1
|
Yang L, Gao Y, Lu J, Wu G. Aurora kinase A promotes epithelial‑mesenchymal transition by regulating P130 and P107 molecules in thyroid cancer cells. Exp Ther Med 2025; 29:93. [PMID: 40162054 PMCID: PMC11947869 DOI: 10.3892/etm.2025.12843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/20/2025] [Indexed: 04/02/2025] Open
Abstract
The mortality associated with thyroid cancer (THCA) has been increasing due to distant metastasis, yet the precise mechanisms remain unclear. The present study examined the role of Aurora kinase A (AURKA) in THCA cells. Reducing AURKA expression led to decreased cell proliferation and inhibited the transition of BHT101 and BCPAP cells from the G0 phase to active division. Interestingly, decreasing AURKA expression also enhanced the cells' ability to move, migrate and invade. It was found that AURKA regulates key molecules involved in cell proliferation. Specifically, reducing AURKA expression increased the levels of P130 and E2F4, while decreasing the level of P107. Furthermore, upregulating AURKA promoted epithelial-mesenchymal transition (EMT), whereas downregulating AURKA had the opposite effect. Blocking the focal adhesion kinase signaling pathway impaired the movement, migration and invasion capabilities of THCA cells, underscoring its crucial role in metastasis. In conclusion, AURKA promotes EMT by regulating P130 and P107, thereby facilitating the metastasis of THCA.
Collapse
Affiliation(s)
- Liyun Yang
- Department of Otolaryngology, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215600, P.R. China
- Department of Otolaryngology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, P.R. China
| | - Yuhuan Gao
- Medical College of Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Jing Lu
- Department of Otolaryngology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, P.R. China
| | - Geping Wu
- Department of Otolaryngology, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215600, P.R. China
| |
Collapse
|
2
|
Rekowska AK, Obuchowska K, Bartosik M, Kimber-Trojnar Ż, Słodzińska M, Wierzchowska-Opoka M, Leszczyńska-Gorzelak B. Biomolecules Involved in Both Metastasis and Placenta Accreta Spectrum-Does the Common Pathophysiological Pathway Exist? Cancers (Basel) 2023; 15:cancers15092618. [PMID: 37174083 PMCID: PMC10177254 DOI: 10.3390/cancers15092618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The process of epithelial-to-mesenchymal transition (EMT) is crucial in the implantation of the blastocyst and subsequent placental development. The trophoblast, consisting of villous and extravillous zones, plays different roles in these processes. Pathological states, such as placenta accreta spectrum (PAS), can arise due to dysfunction of the trophoblast or defective decidualization, leading to maternal and fetal morbidity and mortality. Studies have drawn parallels between placentation and carcinogenesis, with both processes involving EMT and the establishment of a microenvironment that facilitates invasion and infiltration. This article presents a review of molecular biomarkers involved in both the microenvironment of tumors and placental cells, including placental growth factor (PlGF), vascular endothelial growth factor (VEGF), E-cadherin (CDH1), laminin γ2 (LAMC2), the zinc finger E-box-binding homeobox (ZEB) proteins, αVβ3 integrin, transforming growth factor β (TGF-β), β-catenin, cofilin-1 (CFL-1), and interleukin-35 (IL-35). Understanding the similarities and differences in these processes may provide insights into the development of therapeutic options for both PAS and metastatic cancer.
Collapse
Affiliation(s)
- Anna K Rekowska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Karolina Obuchowska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Magdalena Bartosik
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Żaneta Kimber-Trojnar
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Magdalena Słodzińska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | | | | |
Collapse
|
3
|
Biswas S, Mahapatra E, Das S, Roy M, Mukherjee S. PEITC: A resounding molecule averts metastasis in breast cancer cells in vitro by regulating PKCδ/Aurora A interplay. Heliyon 2022; 8:e11656. [PMID: 36458309 PMCID: PMC9706142 DOI: 10.1016/j.heliyon.2022.e11656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 01/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Background/aim Intricate association and aberrant activation of serine/threonine kinase (STK) family proteins like Polo-like kinase (PLK1) and Aurora kinase (Aurora A abruptly regulate mitotic entry whereas activation of PKCδ), another important member of STK family conversely induces apoptosis which is preceded by cell cycle arrest. These STKs are considered as major determinant of oncogenicity. Therefore, the contributory role of Aurora A/PLK-1 axis in mitotic control and PKCδ in apoptosis control and their reciprocity in cancer research is an emerging area to explore. The present study investigated the intricate involvement of STKs in breast cancer cells (MCF-7 and MDA-MB-231) and their disruption by PEITC. Methods Both MCF-7 and MDA-MB-231 cells were checked for clonogenic assay, cell-cycle analysis and the results were compared with normal MCF-10A, Western blotting, TUNEL & DNA-fragmentation assay, wound healing, transwell migration assays in presence and absence of PEITC. Results PEITC was found to increase the expression of PKCδ with subsequent nuclear translocation. Nuclear translocation of PKCδ was accompanied by inhibition of nuclear lamin vis a vis phosphorylation of Nrf2 at Ser 40 alongside nuclear accumulation of phospho-Nrf2. Activated PKCδ furthermore exerted its apoptotic effect by negatively regulating Aurora A and consequentially PLK1; indicating activation of PLK1 by Aurora A. Involvement of PEITC induced PKCδ activation and Aurora A inhibition was ascertained by using Rottlerin/Aurora A Inhibitor. Discussion & conclusion Natural isothiocyanates like PEITC efficiently altered the functional abilities of STKs concerning their entangled functional interplay. Such alterations in protein expression by PEITC was chaperoned with inhibition of the aggressiveness of breast cancer cells and ultimately induction of apoptosis.
Collapse
Affiliation(s)
- Souvick Biswas
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| | - Elizabeth Mahapatra
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| | - Salini Das
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| | - Madhumita Roy
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| | - Sutapa Mukherjee
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| |
Collapse
|
4
|
AKTAN Ç. Identification of Ferroptosis-Related Genes in Laryngeal Carcinoma Using an Integrated Bioinformatics Approach. İSTANBUL GELIŞIM ÜNIVERSITESI SAĞLIK BILIMLERI DERGISI 2022. [DOI: 10.38079/igusabder.1128423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Amaç: Hücre içi demir birikimi ve lipid peroksidasyonu ile karakterize edilen ferroptoz, tümör baskılanmasında önemli rol oynayabilen yeni tanımlanmış bir hücre ölüm şeklidir. Larengeal skuamöz hücreli karsinom (LSHK) ve ferroptozis arasındaki ilişki hakkında yapılan çalışmalar sınırlıdır. Bu çalışmanın amacı, LSHK' nin tanı, tedavisinde ve ferroptozis ile ilgili belirteçleri in siliko yöntemleri kullanarak saptamaktır.Yöntem: Ferroptoz ile ilgili genler, FerrDb veri tabanından elde edildi. The Cancer Genome Atlas (TCGA) veri setlerinden LSHK hastalarının mRNA ekspresyon verileri ve ferroptoz ile ilgili bazı genleri taramak için kullanıldı. LSHK ile ilgili GSE143224 ve GSE84957 mikrodizi veri setleri GEO veri tabanından elde edilmiştir. Tüm veri setleri kullanılarak ferroptoz ve LSHK ile ilişkili genleri elde etmek için örtüşen veriler kullanılmıştır. LSHK grubu ve normal kontroller arasındaki diferansiyel olarak eksprese edilen genler (DEG'ler) ve ferroptoz ile ilgili DEG'ler, biyoinformatik yöntemler kullanılarak analiz edildi. Daha sonra STRING ve Cytoscape yazılımları kullanılarak Gene Ontology (GO), KEGG ve protein-protein etkileşimi (PPE) ağı analizleri gerçekleştirilmiştir.Bulgular: Ferroptoz ile ilgili 259 gen, FerrDb veri tabanından alındı ve ferroptoz DEG'lerini tanımlamak için bunları TCGA-HNSC (523 örnek), GSE143224 (25 örnek) ve GSE84957 (18 örnek) ile analizleri yapıldı. Analiz sonrasında 13 adet yukarı regüle edilmiş (NOX4, BID, ABCC1, TNFAIP3, PANX1, SLC1A4, SLC3A2, FTL, TFRC, AURKA, HSF1, PML, CA9; p<0.05) ve 3 adet aşağı regüle edilmiş gen (CHAC1, LPIN1, MUC1; p<0.05) saptanmıştır. GO, KEGG ve PPE analizleri ile elde edilen hücresel stres, inflamasyon, oksidatif stres ve karsinogenez süreçlerine benzer sonuçlar (p<0.05) ile bu genlerin LSHK' nin ilerlemesinde rol oynayabileceğini göstermektedir.Sonuç: Sonuç olarak, bu çalışmada LSHK'de ferroptoz ile yakından ilişkili olan ve LSHK hastalarını sağlıklı kontrollerden ayırt edebilen 16 potansiyel gen saptanmıştır. Çalışmamız, LSHK’nin moleküler mekanizmasını ve terapötik hedeflerini keşfetmek için daha geniş bir fikir sağlayabilir.
