the relationship between ERα and these genes were validated by RT-PCR and correlation analysis based on TCGA database. By PPI network analysis, we identified TOP5 hub genes, FOS, SP1, CDKN1A, CALCR and JUNB, which were involved in cell proliferation and invasion. Taken together, the whole-genome insights carried in this work can help fully understanding biological roles of ERα in breast cancer.
Collapse
Affiliation(s)
- Shuning Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Xiaoju Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Wangqian Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Kuo Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Zhaowei Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Wei Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
17
|
Ma Z, Xin Z, Hu W, Jiang S, Yang Z, Yan X, Li X, Yang Y, Chen F. Forkhead box O proteins: Crucial regulators of cancer EMT. Semin Cancer Biol 2018; 50:21-31. [PMID: 29427645 DOI: 10.1016/j.semcancer.2018.02.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 12/02/2017] [Accepted: 02/05/2018] [Indexed: 12/12/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is an acknowledged cellular transition process in which epithelial cells acquire mesenchymal-like properties that endow cancer cells with increased migratory and invasive behavior. Forkhead box O (FOXO) proteins have been shown to orchestrate multiple EMT-associated pathways and EMT-related transcription factors (EMT-TFs), thereby modulating the EMT process. The focus of the current review is to evaluate the latest research progress regarding the roles of FOXO proteins in cancer EMT. First, a brief overview of the EMT process in cancer and a general background on the FOXO family are provided. Next, we present the interactions between FOXO proteins and multiple EMT-associated pathways during malignancy development. Finally, we propose several novel potential directions for future research. Collectively, the information compiled herein should serve as a comprehensive repository of information on this topic and should aid in the design of additional studies and the future development of FOXO proteins as therapeutic targets.
Collapse
Affiliation(s)
- Zhiqiang Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069 China; Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Zhenlong Xin
- Department of Occupational and Environmental Health and The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Hu
- Department of Immunology, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069 China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| | - Fulin Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069 China.
| |
Collapse
|
18
|
Hu Y, Huang Y, Liu J, Zhang J, Qin Q, Huang X. TBK1 from orange-spotted grouper exerts antiviral activity against fish viruses and regulates interferon response. FISH & SHELLFISH IMMUNOLOGY 2018; 73:92-99. [PMID: 29222027 DOI: 10.1016/j.fsi.2017.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/06/2017] [Accepted: 12/04/2017] [Indexed: 06/07/2023]
Abstract
TANK-binding kinase-1 (TBK1) has been well studied in mammals because of its importance in type I interferon induction in antiviral immunity. However, the roles of fish TBK1 in virus infection still remained largely uncertain. In the current study, a TBK1 homolog from orange-spotted grouper (Epinephelus coioides) (EcTBK1) was cloned and its roles in fish viral infections were investigated. Sequence analysis showed that EcTBK1 encoded a 723-amino acid peptide which shared 98% and 73% identity to large yellow croaker (Larimichthys crocea) and human (homo sapiens), respectively. Multiple sequence alignments indicated that EcTBK1 contained conserved domains, including N-terminal kinase domain (KD), the middle ubiquitin-like domain (ULD) and C-terminal coiled-coil (CC) domains. The tissue distribution profiles demonstrated that EcTBK1 gene was constitutively expressed in all examined tissues, with predominant expression in intestine. Temporal expression analysis in vitro showed that the expression levels of EcTBK1 were significantly up-regulated in response to both red-spotted grouper nervous necrosis virus (RGNNV) and Singapore grouper iridovirus (SGIV) infection, suggested that EcTBK1 might exert crucial roles in fish virus infection. Subcellular localization indicated that EcTBK1 expression was primarily in the cytoplasm in GS cells. The ectopic expression of EcTBK1 significantly inhibited both SGIV and RGNNV replication. Furthermore, EcTBK1 overexpression significantly increased the expression levels of interferon related cytokines and pro-inflammatory factors. In addition, the overexpression of EcTBK1 increased the IRF3- and IRF7-regulated interferon promoter ISRE and IFN activity, and the regulatory effect on interferon immune response were dependent on its kinase domain. Together, we speculated that grouper TBK1 exerted antiviral activity against iridovirus and nodavirus via regulating the interferon immune and inflammatory response.
