1
|
Phan Van T, Huyen Ton Nu Bao T, Leya M, Zhou Z, Jeong H, Lim CW, Kim B. Amlexanox attenuates LPS-induced neuroinflammatory responses in microglial cells via inhibition of NF-κB and STAT3 signaling pathways. Sci Rep 2024; 14:2744. [PMID: 38302598 PMCID: PMC10834963 DOI: 10.1038/s41598-024-53235-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/30/2024] [Indexed: 02/03/2024] Open
Abstract
Amlexanox is an anti-inflammatory and anti-allergic agent used clinically for the treatment of aphthous ulcers, allergic rhinitis, and asthma. Recent studies have demonstrated that amlexanox, a selective inhibitor of IkB kinase epsilon (IKKε) and TANK-binding kinase 1 (TBK1), suppresses a range of diseases or inflammatory conditions, such as obesity-related metabolic dysfunction and type 2 diabetes. However, the effects of amlexanox on neuroinflammatory responses to amlexanox have not yet been comprehensively studied. In this study, we investigated the novel therapeutic effect of amlexanox on LPS-induced neuroinflammation in vivo, and intraperitoneal injection of amlexanox markedly reduced LPS-induced IKKε levels, proinflammatory cytokines, and microglial activation, as evidenced by ionized calcium-binding adapter molecule 1 (Iba1) immunostaining. Furthermore, amlexanox significantly reduced proinflammatory cytokines and chemokines in LPS-induced bone marrow-derived macrophages (BMDM), murine BV2, and human HMC3 microglial cells. This data provided considerable evidence that amlexanox can be used as a preventive and curative therapy for neuroinflammatory and neurodegenerative diseases. In terms of mechanism aspects, our results demonstrated that the anti-inflammatory action of amlexanox in BV2 microglial cells was through the downregulation of NF-κB and STAT3 signaling pathways. In addition, the combination of amlexanox and SPI (a STAT3 selective inhibitor) showed high efficiency in inhibiting the production of neurotoxic and pro-inflammatory mediators. Overall, our data provide rational insights into the mechanisms of amlexanox as a potential therapeutic strategy for neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Thach Phan Van
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, 54596, Republic of Korea
- Department of Biotechnology, NTT Hi-tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Viet Nam
| | - Tien Huyen Ton Nu Bao
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, 54596, Republic of Korea
| | - Mwense Leya
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, 54596, Republic of Korea
| | - Zixiong Zhou
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Hyuneui Jeong
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, 54596, Republic of Korea
| | - Chae-Woong Lim
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, 54596, Republic of Korea
| | - Bumseok Kim
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, 54596, Republic of Korea.
| |
Collapse
|
2
|
Runde AP, Mack R, S J PB, Zhang J. The role of TBK1 in cancer pathogenesis and anticancer immunity. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:135. [PMID: 35395857 PMCID: PMC8994244 DOI: 10.1186/s13046-022-02352-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/29/2022] [Indexed: 02/07/2023]
Abstract
The TANK-binding kinase 1 (TBK1) is a serine/threonine kinase belonging to the non-canonical inhibitor of nuclear factor-κB (IκB) kinase (IKK) family. TBK1 can be activated by pathogen-associated molecular patterns (PAMPs), inflammatory cytokines, and oncogenic kinases, including activated K-RAS/N-RAS mutants. TBK1 primarily mediates IRF3/7 activation and NF-κB signaling to regulate inflammatory cytokine production and the activation of innate immunity. TBK1 is also involved in the regulation of several other cellular activities, including autophagy, mitochondrial metabolism, and cellular proliferation. Although TBK1 mutations have not been reported in human cancers, aberrant TBK1 activation has been implicated in the oncogenesis of several types of cancer, including leukemia and solid tumors with KRAS-activating mutations. As such, TBK1 has been proposed to be a feasible target for pharmacological treatment of these types of cancer. Studies suggest that TBK1 inhibition suppresses cancer development not only by directly suppressing the proliferation and survival of cancer cells but also by activating antitumor T-cell immunity. Several small molecule inhibitors of TBK1 have been identified and interrogated. However, to this point, only momelotinib (MMB)/CYT387 has been evaluated as a cancer therapy in clinical trials, while amlexanox (AMX) has been evaluated clinically for treatment of type II diabetes, nonalcoholic fatty liver disease, and obesity. In this review, we summarize advances in research into TBK1 signaling pathways and regulation, as well as recent studies on TBK1 in cancer pathogenesis. We also discuss the potential molecular mechanisms of targeting TBK1 for cancer treatment. We hope that our effort can help to stimulate the development of novel strategies for targeting TBK1 signaling in future approaches to cancer therapy.