Collapse
|
5
|
Huang D, Qiu H, Miao L, Guo L, Zhang X, Lin M, Li Z, Li F. Cdc42 promotes thyroid cancer cell proliferation and migration and tumor-associated macrophage polarization through the PTEN/AKT pathway. J Biochem Mol Toxicol 2022; 36:e23115. [PMID: 35822655 DOI: 10.1002/jbt.23115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 04/21/2022] [Accepted: 05/18/2022] [Indexed: 11/09/2022]
Abstract
The purpose of this study was to investigate the potential mechanism and function of Cdc42 in thyroid cancer. We found that knockdown of Cdc42 inhibited the migration and proliferation of WRO cells. This role of Cdc42 is achieved by interacting with PTEN and interfering with its PTEN nuclear translocation. The overexpression of Cdc42 enhances the production of lactic acid and promotes the polarization of M2 macrophages, and therefore M2 macrophages inhibit the function of T cells. Overall, Cdc42 can promote cell proliferation and migration through the PTEN/AKT pathway and promote tumor-related M2 macrophage polarization and inhibit T cell activity by enhancing aerobic glycolysis, animal experiments confirmed that tumor volume increased after Cdc42 overexpressed in TBP-3743 murine thyroid cancer cells. Increased infiltration of Treg and macrophages was also observed. taken together, our results indicate that Cdc42 can be used as a diagnostic and thyroid cancer Prognostic biomarkers and potential therapeutic targets.
Collapse
Affiliation(s)
- Deyi Huang
- Ultrasound Department, The People's Hospital of Yuhuan, Yuhuan, China
| | - Huali Qiu
- Ultrasound Department, The People's Hospital of Yuhuan, Yuhuan, China
| | - Lin Miao
- Thyroid Breast Surgery Department, The People's Hospital of Yuhuan, Yuhuan, China
| | - Lin Guo
- Inspection Department, The People's Hospital of Yuhuan, Yuhuan, China
| | - Xiaoting Zhang
- Ultrasound Department, The People's Hospital of Yuhuan, Yuhuan, China
| | - Mengmeng Lin
- Ultrasound Department, The People's Hospital of Yuhuan, Yuhuan, China
| | - Zhongyun Li
- Ultrasound Department, The People's Hospital of Yuhuan, Yuhuan, China
| | - Fang Li
- Ultrasound Department, The People's Hospital of Yuhuan, Yuhuan, China
| |
Collapse
|
6
|
Guo Z, Liu X, Shao H. E2F4-induced AGAP2-AS1 up-regulation accelerates the progression of colorectal cancer via miR-182-5p/CFL1 axis. Dig Liver Dis 2022; 54:878-889. [PMID: 34838479 DOI: 10.1016/j.dld.2021.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are closely associated with the pathogenesis of numerous diseases including cancers. LncRNA AGAP2 Antisense RNA 1 (AGAP2-AS1) has been found to participate in the tumorigenesis of several kinds of human cancers. Nonetheless, its potential function in colorectal cancer (CRC) was still poorly investigated. METHODS The expression level of RNAs or proteins was assessed by RT-qPCR or western blot analysis. Functional experiments were performed to analyze the role of AGAP2-AS1 in CRC in vitro and in vivo. Mechanism investigations were fulfilled to determine the potential mechanism of the molecules. RESULTS AGAP2-AS1 expression was significantly elevated in CRC cells and could be transcriptionally activated by E2F Transcription Factor 4 (E2F4). Down-regulated AGAP2-AS1 could weaken CRC cell growth, migration, invasion, and epithelial-mesenchymal transition (EMT). MicroRNA-182-5p (miR-182-5p) was the target downstream molecule of AGAP2-AS1. Furthermore, Cofilin 1 (CFL1) was proved as the target of miR-182-5p. Mechanically, AGAP2-AS1 could boost the CFL1 expression via competitively binding to miR-182-5p in CRC. Importantly, CFL1 restoration could counteract the in vitro and in vivo suppression of depleted AGAP2-AS1 on CRC progression. CONCLUSION E2F4-stimulated AGAP2-AS1 aggravated CRC development through regulating miR-182-5p/CFL1 axis, implying that AGAP2-AS1 might become a potent new target for future therapies for CRC.
Collapse
Affiliation(s)
- Zhen Guo
- Gastrointestinal Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Xuezhong Liu
- Gastrointestinal Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Hongjin Shao
- Anorectal Department, Liaocheng People's Hospital, NO.67 Dongchang West Road, Dongchangfu District, Liaocheng, Shandong 252000, China.
| |
Collapse
|
7
|
Kim CH, Kim DE, Kim DH, Min GH, Park JW, Kim YB, Sung CK, Yim H. Mitotic protein kinase-driven crosstalk of machineries for mitosis and metastasis. Exp Mol Med 2022; 54:414-425. [PMID: 35379935 PMCID: PMC9076678 DOI: 10.1038/s12276-022-00750-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence indicates that mitotic protein kinases are involved in metastatic migration as well as tumorigenesis. Protein kinases and cytoskeletal proteins play a role in the efficient release of metastatic cells from a tumor mass in the tumor microenvironment, in addition to playing roles in mitosis. Mitotic protein kinases, including Polo-like kinase 1 (PLK1) and Aurora kinases, have been shown to be involved in metastasis in addition to cell proliferation and tumorigenesis, depending on the phosphorylation status and cellular context. Although the genetic programs underlying mitosis and metastasis are different, the same protein kinases and cytoskeletal proteins can participate in both mitosis and cell migration/invasion, resulting in migratory tumors. Cytoskeletal remodeling supports several cellular events, including cell division, movement, and migration. Thus, understanding the contributions of cytoskeletal proteins to the processes of cell division and metastatic motility is crucial for developing efficient therapeutic tools to treat cancer metastases. Here, we identify mitotic kinases that function in cancer metastasis as well as tumorigenesis. Several mitotic kinases, namely, PLK1, Aurora kinases, Rho-associated protein kinase 1, and integrin-linked kinase, are considered in this review, as an understanding of the shared machineries between mitosis and metastasis could be helpful for developing new strategies to treat cancer. Improving understanding of the mechanisms linking cell division and cancer spread (metastasis) could provide novel strategies for treatment. A group of enzymes involved in cell division (mitosis) are also thought to play critical roles in the spread of cancers. Hyungshin Yim at Hanyang University in Ansan, South Korea, and co-workers in Korea and the USA reviewed the roles of several mitotic enzymes that are connected with metastasis as well as tumorigenesis. They discussed how these enzymes modify cytoskeletal proteins and other substrates during cancer progression. Some regulatory control of cell cytoskeletal structures is required for cancer cells to metastasize. Recent research has uncovered crosstalk between mitotic enzymes and metastatic cytoskeletal molecules in various cancers. Targeting mitotic enzymes and the ways they influence cytoskeletal mechanisms could provide valuable therapeutic strategies for suppressing metastasis.