Collapse
Affiliation(s)
- Yin Hu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing, China; Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Youhua Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Jiaxin Liu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing, China; Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Jingcheng Zhang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing, China; Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China.
| | - Xiaohong Huang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; University of Chinese Academy of Sciences, Beijing, China; Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China.
| |
Collapse
|
19
|
Lee JH, Jung SM, Yang KM, Bae E, Ahn SG, Park JS, Seo D, Kim M, Ha J, Lee J, Kim JH, Kim JH, Ooshima A, Park J, Shin D, Lee YS, Lee S, van Loo G, Jeong J, Kim SJ, Park SH. A20 promotes metastasis of aggressive basal-like breast cancers through multi-monoubiquitylation of Snail1. Nat Cell Biol 2017; 19:1260-1273. [DOI: 10.1038/ncb3609] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/10/2017] [Indexed: 12/16/2022]
|
20
|
Yang L, Lv Q, Chen W, Sun J, Wu Y, Wang Y, Chen X, Chen X, Zhang Z. Presence of embryonic DNA in culture medium. Oncotarget 2017; 8:67805-67809. [PMID: 28978073 PMCID: PMC5620213 DOI: 10.18632/oncotarget.18852] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/01/2017] [Indexed: 11/25/2022] Open
Abstract
Preimplantation genetic diagnosis (PGD) has successfully assisted couples with genetic diseases to conceive healthy babies during the past decades. However, biopsy of the blastomere has potential lesion to the embryos which commonly results in abortion. Thus, a noninvasive PGD is needed. In the past, the presence of genetic materials in maternal plasma or serum has triggered a great innovation of noninvasive prenatal diagnosis. Nevertheless, it is not clear whether embryonic DNA is also present in embryonic culture medium. Here, a rapid-boiling method has been used to harvest DNA from the medium or the discarded embryos, following Polymerase Chain Reaction (PCR) was applied to detect the dissociative DNA by amplifying SRY gene (Y-chromosome). For the first time, the Y sequences were detected in the medium which were used to culture embryo for above 3 days. None of the positive signal was examined in Day 1 and Day 2 embryonic culture medium. Our findings suggest that the Y chromosome fragments from the embryo may release into its culture medium. If validated in a larger cohort, detection of SRY gene may prove to be a useful method to screen Y-linked genetic disease. More importantly, detecting the free DNA in the embryonic culture medium may represent a novel strategy for noninvasive PGD.
Collapse
Affiliation(s)
- Linlin Yang
- The Reproductive Medicine Center of Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China.,Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai 201900, China
| | - Qiaoying Lv
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Wei Chen
- The Reproductive Medicine Center of Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Jian Sun
- The Reproductive Medicine Center of Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Yu Wu
- The Reproductive Medicine Center of Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Yiying Wang
- Department of Obstetrics and Gynecology, Henan Province People's Hospital, Zhengzhou 450000, China
| | - Xiong Chen
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai 201900, China
| | - Xiaojun Chen
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Zhenbo Zhang
- The Reproductive Medicine Center of Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China.,Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai 201900, China
| |
Collapse
|
21
|
Lv QY, Xie BY, Yang BY, Ning CC, Shan WW, Gu C, Luo XZ, Chen XJ, Zhang ZB, Feng YJ. Increased TET1 Expression in Inflammatory Microenvironment of Hyperinsulinemia Enhances the Response of Endometrial Cancer to Estrogen by Epigenetic Modulation of GPER. J Cancer 2017; 8:894-902. [PMID: 28382153 PMCID: PMC5381179 DOI: 10.7150/jca.17064] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/09/2016] [Indexed: 12/27/2022] Open
Abstract