Collapse
Affiliation(s)
- Austin P Runde
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Ryan Mack
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Peter Breslin S J
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA.,Departments of Molecular/Cellular Physiology and Biology, Loyola University Medical Center and Loyola University Chicago, Chicago, IL, 60660, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA. .,Departments of Pathology and Radiation Oncology, Loyola University Medical Center, Maywood, IL, 60153, USA.
| |
Collapse
|
3
|
Chen S, Ni M, Hu T, Gu Y, Feng C, Pan C, Zhang S, Wen S, Zhao N, Wang W, Dai L, Wang J. TANK-binding kinase 1 inhibitor GSK8612 enhances daunorubicin sensitivity in acute myeloid leukemia cells via the AKT-CDK2 pathway. Am J Transl Res 2021; 13:13640-13653. [PMID: 35035703 PMCID: PMC8748083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/01/2021] [Indexed: 06/14/2023]
Abstract
PURPOSE It has been established in previous studies that TANK-binding kinase 1 (TBK1) is upregulated in malignant tumors and is therefore associated with poor prognosis. However, the role of TBK1 in acute myeloid leukemia (AML) remains unclear. In this study, we investigated the expression levels and the function of TBK1 in AML. METHODS First, TBK1 expression was detected and analyzed using Western blot and qRT-PCR. Then, GSK8612, a novel TBK1 inhibitor, and TBK1-specific siRNA (si-TBK1) were used to inhibit TBK1 function and expression. The effects of TBK1 inhibition on AML were investigated first through a cell counting kit (CCK-8) assay, followed by trypan blue staining to assess cell apoptosis and cell cycle progression in vitro. Finally, the signaling pathway activities in HL-60 and Kasumi-1 cells and patients' mononuclear cells (MNCs) were explored using western blot. RESULTS We found a significantly higher TBK1 expression in AML patients with poor prognoses. GSK8612 successfully inhibited TBK1 expression, resulting in the increased sensitivity of AML cells to daunorubicin. Mechanistically, TBK1 inhibition (by GSK8612 and si-TBK1) regulated cyclin-dependent kinase 2 (CDK2) levels in AML cells via the AKT pathway. Moreover, it was observed that the inhibition of protein kinase B (AKT) activity also resulted in the increased sensitivity of AML cell lines to daunorubicin, validating the relationship between TBK1 and the AKT-CDK2 pathway. Similar results were obtained in MNCs from patients with AML. CONCLUSION TBK1 is a potential prognostic factor for AML, and its inhibition may improve the sensitivity of AML cells to daunorubicin. This regulatory effect is predicted to involve the TBK1-AKT-CDK2 pathway.