Collapse
Affiliation(s)
- Chang-Hyeon Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Da-Eun Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Dae-Hoon Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Ga-Hong Min
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Jung-Won Park
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Yeo-Bin Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea
| | - Chang K Sung
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, Kingsville, TX, 78363, USA
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, 15588, Korea.
| |
Collapse
|
8
|
Ling Z, Long X, Wu Y, Li J, Feng M. LMO3 promotes proliferation and metastasis of papillary thyroid carcinoma cells by regulating LIMK1-mediated cofilin and the β-catenin pathway. Open Med (Wars) 2022; 17:453-462. [PMID: 35350839 PMCID: PMC8919831 DOI: 10.1515/med-2022-0419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/21/2021] [Accepted: 12/07/2021] [Indexed: 11/15/2022] Open
Abstract
Abstract
LIM domain only 3 (LMO3) interacts with transcription factors to regulate target genes involved in embryonic development. The oncogenic role of LMO3 in hepatocellular carcinoma, gastric cancer, and neuroblastoma has been reported recently. However, little is known about the biological function of LMO3 in papillary thyroid carcinoma (PTC). First, expression of LMO3 was dramatically enhanced in the PTC tissues and cell lines. Second, knockdown of LMO3 in PTC cells repressed cell proliferation and promoted cell apoptosis with downregulated Bcl-2 and upregulated cleaved caspase-3/PARP. In vitro cell migration and invasion of PTC were also retarded by siRNA-mediated silence of LMO3. Third, protein expression of LIM kinase (LIMK) 1-mediated phosphorylation of cofilin and nuclear translocation of β-catenin were reduced by the knockdown of LMO3. pcDNA-mediated overexpression of LIMK1 promoted cofilin phosphorylation and attenuated LMO3 silence-induced decrease of cofilin phosphorylation. Last, enhanced LIMK1 expression promoted PTC cell proliferation and metastasis and counteracted the suppressive effects of LMO3 silence on PTC cell proliferation and metastasis. In conclusion, LMO3 promoted PTC cell proliferation and metastasis by regulating LIMK1-mediated cofilin and the β-catenin pathway.
Collapse
Affiliation(s)
- Zeyi Ling
- Department of Otorhinolaryngology Head and Neck Surgery, Yongchuan Hospital of Chongqing Medical University , Chongqing , 402160 , China
| | - Xiaoli Long
- Department of Geriatrics, Yongchuan Hospital of Chongqing Medical University , No. 439, Xuanhua Road, Yongchuan District , Chongqing , 402160 , China
| | - Ying Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Yongchuan Hospital of Chongqing Medical University , Chongqing , 402160 , China
| | - Jie Li
- Department of Otorhinolaryngology Head and Neck Surgery, Yongchuan Hospital of Chongqing Medical University , Chongqing , 402160 , China
| | - Mingliang Feng
- Department of Otorhinolaryngology Head and Neck Surgery, Yongchuan Hospital of Chongqing Medical University , Chongqing , 402160 , China
| |
Collapse
|
9
|
Baltzer S, Bulatov T, Schmied C, Krämer A, Berger BT, Oder A, Walker-Gray R, Kuschke C, Zühlke K, Eichhorst J, Lehmann M, Knapp S, Weston J, von Kries JP, Süssmuth RD, Klussmann E. Aurora Kinase A Is Involved in Controlling the Localization of Aquaporin-2 in Renal Principal Cells. Int J Mol Sci 2022; 23:ijms23020763. [PMID: 35054947 PMCID: PMC8776063 DOI: 10.3390/ijms23020763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/30/2021] [Accepted: 01/08/2022] [Indexed: 02/01/2023] Open
Abstract
The cAMP-dependent aquaporin-2 (AQP2) redistribution from intracellular vesicles into the plasma membrane of renal collecting duct principal cells induces water reabsorption and fine-tunes body water homeostasis. However, the mechanisms controlling the localization of AQP2 are not understood in detail. Using immortalized mouse medullary collecting duct (MCD4) and primary rat inner medullary collecting duct (IMCD) cells as model systems, we here discovered a key regulatory role of Aurora kinase A (AURKA) in the control of AQP2. The AURKA-selective inhibitor Aurora-A inhibitor I and novel derivatives as well as a structurally different inhibitor, Alisertib, prevented the cAMP-induced redistribution of AQP2. Aurora-A inhibitor I led to a depolymerization of actin stress fibers, which serve as tracks for the translocation of AQP2-bearing vesicles to the plasma membrane. The phosphorylation of cofilin-1 (CFL1) inactivates the actin-depolymerizing function of CFL1. Aurora-A inhibitor I decreased the CFL1 phosphorylation, accounting for the removal of the actin stress fibers and the inhibition of the redistribution of AQP2. Surprisingly, Alisertib caused an increase in actin stress fibers and did not affect CFL1 phosphorylation, indicating that AURKA exerts its control over AQP2 through different mechanisms. An involvement of AURKA and CFL1 in the control of the localization of AQP2 was hitherto unknown.
Collapse
Affiliation(s)
- Sandrine Baltzer
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Timur Bulatov
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Christopher Schmied
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
- DKTK (German Translational Research Network), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
| | - Benedict-Tilman Berger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
| | - Andreas Oder
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Ryan Walker-Gray
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Christin Kuschke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Kerstin Zühlke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Jenny Eichhorst
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
- DKTK (German Translational Research Network), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, 60596 Frankfurt am Main, Germany
| | - John Weston
- JQuest Consulting, Carl-Orff-Weg 25, 65779 Kelkheim, Germany;
| | - Jens Peter von Kries
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Roderich D. Süssmuth
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Enno Klussmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence: ; Tel.: +49-30-9406-2596
| |
Collapse
|
10
|
Huang Y, Xie Z, Li X, Chen W, He Y, Wu S, Li X, Hou B, Sun J, Wang S, He Y, Jiang H, Lun Y, Zhang J. Development and validation of a ferroptosis-related prognostic model for the prediction of progression-free survival and immune microenvironment in patients with papillary thyroid carcinoma. Int Immunopharmacol 2021; 101:108156. [PMID: 34624650 DOI: 10.1016/j.intimp.2021.108156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/31/2021] [Accepted: 09/11/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Ferroptosis is an iron-dependent and regulated cell death that has been widely reported in a variety of malignancies. The overall survival of papillary thyroid cancer (PTC) is excellent, but the identification of patients with poor prognosis still faces challenges. Nevertheless, whether ferroptosis-related genes (FRGs) can be used to screen high-risk patients is not clear. METHODS We obtained the clinical data of patients with PTC and FRGs from the UCSC Xena platform and the FerrDb respectively. Differentially expressed genes (DEGs) of FRGs were obtained from the entire The Cancer Genome Atlas (TCGA). Subsequently, the entire TCGA dataset was randomly split into two subsets: training and test datasets. Based on DEGs, we constructed a predictive model which was tested in the test dataset and the entire TCGA dataset to predict progression-free survival (PFS). Patients were categorized into high- or low-risk groups based on their median risk score. We analyzed differences in some aspects, including pathway enrichment analysis, single-sample Gene Set Enrichment Analysis (ssGSEA), tumor microenvironment (TME), human leukocyte antigen (HLA) genes, and tumor mutation burden (TMB) analyses, between high-risk and low-risk groups. RESULTS A predictive model with three FRGs (HSPA5, AURKA, and TSC22D3) was constructed. Patients in the high-risk group had worse PFS compared with patients in the low-risk group. Functional analysis results revealed that ssGSEA, immune cell infiltration, TME, HLA, and TMB were closely associated with ferroptosis. CONCLUSION The prognostic model constructed in this study can effectively predict PFS for patients with PTC.