Background: Insulin resistance (IR) has been well studied in the initiation and development of endometrial endometrioid carcinoma (EEC). As yet, it has been largely neglected for estrogen sensitivity in local endometrium in hyperinsulinemia-induced systemic microenvironment. The aim of this study was to investigate the role of insulin in regulating estrogen sensitivity and explore the potential mechanisms in insulin-driven inflammatory microenvironment. Methods: We first investigated the effect of insulin on estradiol-driven endometrial cancer cells proliferation in vitro to address the roles of insulin in modulating estrogen sensitivity. Then GPER, ERα and TET1 in EEC samples with or without insulin resistance were screened by immunohistochemistry to confirm whether insulin resistance regulates estrogen receptors. Further mechanism analysis was carried out to address whether TET1 was mediated epigenetic modulation of GPER in insulin-induced microenvironment. Results: Insulin enhanced estradiol-driven endometrial cancer cells proliferation by up-regulating G-protein-coupled estrogen receptor (GPER) expression, but not ERα or ERβ. Immunohistochemistry of EEC tissues showed that GPER expression was greatly increased in endometrial tissues from EEC subjects with insulin resistance and was positively correlated with Ten-eleven-translocation 1 (TET1) expression. Mechanistically, insulin up-regulates TET1 expression, and the latter, an important DNA hydroxymethylase, could up-regulate GPER expression through epigenetic modulation. Conclusion: This study identified TET1 as the upstream regulator of GPER expression and provides a possible mechanism that insulin-induced positive regulation of estrogen sensitivity in endometrial cancer cells. Increasing expression of GPER through TET1-mediated epigenetic modulation may emerge as the main regulator to enhance the response of endometrial cancer to estrogen in insulin-driven inflammatory microenvironment.
Collapse
Affiliation(s)
- Qiao-Ying Lv
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Bing-Ying Xie
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Bing-Yi Yang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Cheng-Cheng Ning
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Wei-Wei Shan
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Chao Gu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Xue-Zhen Luo
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Xiao-Jun Chen
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Zhen-Bo Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University school of medicine, Shanghai, 201620, China
| | - You-Ji Feng
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University school of medicine, Shanghai, 201620, China
| |
Collapse
|
22
|
Gao Y, Wang Z, Hao Q, Li W, Xu Y, Zhang J, Zhang W, Wang S, Liu S, Li M, Xue X, Zhang W, Zhang C, Zhang Y. Loss of ERα induces amoeboid-like migration of breast cancer cells by downregulating vinculin. Nat Commun 2017; 8:14483. [PMID: 28266545 PMCID: PMC5344302 DOI: 10.1038/ncomms14483] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/20/2016] [Indexed: 12/16/2022] Open
Abstract
Oestrogen receptor alpha (ERα) is a well-known target of endocrine therapy for ERα-positive breast cancer. ERα-negative cells, which are enriched during endocrine therapy, are associated with metastatic relapse. Here we determine that loss of ERα in the invasive front and in lymph node metastasis in human breast cancer is significantly correlated with lymphatic metastasis. Using in vivo and in vitro experiments, we demonstrate that ERα inhibits breast cancer metastasis. Furthermore, we find that ERα is a novel regulator of vinculin expression in breast cancer. Notably, ERα suppresses the amoeboid-like movement of breast cancer cells by upregulating vinculin in 3D matrix, which in turn promotes cell–cell and cell–matrix adhesion and inhibits the formation of amoeboid-like protrusions. A positive association between ERα and vinculin expression is found in human breast cancer tissues. The results show that ERα inhibits breast cancer metastasis and suggest that ERα suppresses cell amoeboid-like movement by upregulating vinculin. Estrogen receptor alpha (ERα)-negative cells, which are enriched during endocrine therapy, are associated with metastatic relapse of breast cancer. Here the authors show that ERα inhibits breast cancer metastasis and suggest that ERα suppresses the amoeboid-like migration of breast cancer cells by upregulating vinculin.