Collapse
Affiliation(s)
- Siyu Chen
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
| | - Ming Ni
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
| | - Tianzhen Hu
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
| | - Yangguang Gu
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
| | - Cheng Feng
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
| | - Chengyun Pan
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
| | - Siyu Zhang
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
| | - Shuangshuang Wen
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
| | - Naiqin Zhao
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
| | - Weili Wang
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
| | - Lihong Dai
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
| | - Jishi Wang
- Guizhou Medical UniversityGuiyang 550025, Guizhou, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation CentreGuiyang 550005, Guizhou, China
| |
Collapse
|
4
|
Xu R, Jones W, Wilcz-Villega E, Costa AS, Rajeeve V, Bentham RB, Bryson K, Nagano A, Yaman B, Olendo Barasa S, Wang Y, Chelala C, Cutillas P, Szabadkai G, Frezza C, Bianchi K. The breast cancer oncogene IKKε coordinates mitochondrial function and serine metabolism. EMBO Rep 2020; 21:e48260. [PMID: 32783398 PMCID: PMC7116048 DOI: 10.15252/embr.201948260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/29/2020] [Accepted: 07/09/2020] [Indexed: 12/25/2022] Open
Abstract
IκB kinase ε (IKKε) is a key molecule at the crossroads of inflammation and cancer. Known to regulate cytokine secretion via NFκB and IRF3, the kinase is also a breast cancer oncogene, overexpressed in a variety of tumours. However, to what extent IKKε remodels cellular metabolism is currently unknown. Here, we used metabolic tracer analysis to show that IKKε orchestrates a complex metabolic reprogramming that affects mitochondrial metabolism and consequently serine biosynthesis independently of its canonical signalling role. We found that IKKε upregulates the serine biosynthesis pathway (SBP) indirectly, by limiting glucose‐derived pyruvate utilisation in the TCA cycle, inhibiting oxidative phosphorylation. Inhibition of mitochondrial function induces activating transcription factor 4 (ATF4), which in turn drives upregulation of the expression of SBP genes. Importantly, pharmacological reversal of the IKKε‐induced metabolic phenotype reduces proliferation of breast cancer cells. Finally, we show that in a highly proliferative set of ER negative, basal breast tumours, IKKε and PSAT1 are both overexpressed, corroborating the link between IKKε and the SBP in the clinical context.
Collapse
Affiliation(s)
- Ruoyan Xu
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - William Jones
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Ewa Wilcz-Villega
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Ana Sh Costa
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK.,Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Vinothini Rajeeve
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Robert B Bentham
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK.,Francis Crick Institute, London, UK
| | - Kevin Bryson
- Department of Computer Sciences, University College London, London, UK
| | - Ai Nagano
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Busra Yaman
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Sheila Olendo Barasa
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Yewei Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| | - Pedro Cutillas
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - 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
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK
| | - Katiuscia Bianchi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, UK
| |
Collapse
|
5
|
Zhou Z, Qi J, Zhao J, Lim CW, Kim J, Kim B. Dual TBK1/IKKɛ inhibitor amlexanox attenuates the severity of hepatotoxin-induced liver fibrosis and biliary fibrosis in mice. J Cell Mol Med 2020; 24:1383-1398. [PMID: 31821710 PMCID: PMC6991653 DOI: 10.1111/jcmm.14817] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/23/2019] [Accepted: 10/26/2019] [Indexed: 12/31/2022] Open
Abstract
Although numerous studies have suggested that canonical IκB kinases (IKK) play a key role in the progression of liver fibrosis, the role of non-canonical IKKε and TANK-binding kinase 1 (TBK1) on the development and progression of liver fibrosis remains unclear. To demonstrate such issue, repeated injection of CCl4 was used to induce hepatotoxin-mediated chronic liver injury and biliary fibrosis was induced by 0.1% diethoxycarbonyl-1, 4-dihydrocollidine diet feeding for 4 weeks. Mice were orally administered with amlexanox (25, 50, and 100 mg/kg) during experimental period. Significantly increased levels of TBK1 and IKKε were observed in fibrotic livers or hepatic stellate cells (HSCs) isolated from fibrotic livers. Interestingly, amlexanox treatment significantly inhibited the phosphorylation of TBK1 and IKKε accompanied by reduced liver injury as confirmed by histopathologic analysis, decreased serum biochemical levels and fibro-inflammatory responses. Additionally, treatment of amlexanox promoted the fibrosis resolution. In accordance with these findings, amlexanox treatment suppressed HSC activation and its related fibrogenic responses by partially inhibiting signal transducer and activator of transcription 3. Furthermore, amlexanox decreased the activation and inflammatory responses in Kupffer cells. Collectively, we found that inhibition of the TBK1 and IKKε by amlexanox is a promising therapeutic strategy to cure liver fibrosis.