Collapse
Affiliation(s)
- Yinde Huang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Zhenyu Xie
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Xin Li
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Wenbin Chen
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Yuzhen He
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Song Wu
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Xinyang Li
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Bingchen Hou
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Jianjian Sun
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Shiyue Wang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Yuchen He
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Han Jiang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Yu Lun
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Jian Zhang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
11
|
Lin X, Xiang X, Feng B, Zhou H, Wang T, Chu X, Wang R. Targeting Long Non-Coding RNAs in Hepatocellular Carcinoma: Progress and Prospects. Front Oncol 2021; 11:670838. [PMID: 34249710 PMCID: PMC8267409 DOI: 10.3389/fonc.2021.670838] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/07/2021] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma is the fifth-ranked cancer worldwide with a relatively low five-year survival rate. Long non-coding RNAs are a group of RNAs with remarkable aberrant expression which could act on multiple bioprocesses and ultimately impact upon tumor proliferation, invasion, migration, metastasis, apoptosis, and therapy resistance in cancer cells including hepatocellular carcinoma cells. In recent years, long non-coding RNAs have been reported to be indispensable targets in clinical target therapy to stop the growth of cancer and prolong the lifespan of patients with hepatocellular carcinoma. In this review, we enumerate the signaling pathways and life activities affected by long non-coding RNAs in hepatocellular carcinoma cells to illustrate the role of long non-coding RNAs in the development and therapy resistance of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xinrong Lin
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaosong Xiang
- Affiliated Jingling Hospital Research Institution of General Surgery, School of Medicine, Nanjing University, Nanjing, China
| | - Bing Feng
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hao Zhou
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ting Wang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaoyuan Chu
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Rui Wang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
12
|
Xu M, Zhou J, Zhang Q, Le K, Xi Z, Yi P, Zhao X, Tan J, Huang T. MiR-3121-3p promotes tumor invasion and metastasis by suppressing Rap1GAP in papillary thyroid cancer in vitro. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1229. [PMID: 33178761 PMCID: PMC7607113 DOI: 10.21037/atm-20-4469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Rap1GAP is a tumor suppressor and is downregulated in human malignancies including papillary thyroid cancer (PTC). The mechanism of its suppression in PTC remains unclear. Methods Bioinformatic analyses were carried out to evaluate clinical significance and to predict upstream miRNA bindings of Rap1GAP. Three PTC cell lines, TPC-1, B-CPAP, and K1, were employed for functional verification and further experiments. We used dual-luciferase reporter gene assay to confirm the miRNA binding prediction, Western blotting and quantitative polymerase chain reaction (qPCR) to explore miRNA and Rap1GAP regulation, Transwell and wound healing assays to compare cell migration and invasion after protein knockout or overexpression, and Cell Counting Kit-8 (CCK-8) assay to evaluate cell proliferation. Results Rap1GAP expression was suppressed in thyroid cancer compared to adjacent normal tissues and was a potential diagnostic marker of PTC. Rap1GAP suppression was correlated to younger age, advanced T stage, N stage, extrathyroidal extension, BRAF-like tumors, and higher risk of recurrence. Combined analysis of bioinformatic prediction and dual-luciferase assay revealed binding between miR-3121-3p with 3'UTR of Rap1GAP promoter. MiR-3121-3p promoted cell migration, invasion, and proliferation via inhibiting Rap1GAP and thus upregulating MAPK pathway. Overexpression and knockdown of Rap1GAP could counteract the influence on cell migration and invasion carried out by miR-3121-3p mimic and inhibitor, respectively. Rap1GAP partially impaired the effect of miR-3121-3p in cell growth in the CCK-8 assay. Conclusions Rap1GAP expression is suppressed in PTC and is a potential diagnostic marker. Its upstream regulator, miR-3121-3p, affects tumor metastasis and proliferation via regulating Rap1GAP expression. MAPK signaling pathway may be involved in this effect.
Collapse
Affiliation(s)
- Ming Xu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiulei Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kehao Le
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihan Xi
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengfei Yi
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangwang Zhao
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Tan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Hirokawa YS, Kanayama K, Kagaya M, Shimojo N, Uchida K, Imai H, Ishii K, Watanabe M. SOX11-induced decrease in vimentin and an increase in prostate cancer cell migration attributed to cofilin activity. Exp Mol Pathol 2020; 117:104542. [PMID: 32971115 DOI: 10.1016/j.yexmp.2020.104542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/13/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022]
Abstract
SOX11 is a transcription factor in the SOX family of genes that regulate multiple cellular events by influencing the expression of key genes in developmental, physiological, and tumorigenic cells. To elucidate the role of SOX11 in prostate cancer cells, PC-3 prostate cancer cells were cloned (S6 and S9 cells) to highly express SOX11. We demonstrated that both S6 and S9 lose vimentin expression, acquiring epithelial marker proteins, which indicates the Epithelial state phenotype. S6 and S9 cells have cancer-promoting characteristics that include higher migratory properties compared with control cells. The mechanisms that are responsible for the enhanced migration are cofilin activity and keratin 18 expression. TCGA (The Cancer Genome Atlas) dataset analysis revealed that metastatic prostate cancer tumors tend to have more SOX11 gene amplification compared with primary tumors. These results suggest the tumor promotive role and epithelial protein induction of SOX11 in prostate cancer cell.