Collapse
Affiliation(s)
- Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhaowei Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Yujin Xu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Juliang Zhang
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Wangqian Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shuning Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shuo Liu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Xiaochang Xue
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| |
Collapse
|
23
|
Xie BY, Lv QY, Ning CC, Yang BY, Shan WW, Cheng YL, Gu C, Luo XZ, Zhang ZB, Chen XJ, Xi XW, Feng YJ. TET1-GPER-PI3K/AKT pathway is involved in insulin-driven endometrial cancer cell proliferation. Biochem Biophys Res Commun 2016; 482:857-862. [PMID: 27889612 DOI: 10.1016/j.bbrc.2016.11.124] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 11/22/2016] [Indexed: 10/20/2022]
Abstract
Large amount of clinical evidence has demonstrated that insulin resistance is closely related to oncogenesis of endometrial cancer (EC). Despite recent studies showed the up-regulatory role of insulin in G protein-coupled estrogen receptor (GPER/GPR30) expression, GPER expression was not decreased compared to control when insulin receptor was blocked even in insulin treatment. The purpose of this study was to explore the possible mechanism by which insulin up-regulates GPER that drives EC cell proliferation. For this purpose, we first investigated the GPER expression in tissues of endometrial lesions, further explored the effect of GPER on EC cell proliferation in insulin resistance context. Then we analyzed the role of Ten-Eleven Translocation 1 (TET1) in insulin-induced GEPR expression and EC cell proliferation. The results showed that GPER was highly expressed in endometrial atypical hyperplasia and EC tissues. Mechanistically, insulin up-regulated TET1 expression and the latter played an important role in up-regulating GPER expression and activating PI3K/AKT signaling pathway. TET1 mediated GPER up-regulation was another mechanism that insulin promotes EC cell proliferation.
Collapse
Affiliation(s)
- Bing-Ying Xie
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Qiao-Ying Lv
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Cheng-Cheng Ning
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Bing-Yi Yang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Wei-Wei Shan
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Ya-Li Cheng
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Chao Gu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Xue-Zhen Luo
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Zhen-Bo Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Xiao-Jun Chen
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China.
| | - Xiao-Wei Xi
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
| | - You-Ji Feng
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| |
Collapse
|
24
|
Yu T, Yang Y, Yin DQ, Hong S, Son YJ, Kim JH, Cho JY. TBK1 inhibitors: a review of patent literature (2011 - 2014). Expert Opin Ther Pat 2015; 25:1385-96. [PMID: 26293650 DOI: 10.1517/13543776.2015.1081168] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION TANK-binding kinase 1 (TBK1) is a noncanonical IκB kinase family member that regulates the innate immune response. Misregulation of TBK1 activity can promote inflammatory disorders and oncogenesis; therefore, TBK1 inhibitors are considered a promising therapy for inflammation and cancer. AREAS COVERED In this review, the authors provide information on the role of TBK1 in human health and on recently developed inhibitors from patents granted from 2011 to 2014. The reader will gain an understanding of the mechanisms of TBK1 function as well as the structure and biological activity of recently developed TBK1 inhibitors. Google and NCBI search engines were used to find relevant patents and clinical information using "TBK1 inhibitor" as the search term. EXPERT OPINION The role of TBK1 in various diseases has prompted the further investigation of significant targets. Although research on TBK1 inhibitors has increased over the last few years, only a few inhibitors of this kinase have been identified. In addition, almost all of the chemical inhibitors are modified from different scaffolds and/or chemotypes of pyrimidine. Specifically, compound BX795 is the representative one, which was first patented as a potent TBK1 inhibitor. Even though some compounds have displayed interesting potential inhibition and selectivity of TBK1 in vitro and in in vivo trials, the development of more efficient and selective TBK1 inhibitors is still required.