Collapse
Affiliation(s)
- Zixiong Zhou
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program)College of Veterinary MedicineJeonbuk National UniversityIksanKorea
| | - Jing Qi
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program)College of Veterinary MedicineJeonbuk National UniversityIksanKorea
| | - Jing Zhao
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program)College of Veterinary MedicineJeonbuk National UniversityIksanKorea
| | - Chae Woong Lim
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program)College of Veterinary MedicineJeonbuk National UniversityIksanKorea
| | - Jong‐Won Kim
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program)College of Veterinary MedicineJeonbuk National UniversityIksanKorea
| | - Bumseok Kim
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program)College of Veterinary MedicineJeonbuk National UniversityIksanKorea
| |
Collapse
|
6
|
Yin M, Wang X, Lu J. Advances in IKBKE as a potential target for cancer therapy. Cancer Med 2020; 9:247-258. [PMID: 31733040 PMCID: PMC6943080 DOI: 10.1002/cam4.2678] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 12/16/2022] Open
Abstract
IKBKE (inhibitor of nuclear factor kappa-B kinase subunit epsilon), a member of the nonclassical IKK family, plays an important role in the regulation of inflammatory reactions, activation and proliferation of immune cells, and metabolic diseases. Recent studies have demonstrated that IKBKE plays a crucial regulatory role in malignant tumor development. In recent years, IKBKE, an important oncoprotein in several kinds of tumors, has been widely found to regulate a variety of cytokines and signaling pathways. IKBKE promotes the growth, proliferation, invasion, and drug resistance of various cancers. This paper makes a detailed review that focuses on the recent discoveries of IKBKE in the malignant tumors, and puts forward that IKBKE is becoming an important therapeutic target for clinical treatment, which has been more and more realized.
Collapse
Affiliation(s)
- Min Yin
- Department of OncologyJinan Fifth People's HospitalJinanPR China
| | - Xin Wang
- Department of OncologyRenmin Hospital of Wuhan UniversityHubei ProvinceWuhanPR China
- Department of Radiation OncologyShandong Cancer Hospital Affiliated to Shandong UniversityShandong Academy of Medical ScienceJinanPR China
| | - Jie Lu
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical UniversityJinanPR China
| |
Collapse
|
7
|
He Q, Xia X, Yao K, Zeng J, Wang W, Wu Q, Tang R, Zou X. Amlexanox reversed non-alcoholic fatty liver disease through IKKε inhibition of hepatic stellate cell. Life Sci 2019; 239:117010. [DOI: 10.1016/j.lfs.2019.117010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/07/2019] [Accepted: 10/21/2019] [Indexed: 01/07/2023]
|
8
|
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
|
9
|
The human T-cell leukemia virus type-1 tax oncoprotein dissociates NF-κB p65 RelA-Stathmin complexes and causes catastrophic mitotic spindle damage and genomic instability. Virology 2019; 535:83-101. [PMID: 31299491 DOI: 10.1016/j.virol.2019.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 12/23/2022]
Abstract
Genomic instability is a hallmark of many cancers; however, the molecular etiology of chromosomal dysregulation is not well understood. The human T-cell leukemia virus type-1 (HTLV-1) oncoprotein Tax activates NF-κB-signaling and induces DNA-damage and aberrant chromosomal segregation through diverse mechanisms which contribute to viral carcinogenesis. Intriguingly, Stathmin/oncoprotein-18 (Op-18) depolymerizes tubulin and interacts with the p65RelA subunit and functions as a cofactor for NF-κB-dependent transactivation. We thus hypothesized that the dissociation of p65RelA-Stathmin/Op-18 complexes by Tax could lead to the catastrophic destabilization of microtubule (MT) spindle fibers during mitosis and provide a novel mechanistic link between NF-κB-signaling and genomic instability. Here we report that the inhibition of Stathmin expression by the retroviral latency protein, p30II, or knockdown with siRNA-stathmin, dampens Tax-mediated NF-κB transactivation and counters Tax-induced genomic instability and cytotoxicity. The Tax-G148V mutant, defective for NF-κB activation, exhibited reduced p65RelA-Stathmin binding and diminished genomic instability and cytotoxicity. Dominant-negative inhibitors of NF-κB also prevented Tax-induced multinucleation and apoptosis. Moreover, cell clones containing the infectious HTLV-1 ACH. p30II mutant provirus, impaired for p30II production, exhibited increased multinucleation and the accumulation of cytoplasmic tubulin aggregates following nocodozole-treatment. These findings allude to a mechanism whereby NF-κB-signaling regulates tubulin dynamics and mitotic instability through the modulation of p65RelA-Stathmin/Op-18 interactions, and support the notion that p30II enhances the survival of Tax-expressing HTLV-1-transformed cells.