Collapse
Affiliation(s)
- Yoshifumi S Hirokawa
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Kazuki Kanayama
- Department of Clinical Nutrition, Suzuka University of Medical Science, Suzuka, Mie 510-0293, Japan
| | - Michiko Kagaya
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Naoshi Shimojo
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Katsunori Uchida
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hiroshi Imai
- Pathology Division, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Kenichiro Ishii
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masatoshi Watanabe
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
14
|
Melo-Hanchuk TD, Martins MB, Cunha LL, Soares FA, Ward LS, Vassallo J, Kobarg J. Expression of the NEK family in normal and cancer tissue: an immunohistochemical study. BMC Cancer 2020; 20:23. [PMID: 31906878 PMCID: PMC6945616 DOI: 10.1186/s12885-019-6408-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The NEK serine/threonine protein kinases are involved in cell cycle checkpoints, DNA damage repair, and apoptosis. Alterations in these pathways are frequently associated with cell malignant cellular transformations. Thyroid cancer is the most common malignant tumour in the endocrine system. Despite good treatment methods, the number of cases has increased significantly in recent years. Here, we studied the expression of NEK1, NEK2, NEK3, and NEK5 in different types of normal and malignant tissues, using tissue microarray analysis, and identified NEKs as potential markers in thyroid malignancy. METHODS The studied cases comprised multiple cancer tissue microarrays, including breast, colon, esophagus, kidney, lung, pancreas, prostate, stomach, thyroid and uterine cervix, as well as 281 patients who underwent thyroid resection for thyroid cancer or thyroid nodules. The expression of NEK1, NEK2, NEK3, and NEK5 was analyzed by immunohistochemistry. The expression pattern was evaluated in terms of intensity by two methods, semiquantitative and quantitative, and was compared between normal and cancer tissue. RESULTS We analysed the expression of each member of the NEK family in a tissue-dependent manner. Compared to normal tissue, most of the evaluated proteins showed lower expression in lung tumour. However, in the thyroid, the expression was higher in malignant tissue, especially for NEK 1, NEK3 and NEK5. Concerning characteristics of the thyroid tumour, such as aggressiveness, NEK1 expression was higher in tumours with multifocality and in patients with lymph node metastasis. NEK3 expression was stronger in patients with stage II, that involved metastasis. NEK5, on the other hand, showed high expression in patients with invasion and metastasis and in patients with tumour size > 4 cm. Furthermore, this work, demonstrated for the first time a high specificity and sensitivity of over-expression of NEK1 in classical and follicular variants of papillary thyroid cancer and NEK3 in tall-cell papillary thyroid cancer. CONCLUSION Taken together, the NEK protein kinases emerge as important proteins in thyroid cancer development and may help to identify malignancy and aggressiveness features during diagnosis. TRIAL REGISTRATION This study was retrospectively registered. www.accamargo.org.br/cientistas-pesquisadores/comite-de-etica-em-pequisa-cep.
Collapse
Affiliation(s)
- Talita Diniz Melo-Hanchuk
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Mariana Bonjiorno Martins
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Lucas Leite Cunha
- Laboratório de Genética Molecular do Câncer, Faculdade de Ciências Médicas Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | | | - Laura Sterian Ward
- Laboratório de Genética Molecular do Câncer, Faculdade de Ciências Médicas Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - José Vassallo
- Departamento de Anatomia Patológica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Jörg Kobarg
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil. .,Faculdade de Ciências Farmacêuticas-UNICAMP, Universidade Estadual de Campinas, Campinas, Inst. de Biologia, Dep. Bioquímica e Biologia Tecidual, Rua Monteiro Lobato 255, CEP 13083-862, Campinas-SP, Brazil.
| |
Collapse
|
15
|
Shen ZT, Chen Y, Huang GC, Zhu XX, Wang R, Chen LB. Aurora-a confers radioresistance in human hepatocellular carcinoma by activating NF-κB signaling pathway. BMC Cancer 2019; 19:1075. [PMID: 31703572 PMCID: PMC6842208 DOI: 10.1186/s12885-019-6312-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 10/30/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Radiotherapy failure is a significant clinical challenge due to the development of resistance in the course of treatment. Therefore, it is necessary to further study the radiation resistance mechanism of HCC. In our early study, we have showed that the expression of Aurora-A mRNA was upregulated in HCC tissue samples or cells, and Aurora-A promoted the malignant phenotype of HCC cells. However, the effect of Aurora-A on the development of HCC radioresistance is not well known. METHODS In this study, colony formation assay, MTT assays, flow cytometry assays, RT-PCR assays, Western blot, and tumor xenografts experiments were used to identify Aurora-A promotes the radioresistance of HCC cells by decreasing IR-induced apoptosis in vitro and in vivo. Dual-luciferase reporter assay, MTT assays, flow cytometry assays, and Western blot assay were performed to show the interactions of Aurora-A and NF-κB. RESULTS We established radioresistance HCC cell lines (HepG2-R) and found that Aurora-A was significantly upregulated in those radioresistant HCC cells in comparison with their parental HCC cells. Knockdown of Aurora-A increased radiosensitivity of radioresistant HCC cells both in vivo and in vitro by enhancing irradiation-induced apoptosis, while upregulation of Aurora-A decreased radiosensitivity by reducing irradiation-induced apoptosis of parental cells. In addition, we have showed that Aurora-A could promote the expression of nuclear IkappaB-alpha (IκBα) protein while enhancing the activity of NF-kappaB (κB), thereby promoted expression of NF-κB pathway downstream effectors, including proteins (Mcl-1, Bcl-2, PARP, and caspase-3), all of which are associated with apoptosis. CONCLUSIONS Aurora-A reduces radiotherapy-induced apoptosis by activating NF-κB signaling, thereby contributing to HCC radioresistance. Our results provided the first evidence that Aurora-A was essential for radioresistance in HCC and targeting this molecular would be a potential strategy for radiosensitization in HCC.
Collapse
Affiliation(s)
- Ze-Tian Shen
- Department of Radiation Oncology, Jinling Hospital, Nanjing Medical School University, Nanjing, Jiangsu, China
| | - Ying Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Gui-Chun Huang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Xi-Xu Zhu
- Department of Radiation Oncology, Jinling Hospital, Nanjing Medical School University, Nanjing, Jiangsu, China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China.
| | - Long-Bang Chen
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical School University, Nanjing, Jiangsu, China.
| |
Collapse
|
16
|
Giardino E, Catalano R, Barbieri AM, Treppiedi D, Mangili F, Spada A, Arosio M, Mantovani G, Peverelli E. Cofilin is a mediator of RET-promoted medullary thyroid carcinoma cell migration, invasion and proliferation. Mol Cell Endocrinol 2019; 495:110519. [PMID: 31352037 DOI: 10.1016/j.mce.2019.110519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/27/2019] [Accepted: 07/24/2019] [Indexed: 11/20/2022]
Abstract
Medullary thyroid carcinoma (MTC) is a rare neuroendocrine tumor that originates from parafollicular thyroid C cells and accounts for 5% of thyroid cancers. In inherited cases of MTC, and in about 40% of sporadic cases, activating mutations of the receptor tyrosine kinase proto-oncogene RET are found. Constitutively active RET triggers signaling pathways involved in cell proliferation, survival and motility, but the mechanisms underlying malignant transformation of C-cells have been only partially elucidated. Cofilin is a key regulator of actin cytoskeleton dynamics. A crucial role of cofilin in tumor development, progression, invasion and metastasis has been demonstrated in different human cancers, but no data are available in MTC. Interestingly, RET activation upregulates cofilin gene expression. The aim of this study was to investigate cofilin contribution in invasiveness and growth of MTC cells, and its relevance in the context of mutant RET signaling. We found that cofilin transfection in human MTC cell line TT significantly increased migration (178 ± 44%, p < 0.001), invasion (165 ± 28%, p < 0.01) and proliferation (146 ± 18%, p < 0.001), accompanied by an increase of ERK1/2 phosphorylation (2.23-fold) and cyclin D1 levels (1.43-fold). Accordingly, all these responses were significantly reduced after genetic silencing of cofilin (-55 ± 10% migration, p < 0.001, -41 ± 8% invasion, p < 0.001, -17 ± 3% proliferation, p < 0.001). These results have been confirmed in primary cells cultures obtained from human MTCs. The inhibition of constitutively active RET in TT cells by both the RET pharmacological inhibitor RPI-1 and the transfection of dominant negative RET mutant (RETΔTK) resulted in a reduction of cofilin expression (-37 ± 8%, p < 0.001 and -31 ± 16%, p < 0.01, respectively). Furthermore, RPI-1 inhibitory effects on TT cell migration (-57 ± 13%, p < 0.01), but not on cell proliferation, were completely abolished in cells transfected with cofilin. In conclusion, these data indicate that an unbalanced cofilin expression, induced by oncogenic RET, contributes to promote MTC invasiveness and growth, suggesting the possibility of targeting cofilin pathway for more effective treatment of MTC.