Collapse
Affiliation(s)
- Tao Yu
- a 1 Qingdao University, Medical College , Qingdao 266071, China
| | - Yanyan Yang
- a 1 Qingdao University, Medical College , Qingdao 266071, China
| | - De Qing Yin
- b 2 Linyi Center for Disease Control and Prevention , Linyi 276000, China
| | - Sungyoul Hong
- c 3 Sungkyunkwan University, Department of Genetic Engineering , 300 Chuncheon-Dong, Suwon 440-746, Korea +82 312 907 868 ; +82 312 907 870 ;
| | - Young-Jin Son
- d 4 Sunchon National University, Department of Pharmacy , Suncheon 540-742, Republic of Korea +82 617 503 755 ; +82 617 503 708 ;
| | - Jong-Hoon Kim
- e 5 Chonbuk National University, College of Veterinary Medicine, Biosafety Research Institute, Department of Veterinary Physiology , Jeonju 561-756, Republic of Korea +82 632 702 563 ; +82 632 703 780 ;
| | - Jae Youl Cho
- c 3 Sungkyunkwan University, Department of Genetic Engineering , 300 Chuncheon-Dong, Suwon 440-746, Korea +82 312 907 868 ; +82 312 907 870 ;
| |
Collapse
|
25
|
Capdevila-Busquets E, Badiola N, Arroyo R, Alcalde V, Soler-López M, Aloy P. Breast cancer genes PSMC3IP and EPSTI1 play a role in apoptosis regulation. PLoS One 2015; 10:e0115352. [PMID: 25590583 PMCID: PMC4295872 DOI: 10.1371/journal.pone.0115352] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/22/2014] [Indexed: 11/19/2022] Open
Abstract
A key element to delineate the biology of individual tumors is the regulation of apoptosis. In this work, we functionally characterize two breast cancer associated genes, the proteasome 26S subunit ATPase 3 interacting protein (PSMC3IP) and the epithelial-stromal interaction 1 (EPSTI1), to explore their potential apoptotic role in breast cancer. We first explore the existence of direct physical interactions with annotated BC-apoptotic genes. Based on the generated interaction network, we examine several apoptotic markers to determine the effect of PSMC3IP and EPSTI1 gene expression modulation in two different human breast cancer cell lines to suggest potential molecular mechanisms to unveil their role in the disease. Our results show that PSMC3IP and EPSTI1 are able to modulate the extrinsic apoptotic pathway in estrogen receptor positive and triple negative breast cancer cell lines, highlighting them as potential therapeutic targets.
Collapse
Affiliation(s)
- Eva Capdevila-Busquets
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Nahuai Badiola
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Rodrigo Arroyo
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Víctor Alcalde
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Montserrat Soler-López
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
- * E-mail:
| |
Collapse
|
26
|
Wu J, Zhao X, Lin Z, Shao Z. A system level analysis of gastric cancer across tumor stages with RNA-seq data. MOLECULAR BIOSYSTEMS 2015; 11:1925-32. [DOI: 10.1039/c5mb00105f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gastric cancer is the third leading cause of cancer-related death in the world.