Collapse
|
10
|
Shin CH, Choi DS. Essential Roles for the Non-Canonical IκB Kinases in Linking Inflammation to Cancer, Obesity, and Diabetes. Cells 2019; 8:cells8020178. [PMID: 30791439 PMCID: PMC6406369 DOI: 10.3390/cells8020178] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 12/17/2022] Open
Abstract
Non-canonical IκB kinases (IKKs) TBK1 and IKKε have essential roles as regulators of innate immunity and cancer. Recent work has also implicated these kinases in distinctively controlling glucose homeostasis and repressing adaptive thermogenic and mitochondrial biogenic response upon obesity-induced inflammation. Additionally, TBK1 and IKKε regulate pancreatic β-cell regeneration. In this review, we summarize current data on the functions and molecular mechanisms of TBK1 and IKKε in orchestrating inflammation to cancer, obesity, and diabetes.
Collapse
Affiliation(s)
- Chong Hyun Shin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
11
|
Curbing Lipids: Impacts ON Cancer and Viral Infection. Int J Mol Sci 2019; 20:ijms20030644. [PMID: 30717356 PMCID: PMC6387424 DOI: 10.3390/ijms20030644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 12/13/2022] Open
Abstract
Lipids play a fundamental role in maintaining normal function in healthy cells. Their functions include signaling, storing energy, and acting as the central structural component of cell membranes. Alteration of lipid metabolism is a prominent feature of cancer, as cancer cells must modify their metabolism to fulfill the demands of their accelerated proliferation rate. This aberrant lipid metabolism can affect cellular processes such as cell growth, survival, and migration. Besides the gene mutations, environmental factors, and inheritance, several infectious pathogens are also linked with human cancers worldwide. Tumor viruses are top on the list of infectious pathogens to cause human cancers. These viruses insert their own DNA (or RNA) into that of the host cell and affect host cellular processes such as cell growth, survival, and migration. Several of these cancer-causing viruses are reported to be reprogramming host cell lipid metabolism. The reliance of cancer cells and viruses on lipid metabolism suggests enzymes that can be used as therapeutic targets to exploit the addiction of infected diseased cells on lipids and abrogate tumor growth. This review focuses on normal lipid metabolism, lipid metabolic pathways and their reprogramming in human cancers and viral infection linked cancers and the potential anticancer drugs that target specific lipid metabolic enzymes. Here, we discuss statins and fibrates as drugs to intervene in disordered lipid pathways in cancer cells. Further insight into the dysregulated pathways in lipid metabolism can help create more effective anticancer therapies.