Collapse
Affiliation(s)
- E Giardino
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - R Catalano
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; PhD Program in Endocrinological Sciences, Sapienza University of Rome, Rome, Italy
| | - A M Barbieri
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - D Treppiedi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - F Mangili
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - A Spada
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - M Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - G Mantovani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - E Peverelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
17
|
Yi L, Hu N, Mu H, Sun J, Yin J, Dai K, Xu F, Yang N, Ding D. Identification of Cofilin-1 and Destrin as Potential Early-warning Biomarkers for Gamma Radiation in Mouse Liver Tissues. HEALTH PHYSICS 2019; 116:749-759. [PMID: 30913056 DOI: 10.1097/hp.0000000000001012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Gamma radiation causes cell injury and leads to an increased risk of cancer, so it is of practical significance to identify biomarkers for gamma radiation. We used proteomic analysis to identify differentially expressed proteins in liver tissues of C57BL/6J mice treated with gamma radiation from Cs for 360 d. We confirmed obvious pathological changes in mouse liver tissues after irradiation. Compared with the control group, 74 proteins showed a fold change of ≥1.5 in the irradiated groups. We selected 24 proteins for bioinformatics analysis and peptide mass fingerprinting and found that 20 of the identified proteins were meaningful. These proteins were associated with tumorigenesis, tumor suppression, catalysis, cell apoptosis, cytoskeleton, metabolism, gene transcription, T-cell response, and other pathways. We confirmed that both cofilin-1 and destrin were up regulated in the irradiated groups by western blot and real-time polymerase chain reaction. Our findings indicate that cofilin-1 and destrin are sensitive to gamma radiation and may be potential biomarkers for gamma radiation. Whether these proteins are involved in radiation-induced tumorigenesis requires further investigation.
Collapse
Affiliation(s)
- Lan Yi
- Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China
- Institute of Cytology and Genetics, College of Pharmaceutical and Biological Science, University of South China
| | - Nan Hu
- Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China
| | - Hongxiang Mu
- Institute of Cytology and Genetics, College of Pharmaceutical and Biological Science, University of South China
| | - Jing Sun
- Institute of Cytology and Genetics, College of Pharmaceutical and Biological Science, University of South China
| | - Jie Yin
- Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China
- Institute of Cytology and Genetics, College of Pharmaceutical and Biological Science, University of South China
| | - Keren Dai
- Institute of Cytology and Genetics, College of Pharmaceutical and Biological Science, University of South China
| | - Fanghui Xu
- Institute of Cytology and Genetics, College of Pharmaceutical and Biological Science, University of South China
| | - Nanyang Yang
- Institute of Cytology and Genetics, College of Pharmaceutical and Biological Science, University of South China
| | - Dexin Ding
- Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China
| |
Collapse
|
18
|
Kopp S, Krüger M, Bauer J, Wehland M, Corydon TJ, Sahana J, Nassef MZ, Melnik D, Bauer TJ, Schulz H, Schütte A, Schmitz B, Oltmann H, Feldmann S, Infanger M, Grimm D. Microgravity Affects Thyroid Cancer Cells during the TEXUS-53 Mission Stronger than Hypergravity. Int J Mol Sci 2018; 19:E4001. [PMID: 30545079 PMCID: PMC6321223 DOI: 10.3390/ijms19124001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/06/2018] [Accepted: 12/09/2018] [Indexed: 12/24/2022] Open
Abstract
Thyroid cancer is the most abundant tumor of the endocrine organs. Poorly differentiated thyroid cancer is still difficult to treat. Human cells exposed to long-term real (r-) and simulated (s-) microgravity (µg) revealed morphological alterations and changes in the expression profile of genes involved in several biological processes. The objective of this study was to examine the effects of short-term µg on poorly differentiated follicular thyroid cancer cells (FTC-133 cell line) resulting from 6 min of exposure to µg on a sounding rocket flight. As sounding rocket flights consist of several flight phases with different acceleration forces, rigorous control experiments are mandatory. Hypergravity (hyper-g) experiments were performed at 18g on a centrifuge in simulation of the rocket launch and s-µg was simulated by a random positioning machine (RPM). qPCR analyses of selected genes revealed no remarkable expression changes in controls as well as in hyper-g samples taken at the end of the first minute of launch. Using a centrifuge initiating 18g for 1 min, however, presented moderate gene expression changes, which were significant for COL1A1, VCL, CFL1, PTK2, IL6, CXCL8 and MMP14. We also identified a network of mutual interactions of the investigated genes and proteins by employing in-silico analyses. Lastly, µg-samples indicated that microgravity is a stronger regulator of gene expression than hyper-g.
Collapse
Affiliation(s)
- Sascha Kopp
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Marcus Krüger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Johann Bauer
- Max Planck Institute of Biochemistry, D-82152 Martinsried, Germany.
| | - Markus Wehland
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark.
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, 8000 Aarhus C, Denmark.
| | - Jayashree Sahana
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark.
| | - Mohamed Zakaria Nassef
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Daniela Melnik
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Thomas J Bauer
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Herbert Schulz
- Cologne Center for Genomics, University of Cologne, D-50931 Cologne, Germany.
| | - Andreas Schütte
- Airbus Defence and Space GmbH, Airbus-Allee 1, D-28199 Bremen, Germany.
| | - Burkhard Schmitz
- Airbus Defence and Space GmbH, Airbus-Allee 1, D-28199 Bremen, Germany.
| | - Hergen Oltmann
- Airbus Defence and Space GmbH, Airbus-Allee 1, D-28199 Bremen, Germany.
| | - Stefan Feldmann
- Airbus Defence and Space GmbH, Airbus-Allee 1, D-28199 Bremen, Germany.
| | - Manfred Infanger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Daniela Grimm
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus C, Denmark.
- Gravitational Biology and Translational Regenerative Medicine, Faculty of Medicine and Mechanical Engineering, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany.