Collapse
Affiliation(s)
- Jun Wu
- Department of Automation
- Shanghai Jiao Tong University
- and Key Laboratory of System Control and Information Processing of Ministry of Education
- Shanghai
- China
| | - Xiaodong Zhao
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai
- China
| | - Zongli Lin
- Charles L. Brown Department of Electrical and Computer Engineering
- University of Virginia
- Charlottesville
- USA
| | - Zhifeng Shao
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai
- China
| |
Collapse
|
27
|
Cytoplasmic DRAK1 overexpressed in head and neck cancers inhibits TGF-β1 tumor suppressor activity by binding to Smad3 to interrupt its complex formation with Smad4. Oncogene 2014; 34:5037-45. [PMID: 25531329 DOI: 10.1038/onc.2014.423] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/11/2014] [Accepted: 11/15/2014] [Indexed: 12/11/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an extremely aggressive cancer with a poor prognosis and low patient survival. Because chemotherapy for advanced HNSCC is often ineffective, discovering new therapeutic targets that are important for HNSCC development and progression and elucidating their molecular mechanisms are required. In the present study, we describe the role of DRAK1 (death-associated protein kinase-related apoptosis-inducing kinase 1) as a novel negative regulator of the transforming growth factor-β (TGF-β) tumor suppressor signaling pathway for the first time in human HNSCC cells. DRAK1 was significantly overexpressed in primary human HNSCCs and in HNSCC cell lines. Through gain- and loss-of-function experiments, we demonstrated that the DRAK1 expression level regulated TGF-β1-induced transcriptional activity and expression of the tumor suppressor gene p21(Waf1/Cip1). DRAK1 depletion enhanced TGF-β1-induced growth inhibition in vitro and suppressed tumorigenicity in xenograft models in vivo. Mechanistically, DRAK1 was predominantly localized in the cytoplasm and bound to Smad3, thereby interrupting Smad3/Smad4 complex formation, which is the core process for the induction of tumor suppressor genes by TGF-β1. Thus, our findings suggest that cytoplasmic DRAK1 increases tumorigenic potential through inhibition of TGF-β1-mediated tumor suppressor activity in HNSCC cells and may be a potential therapeutic target for HNSCCs.
Collapse
|
28
|
Vu HL, Aplin AE. Targeting TBK1 inhibits migration and resistance to MEK inhibitors in mutant NRAS melanoma. Mol Cancer Res 2014; 12:1509-19. [PMID: 24962318 DOI: 10.1158/1541-7786.mcr-14-0204] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
UNLABELLED Melanoma is a devastating form of skin cancer with limited therapeutic options. Fifteen to 20% of patients with melanoma have an activating mutation in the GTPase, NRAS. The major downstream effectors of RAS are RAFs (ARAF, BRAF, and CRAF), phosphoinositide 3-kinase (PI3K), and the Ral guanine exchange factors (RalGEF). TANK-binding kinase 1 (TBK1) is an atypical IκB kinase family member that acts downstream of RalGEFs. Whereas many studies have analyzed RAF and PI3K signaling in mutant NRAS melanoma, the role of RalGEF/Ral is understudied and TBK1 has not been examined. To address this, TBK1 was modulated with knockdown approaches and targeted therapies to determine the role of TBK1 in motility, apoptosis, and signaling. In melanoma, NRAS overexpression increased TBK1 phosphorylation. TBK1 depletion inhibited migration and invasion, whereas its constitutive overexpression led to an increase in invasion. In three-dimensional systems that mimic the dermal microenvironment, TBK1 depletion or inhibition cooperated with MEK inhibitors to promote apoptosis, particularly in the context of MEK-insensitive mutant NRAS. This effect was absent in melanoma cells that are wild-type for NRAS. These results suggest the utility of TBK1 inhibitors as part of a treatment regimen for patients with mutant NRAS melanoma, for whom there are no current effective therapies. IMPLICATIONS TBK1 promotes the malignant properties of NRAS-mutant melanoma and its targeting, in combination with MEK, promotes apoptosis, thus providing a potential novel targeted therapeutic option.