Collapse
|
12
|
Subramanian K, Dierckx T, Khouri R, Menezes SM, Kagdi H, Taylor GP, Farre L, Bittencourt A, Kataoka K, Ogawa S, Van Weyenbergh J. Decreased RORC expression and downstream signaling in HTLV-1-associated adult T-cell lymphoma/leukemia uncovers an antiproliferative IL17 link: A potential target for immunotherapy? Int J Cancer 2018; 144:1664-1675. [PMID: 30303535 PMCID: PMC6590643 DOI: 10.1002/ijc.31922] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 09/13/2018] [Accepted: 09/18/2018] [Indexed: 01/05/2023]
Abstract
Retinoic acid‐related drugs have shown promising pre‐clinical activity in Adult T‐cell Leukemia/Lymphoma, but RORC signaling has not been explored. Therefore, we investigated transcriptome‐wide interactions of the RORC pathway in HTLV‐1 and ATL, using our own and publicly available gene expression data for ATL and other leukemias. Gene expression data from ATL patients were analyzed using WGCNA to determine gene modules and their correlation to clinical and molecular data. Both PBMCs and CD4+ T‐Cells exhibited decreased RORC expression in four different ATL cohorts. A small subset of RORChi ATL patients was identified with significantly lower pathognomonic CADM1 and HBZ levels but similar levels of other ATL markers (CD4/CD25/CCR4), hinting at a less aggressive ATL subtype. An age‐dependent decrease in RORC expression was found in HTLV‐1‐infected individuals, but not in healthy controls, suggesting an early molecular event predisposing to leukemogenesis. Genes upstream of RORC signaling were members of a proliferative gene module (containing proliferation markers PCNA/Ki67), whereas downstream members clustered in an anti‐proliferative gene module. IL17C transcripts showed the strongest negative correlation to PCNA in both ATL cohorts, which was replicated in two large cohorts of T‐ and B‐cell acute lymphoid leukemia (ALL). Finally, IL17C expression in purified CD4 + CCR4 + CD26‐CD7‐ “ATL‐like” cells from HTLV‐1‐infected individuals and ATL patients was negatively correlated with clonality, underscoring a possible antileukemic/antiproliferative role. In conclusion, decreased RORC expression and downstream signaling might represent an early event in ATL pathogenesis. An antiproliferative IL17C/PCNA link is shared between ATL, T‐ALL and B‐ALL, suggesting (immuno)therapeutic benefit of boosting RORC/IL17 signaling. What's new? Drugs that affect the retinoic acid pathway are of interest for the treatment of adult T‐cell leukemia (ATL). Here, investigation of the role of retinoic acid‐related orphan receptor C (RORC), a regulator of the proinflammatory Th17/IL‐17 axis, reveals a prevailing occurrence of low RORC expression among ATL patients. By comparison, fewer patients exhibited a RORChi phenotype, which was associated with reduced levels of pathognomonic biomarkers CADM1 and HbZ, indicating a protective role for elevated RORC. An antiproliferative link was identified between RORC and IL17C. The data suggest that strategies to increase RORC/IL17C signaling could be important to improving ATL outcomes.
Collapse
Affiliation(s)
- Kritika Subramanian
- St. George's University School of MedicineUniversity CentreGrenadaWest Indies
- KU Leuven – University of Leuven, Department of Microbiology and ImmunologyRega Institute for Medical Research, Clinical and Epidemiological VirologyLeuvenBelgium
| | - Tim Dierckx
- KU Leuven – University of Leuven, Department of Microbiology and ImmunologyRega Institute for Medical Research, Clinical and Epidemiological VirologyLeuvenBelgium
| | - Ricardo Khouri
- KU Leuven – University of Leuven, Department of Microbiology and ImmunologyRega Institute for Medical Research, Clinical and Epidemiological VirologyLeuvenBelgium
- Instituto Gonçalo Moniz – FIOCRUZSalvadorBahiaBrazil
| | - Soraya Maria Menezes
- KU Leuven – University of Leuven, Department of Microbiology and ImmunologyRega Institute for Medical Research, Clinical and Epidemiological VirologyLeuvenBelgium
| | - Huseini Kagdi
- Department of MedicineImperial College LondonLondonUK
| | | | - Lourdes Farre
- Instituto Gonçalo Moniz – FIOCRUZSalvadorBahiaBrazil
| | | | - Keisuke Kataoka
- Department of Pathology and Tumor BiologyGraduate School of Medicine, Kyoto UniversityKyotoJapan
- Division of Molecular OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Seishi Ogawa
- Department of Pathology and Tumor BiologyGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Johan Van Weyenbergh
- KU Leuven – University of Leuven, Department of Microbiology and ImmunologyRega Institute for Medical Research, Clinical and Epidemiological VirologyLeuvenBelgium
| |
Collapse
|
13
|
Roles for the IKK-Related Kinases TBK1 and IKKε in Cancer. Cells 2018; 7:cells7090139. [PMID: 30223576 PMCID: PMC6162516 DOI: 10.3390/cells7090139] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 01/21/2023] Open
Abstract
While primarily studied for their roles in innate immune response, the IκB kinase (IKK)-related kinases TANK-binding kinase 1 (TBK1) and IKKε also promote the oncogenic phenotype in a variety of cancers. Additionally, several substrates of these kinases control proliferation, autophagy, cell survival, and cancer immune responses. Here we review the involvement of TBK1 and IKKε in controlling different cancers and in regulating responses to cancer immunotherapy.