| |
Collapse
|
19
|
Targeting the NRF-2/RHOA/ROCK signaling pathway with a novel aziridonin, YD0514, to suppress breast cancer progression and lung metastasis. Cancer Lett 2018; 424:97-108. [DOI: 10.1016/j.canlet.2018.03.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/06/2018] [Accepted: 03/21/2018] [Indexed: 01/08/2023]
|
20
|
Yu BB, Lin GX, Li L, Qu S, Liang ZG, Chen KH, Zhou L, Lu QT, Sun YC, Zhu XD. Cofilin-2 Acts as a Marker for Predicting Radiotherapy Response and Is a Potential Therapeutic Target in Nasopharyngeal Carcinoma. Med Sci Monit 2018; 24:2317-2329. [PMID: 29664897 PMCID: PMC5921956 DOI: 10.12659/msm.909832] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background The purpose of this study was to determine whether cofilin-2 could serve as a protein marker for predicting radiotherapy response and as a potential therapeutic target in nasopharyngeal carcinoma (NPC). Material/Methods Cofilin-2 protein levels in serum and tissue samples from patients with NPC were assessed by sandwich ELISA and IHC. In vitro, cofilin-2 levels in CNE-2R cells were significantly higher than those of CNE-2 cells. Meanwhile, CNE-2R cells were silenced for cofilin-2 to obtain a stable cofilin-2-RNAi-LV3 cell line. Then, cell proliferation, radiosensitivity, invasion and migration abilities, cell cycle, and apoptosis were evaluated by Cell Counting Kit 8 assay (CCK-8), flow cytometry (FCM), clone formation assay, and in vitro. Results The secreted levels of the cofilin-2 protein in radioresistant NPC patients were significantly higher than those of radiosensitive cases. After cofilin-2 knockdown in nasopharyngeal carcinoma CNE-2R cells, proliferation was decreased, while apoptosis and radiosensitivity were enhanced; cell cycle distribution was altered, and the transplanted tumors in nude mice grew significantly less. Conclusions Overall, our findings suggest that cofilin-2 acts as a marker for predicting radiotherapy response and is a potential therapeutic target in nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Bin-Bin Yu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Guo-Xiang Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Ling Li
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Key Laboratory of High-Incidence Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China (mainland)
| | - Song Qu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Key Laboratory of High-Incidence Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China (mainland)
| | - Zhong-Guo Liang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Kai-Hua Chen
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Lei Zhou
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Qi-Teng Lu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yong-Chu Sun
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Xiao-Dong Zhu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumors, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Key Laboratory of High-Incidence Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China (mainland).,Department of Oncology, Affiliated Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
21
|
Tan J, Liu Y, Maimaiti Y, Wang C, Yan Y, Zhou J, Ruan S, Huang T. Combination of SIRT1 and Src overexpression suggests poor prognosis in luminal breast cancer. Onco Targets Ther 2018; 11:2051-2061. [PMID: 29695913 PMCID: PMC5905521 DOI: 10.2147/ott.s162503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objectives 1) Analyze the correlation of SIRT1 and Src with human breast cancer (BC) prognosis; 2) explore the roles of SIRT1 and Src in BC cell proliferation, tumor invasion, and metastasis; and 3) analyze the correlation and interaction between SIRT1 and Src. Materials and methods 1) Tissue microarray was used to analyze the expression of SIRT1 and Src in human BC tissues and the correlation between protein expression and cancer prognosis; 2) CCK8 assay was used to determine the influence of SIRT1 and Src inhibitors on BC cell proliferation; 3) Transwell migration assay and wound healing assay were used to determine the effect of SIRT1 and Src inhibitors on BC cell migration and invasion; and 4) Western blotting was used to analyze the correlation and interaction between SIRT1 and Src. Results 1) Combination of SIRT1 and/or Src positivity is a prognosis factor in BC, especially in luminal type; 2) MCF-7 cell proliferation is suppressed by SIRT1 inhibitor Ex527, and cell migration and invasion were inhibited by Src inhibitor bosutinib; 3) combined with Ex527, bosutinib has a significantly increased effect on MCF-7 cell migration suppression; and 4) there is a positive association between SIRT1 and Src both in BC tissues and in MCF-7 cells. Conclusion 1) SIRT1 and Src overexpression are both correlated with poor prognosis in human BC; 2) SIRT1 + Src (SIRT1 and/or Src positivity) is a fine prognosis model for luminal-type BC; 3) SIRT1 is a copromotor of Src in BC migration and invasion, but not in cell proliferation; and 4) our results suggest a potential interaction or a common regulation pathway between SIRT1 and Src expression and activity.
Collapse
Affiliation(s)
- Jie Tan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuyin Liu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yusufu Maimaiti
- Department of General Surgery, Research Institute of Minimally Invasive, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Changwen Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengnan Ruan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Liu F, Wang G, Wang X, Che Z, Dong W, Guo X, Wang Z, Chen P, Hou D, Zhang Q, Zhang W, Pan Y, Yang D, Liu H. Targeting high Aurora kinases expression as an innovative therapy for hepatocellular carcinoma. Oncotarget 2018; 8:27953-27965. [PMID: 28427193 PMCID: PMC5438621 DOI: 10.18632/oncotarget.15853] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/20/2017] [Indexed: 12/16/2022] Open
Abstract
The Aurora kinases A and B control tumorigenesis by inhibiting apoptosis and promoting proliferation and metastasis, however, it remains unknown whether Aurora A and B overexpressed concomitantly and its clinical significance in hepatocellular carcinoma (HCC). Here, we obsearved Aurora A and B tended to overexpress parallelly on protein level (r = 0.8679, P < 0.0001) and their co-overexpression (Aurora AHBH), associated with the worst prognosis, was an independent predictor for the survival. Importantly, with the lower IC50 and stronger anti-tumor effect than selective inhibitors, SNS-314, the pan-inhibitor of Aurora kinases, which induced YAP (Yes-associated protein) reduction and resulted in P21 accumulation, significantly promoted the polyploidy (> 4N) formation and apoptosis in HCC. High YAP expression (YAPH) was associated with Aurora AHBH, and appeared to be an independent predictor for survival, but P21 not. Moreover, silencing YAP also induced P21 accumulation, and knockdown P21, which enhanced YAP accumulation and weakened the SNS-314-induced YAP reduction, impaired SNS-314-induced apoptosis. Therefore, P21 enhanced the apoptotic effect of SNS-314 in HCC. Taken together, our findings indicated Aurora kinases/YAP/P21 was an oncogenic signaling axis in HCC, and revealed targeting Aurora AHBH induced apoptosis by YAP suppression. Our results also provided a solid evidence for SNS-314 as a potential targeted therapy, and a proof-of-concept evidence for a possible combined therapy of SNS-314 plus Hippo pathway inhibitors on HCC.