Collapse
Affiliation(s)
- Ha Linh Vu
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
29
|
Xie G, Ji A, Yuan Q, Jin Z, Yuan Y, Ren C, Guo Z, Yao Q, Yang K, Lin X, Chen L. Tumour-initiating capacity is independent of epithelial-mesenchymal transition status in breast cancer cell lines. Br J Cancer 2014; 110:2514-23. [PMID: 24755887 PMCID: PMC4021510 DOI: 10.1038/bjc.2014.153] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 12/18/2022] Open
Abstract
Background: Epithelial–mesenchymal transition (EMT) and cancer stem cells (CSCs) are considered to be crucial for cancer biology. The purpose of this study was to determine whether EMT directly led to the acquisition of tumour-initiating capacity in breast cancer cell lines. Methods: Epithelial–mesenchymal transition was induced in five breast cancer cell lines and one normal breast cell line by EMT-related cytokine stimulation. Mesenchymal–epithelial transition (MET) was induced by stably overexpressing miR-200c in three mesenchymal-like breast cancer cell lines. Molecular expression and cell function analysis were performed to evaluate the effect of EMT or MET on tumour-initiating capacity and other biological characteristics. Results: The induction of EMT did not enhance tumour-initiating capacity but, instead, conferred a CD44+/CD24−/low phenotype as well as cell proliferation, migration, and resistance to doxorubicin and radiation on breast cancer cell lines. Furthermore, MET did not lead to inhibition or loss of the tumour-initiating capacity in mesenchymal-like breast cancer cell lines, but it markedly attenuated other malignant properties, including proliferation, invasion, and resistance to therapy. Conclusions: Epithelial–mesenchymal transition does not alter tumour-initiating capacity of breast cancer cells but some other biological characteristics. Therefore, EMT and tumour-initiating capacity may not be directly linked in breast cancer cell lines.
Collapse
Affiliation(s)
- G Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - A Ji
- Department of Pharmaceutical Science, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, PR China
| | - Q Yuan
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Z Jin
- Department of Pharmacy, Jining First People's Hospital, Jining, Shandong 272111, PR China
| | - Y Yuan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - C Ren
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Z Guo
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Q Yao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - K Yang
- Department of Neurosurgery, Institute of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - X Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - L Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| |
Collapse
|
30
|
Jiang Z, Liu JC, Chung PED, Egan SE, Zacksenhaus E. Targeting HER2(+) breast cancer: the TBK1/IKKε axis. Oncoscience 2014; 1:180-2. [PMID: 25594009 PMCID: PMC4278282 DOI: 10.18632/oncoscience.18] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/05/2014] [Indexed: 11/25/2022] Open
Abstract
HER2+ breast cancer (BC) is a highly aggressive subtype, affecting ~20% of BC patients. Current treatments include adjuvant or neoadjuvant chemotherapy plus anti-HER2 agents such as trastuzumab, a monoclonal antibody directed against HER2. Despite improvement in disease free survival, most patients eventually succumb to metastatic disease, which is largely incurable. Consequently, there is an urgent need to identify novel drugs that can efficiently kill HER2+ BC and/or potentiate the effect of existing anti-HER2 therapies. We performed a lenti-viral shRNA kinome screen on non-adherent mouse Her2/Neu tumorspheres and identified TBK1, a non-canonical IκB kinase (IKK), as the most potent target [1]. TBK1 knock-down, or treatment with TBK1-II, a drug that efficiently inhibits TBK1 and its close relative IKKε (IKBKE), suppressed growth of human HER2+ BC cells and induced cellular senescence. Senescence was associated with inhibition of phosphorylated/active p65-NFkB and induction of the cell cycle inhibitor, p16ink4a. In addition, TBK1-II cooperated with lapatinib, a EGFR/HER2 inhibitor, to accelerate apoptosis in vitro and suppress tumor growth in a xenograft model of HER2+ BC. Thus, TBK1/IKKε inhibitors may improve treatment of HER2+ BC in cooperation with anti-HER2 therapy.
Collapse
Affiliation(s)
- Zhe Jiang
- Division of Advanced Diagnostics, Toronto General Research Institute - University Health Network, Toronto, Ontario, Canada
| | - Jeff C Liu
- Division of Advanced Diagnostics, Toronto General Research Institute - University Health Network, Toronto, Ontario, Canada
| | - Philip E D Chung
- Division of Advanced Diagnostics, Toronto General Research Institute - University Health Network, Toronto, Ontario, Canada
| | - Sean E Egan
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Department of Molecular Genetics, University of Toronto
| | - Eldad Zacksenhaus
- Division of Advanced Diagnostics, Toronto General Research Institute - University Health Network, Toronto, Ontario, Canada
| |
Collapse
|