Collapse
|
14
|
Fochi S, Mutascio S, Bertazzoni U, Zipeto D, Romanelli MG. HTLV Deregulation of the NF-κB Pathway: An Update on Tax and Antisense Proteins Role. Front Microbiol 2018; 9:285. [PMID: 29515558 PMCID: PMC5826390 DOI: 10.3389/fmicb.2018.00285] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/07/2018] [Indexed: 12/31/2022] Open
Abstract
Human T-cell lymphotropic virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia (ATL), an aggressive CD4+/CD25+ T-cell malignancy and of a severe neurodegenerative disease, HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP). The chronic activation or deregulation of the canonical and non-canonical nuclear factor kappa B (NF-κB) pathways play a crucial role in tumorigenesis. The HTLV-1 Tax-1 oncoprotein is a potent activator of the NF-κB transcription factors and the NF-κB response is required for promoting the development of HTLV-1 transformed cell lines. The homologous retrovirus HTLV-2, which also expresses a Tax-2 transforming protein, is not associated with ATL. In this review, we provide an updated synopsis of the role of Tax-1 in the deregulation of the NF-κB pathway, highlighting the differences with the homologous Tax-2. Special emphasis is directed toward the understanding of the molecular mechanisms involved in NF-κB activation resulting from Tax interaction with host factors affecting several cellular processes, such as cell cycle, apoptosis, senescence, cell proliferation, autophagy, and post-translational modifications. We also discuss the current knowledge on the role of the antisense viral protein HBZ in down-regulating the NF-κB activation induced by Tax, and its implication in cellular senescence. In addition, we review the recent studies on the mechanism of HBZ-mediated inhibition of NF-κB activity as compared to that exerted by the HTLV-2 antisense protein, APH-2. Finally, we discuss recent advances aimed at understanding the role exerted in the development of ATL by the perturbation of NF-κB pathway by viral regulatory proteins.
Collapse
Affiliation(s)
| | | | | | | | - Maria G. Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
15
|
Krause M, Foks H, Gobis K. Pharmacological Potential and Synthetic Approaches of Imidazo[4,5-b]pyridine and Imidazo[4,5-c]pyridine Derivatives. Molecules 2017; 22:molecules22030399. [PMID: 28273868 PMCID: PMC6155225 DOI: 10.3390/molecules22030399] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/02/2017] [Indexed: 12/23/2022] Open
Abstract
The structural resemblance between the fused imidazopyridine heterocyclic ring system and purines has prompted biological investigations to assess their potential therapeutic significance. They are known to play a crucial role in numerous disease conditions. The discovery of their first bioactivity as GABAA receptor positive allosteric modulators divulged their medicinal potential. Proton pump inhibitors, aromatase inhibitors, and NSAIDs were also found in this chemical group. Imidazopyridines have the ability to influence many cellular pathways necessary for the proper functioning of cancerous cells, pathogens, components of the immune system, enzymes involved in carbohydrate metabolism, etc. The collective results of biochemical and biophysical properties foregrounded their medicinal significance in central nervous system, digestive system, cancer, inflammation, etc. In recent years, new preparative methods for the synthesis of imidazopyridines using various catalysts have been described. The present manuscript to the best of our knowledge is the complete compilation on the synthesis and medicinal aspects of imidazo[4,5-b]pyridines and imidazo[4,5-c]pyridines reported from the year 2000 to date, including structure–activity relationships.
Collapse
Affiliation(s)
- Malwina Krause
- Department of Organic Chemistry, Medical University of Gdańsk, 107 Gen. Hallera Ave., 80-416 Gdańsk, Poland.
| | - Henryk Foks
- Department of Organic Chemistry, Medical University of Gdańsk, 107 Gen. Hallera Ave., 80-416 Gdańsk, Poland.
| | - Katarzyna Gobis
- Department of Organic Chemistry, Medical University of Gdańsk, 107 Gen. Hallera Ave., 80-416 Gdańsk, Poland.
| |
Collapse
|