Collapse
Affiliation(s)
- Fuchen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China.,Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Guangyong Wang
- Department of Gastroenterology, 411 Hospital of PLA, Shanghai 200081, China
| | - Xiaoqiang Wang
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhihui Che
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Dong
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Xinggang Guo
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Zhenguang Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Ping Chen
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Daisen Hou
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Qi Zhang
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Wenli Zhang
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Yida Pan
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Dongqin Yang
- Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| |
Collapse
|
23
|
Zhang Y, Wang Y, Xue J. Paclitaxel inhibits breast cancer metastasis via suppression of Aurora kinase-mediated cofilin-1 activity. Exp Ther Med 2018; 15:1269-1276. [PMID: 29434713 PMCID: PMC5776659 DOI: 10.3892/etm.2017.5588] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022] Open
Abstract
The main problem in breast cancer treatment is the recurrence of tumor growth and metastases. Previous studies have suggested that Paclitaxel is widely used to treat various cancers. The present study analyzed the potential signaling pathway of Paclitaxel-inhibited breast cancer metastasis. It was demonstrated that Paclitaxel treatment significantly inhibited growth of breast cancer cell lines including MCF-7 and SKBR3 cells. Results demonstrated that Paclitaxel significantly inhibited breast cancer cell migration and invasion. Results additionally demonstrated that Paclitaxel treatment suppressed Aurora kinase and cofilin-1 activity in breast cancer cells. The potential mechanism indicated that activation of Aurora kinase activity stimulated cofilin-1 activity, which canceled Paclitaxel-inhibited growth and aggressiveness of breast cancer cells. An in vivo assay revealed that Paclitaxel treatment significantly inhibited breast cancer growth. Immunohistochemistry demonstrated that Paclitaxel treatment increased apoptosis of tumor cells in tumor tissue. Notably, Aurora kinase and cofilin-1 activity were downregulated by Paclitaxel in tumor tissues. In conclusion, these results indicated that Paclitaxel inhibited breast cancer cell growth and metastasis via suppression of Aurora kinase-mediated cofilin-1 activity, suggesting Paclitaxel may be an efficient anticancer agent for the treatment of this disease.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Mammography Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Yaoyi Wang
- Department of Radiology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Jun Xue
- Department of Vessels and Glands Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| |
Collapse
|
24
|
Rashid FA, Mansoor Q, Tabassum S, Aziz H, Arfat WO, Naoum GE, Ismail M, Farooqi AA. Signaling cascades in thyroid cancer: Increasing the armory of archers to hit bullseye. J Cell Biochem 2018; 119:3798-3808. [PMID: 29243843 DOI: 10.1002/jcb.26620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022]
Abstract
Thyroid cancer is a multifaceted and therapeutically challenging disease and rapidly accumulating experimentally verified findings have considerably improve our understanding of the molecular mechanisms which underlie its development. Substantial fraction of information has been added into existing landscape of molecular oncology and we have started to develop a sharper understanding of the underlying mechanisms of thyroid cancer. Wealth of information demystified different intracellular signaling cascades which are frequently deregulated in thyroid cancer. In vitro assays and xenografted mice based studies have helped us to identify drug targets and different synthetic and natural products are currently being tested to effectively treat thyroid cancer. Cabozantinib and vandetanib have been approved to treat medullary thyroid cancer (MTC) and two agents (lenvatinib and sorafenib) are also being used to treat radioactive-iodine refractory differentiated thyroid cancer. This review comprehensively summarizes most recent advancements in our knowledge related to dysregulated intracellular signaling cascades in thyroid cancer and how different proteins can be therapeutically exploited. (1) We discuss how loss of TRAIL mediated apoptosis occurred in thyroid cancer cells and how different strategies can be used to restore apoptosis in resistant cancer cells; (2) We provide detailed account of seemingly opposite roles of NOTCH signaling in thyroid cancers; (3) TGF/SMAD mediated signaling also needs detailed research because of context dependent role in thyroid cancer. Researchers have only begun to scratch the surface of how TGF signaling works in thyroid cancer and metastasis; and (4) Role of SHH signaling in thyroid cancer stem cells is also well appreciated and targeting of SHH pathway will be an important aspect in treatment of thyroid cancer. Better concepts and improved knowledge will be helpful for clinicians in getting a step closer to individualized medicine.
Collapse
Affiliation(s)
- Faiza Abdul Rashid
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan.,Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Qaisar Mansoor
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Sobia Tabassum
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Hafsa Aziz
- Nuclear Medicine, Oncology and Radiotherapy Institute, H-10 Campus, Islamabad, Pakistan
| | - Waleed O Arfat
- Alexandria Comprehensive Cancer Center, Alexandria, Egypt.,Department of Radiation Oncology, Alexandria University, Alexandria, Egypt
| | - George E Naoum
- Alexandria Comprehensive Cancer Center, Alexandria, Egypt.,Department of Radiation oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Muhammad Ismail
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | | |
Collapse
|
25
|
He JG, Li L, Qin Y, Yu W, He X, Gang R. Aurora-A Regulates Progression and Metastasis of Colorectal Cancer by Promoting Slug Activity. Technol Cancer Res Treat 2017. [PMCID: PMC5762031 DOI: 10.1177/1533034616682172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) is associated with cancer metastasis and poor prognosis, but the exact mechanism has not been clarified. Centrosomal Aurora-A kinase gene is frequently overexpressed in a variety of cancers and plays a pivotal role in the growth and survival of cancer cells. However, its role in colorectal cancer metastasis has not been confirmed. Here we demonstrate that Aurora-A plays a crucial role in the progression and metastasis of colorectal cancer by regulating epithelial–mesenchymal transition. In our study, increased Aurora-A expression was detected in colorectal cancer clinical specimens compared to normal colorectal tissues. Moreover, overexpressed Aurora-A significantly promoted the proliferation, migration, and invasion capacity of colorectal cancer cells and then enhanced metastatic capacity of colorectal cancer in vitro and in vivo and eventually led to poor prognosis. Conversely, silencing Aurora-A expression in colorectal cancer cells decreased the capacity of proliferation, migration, and invasion and further reduced colorectal cancer metastasis. Mechanistically, we found that Slug was involved in Aurora-A–induced migration and invasion of colorectal cancer cells. Silencing Slug expression could block Aurora-A–induced migration, invasion, and metastasis of colorectal cancer cells. Furthermore, the expression of Aurora-A and Slug were positively correlated in colorectal cancer tissues and paired normal colorectal tissue. Taken together, our findings revealed a critical role of Aurora-A in colorectal cancer progression and metastasis by regulating epithelial–mesenchymal transition.
Collapse
Affiliation(s)
- Jin-Guang He
- Department of Oncology, Heze Municiple Hospital, Heze, Shandong, People’s Republic of China
| | - Luming Li
- Department of Cardiology, Weihai Municiple Hospital, Weihai, Shandong, People’s Republic of China
| | - Ying Qin
- Department of Anatomy and Histology, Shandong Medical College, Jinan, Shandong, People’s Republic of China
| | - Wenfei Yu
- Shandong University School of Medicine, Jinan, Shandong, People’s Republic of China
| | - Xiuquan He
- Department of Human Anatomy, Shandong University School of Medicine, Jinan, Shandong, People’s Republic of China
| | - Ren Gang
- Department of Oncology, People’s Hospital of Laiwu City, Laiwu, Shandong, People’s Republic of China
| |
Collapse
|
26
|
Chen C, Maimaiti Y, Zhijun S, Zeming L, Yawen G, Pan Y, Tao H. Slingshot-1L, a cofilin phosphatase, induces primary breast cancer metastasis. Oncotarget 2017; 8:66195-66203. [PMID: 29029503 PMCID: PMC5630403 DOI: 10.18632/oncotarget.19855] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/28/2017] [Indexed: 01/06/2023] Open
Abstract
Slingshot (SSH) is a member of the conserved family of cofilin phosphatases that plays a critical role in cell membrane protrusion and migration by transforming inactive phosphorylated cofilin to an active form. SSH-like protein 1 (SSH-1L) expression is detected in various types of tumors; insulin induces the phosphatases activity of SSH-1L in a phosphoinositide 3-kinase-dependent manner. However, little is known about the expression and role of SSH-1L in breast cancer. Here, we analyzed 295 human breast cancer tissue specimens for SSH-1L expression by immunohistochemistry. The correlation between SSH-1L level and patients' clinical characteristics was analyzed with Pearson's χ2 test. The function of SSH-1L was evaluated by gene knockdown and quantitative real-time polymerase chain reaction detection of cofilin expression in MDA-MB-231, MCF-7, and SK-BR-3 human breast cancer cell lines. SSH-1L expression was detected in 88.1% of tissue specimens by immunohistochemistry and was strongly associated with increased metastasis and mortality. Loss of SSH-1L expression decreased the nonphosphorylated, active form of cofilin in SK-BR-3 and MDA-MB-231 cell lines, which was associated with reduced cell motility. Accordingly, SSH-1L/cofilin signaling played a critical role in primary breast cancer metastasis and was a potential therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Chen Chen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Yusufu Maimaiti
- Department of General Surgery (Research Institute of Minimally Invasive), People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830000, P.R. China
| | - Shen Zhijun
- Clinical Laboratory, The Third People's Hospital of Hubei Province, Wuhan 430000, P.R. China
| | - Liu Zeming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Guo Yawen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Yu Pan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Huang Tao
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| |
Collapse
|