1
|
Zhu J, Jia X, Ren S, Zhang Z, Li H, Wang J, Song B, Wu W, Peng C. Inhibition of Polo-Like Kinase 1 Dampens the Replication of Vaccinia Virus in Mammalian Cells. J Med Virol 2025; 97:e70240. [PMID: 39953955 DOI: 10.1002/jmv.70240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Since the eradication of smallpox, zoonotic poxviruses, such as the mpox virus (MPXV), continue to pose a threat to public health. Identifying drugs that reduce poxvirus infection and replication, as well as understanding their molecular mechanisms, is essential for epidemic control. Polo-like kinase 1 (PLK1) has been shown to facilitate vaccinia virus (VACV) infection and replication. This study confirms the effects of the PLK1 inhibitors HMN-214 and ON-01910 on VACV replication in A549 cells. Both viral titers and DNA loads were significantly reduced in treated cells after infection. Additionally, ON-01910 demonstrated broad-spectrum antiviral activity against the lumpy skin disease virus (LSDV) and the infectious bovine rhinotracheitis virus (IBRV) in vitro. PLK1 knockdown in A549 cells also led to a reduction in VACV protein expression, viral titers, and DNA levels. Further analysis showed that VACV infection leads to the accumulation of PLK1 near viral factories. However, despite its strong in vitro effects, ON-01910 did not significantly reduce VACV replication in mice. These findings highlight the critical role of PLK1 in VACV replication and its potential as a target for antiviral therapy against orthopoxviruses.
Collapse
Affiliation(s)
- Junda Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xuejiao Jia
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shuning Ren
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zihui Zhang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hua Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jing Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Baifen Song
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wenxue Wu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chen Peng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
2
|
Zhang L, Wang S, Wang L. Comprehensive analysis identifies YKT6 as a potential prognostic and diagnostic biomarker in lung adenocarcinoma. BMC Cancer 2024; 24:1235. [PMID: 39375639 PMCID: PMC11460176 DOI: 10.1186/s12885-024-12975-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Lung cancer is the most common cause of cancer-related death worldwide. The most prevalent histological subtype of lung cancer is lung adenocarcinoma (LUAD), with incidence rising each year. Treating LUAD remains a significant issue due to a lack of early diagnosis and poor therapy outcomes. YKT6 is a member of the SNARE protein family, whose clinical value and biological function in LUAD has yet to be established. METHODS TCGA, HPA and UALCAN were used to analyze YKT6 mRNA and protein levels, the correlation between YKT6 expression and clinicopathological features and prognosis. YKT6 mRNA and protein expression were verified by qRT-PCR, immunohistochemistry (IHC) and tissue microarrays (TMA). Additionally, lung cancer cell lines were chosen for YKT6 silencing to explore the effects on cell proliferation and migration. The cBioPortal was used to select YKT6-related genes. Protein-protein interaction (PPI) network was created based on STRING database and hub genes were screened, with their expression levels and prognosis values in LUAD analyzed accordingly. YKT6-related genes were enriched by gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analyses. RESULTS In LUAD, YKT6 was distinctly highly expressed with relation to clinical features of staging, smoking, lymph node metastasis, and TP53 mutation. Elevated YKT6 expression was linked to adverse prognosis, serving as an independent unfavorable prognostic factor. Moreover, YKT6 presented high diagnostic value in LUAD patients (AUC = 0.856). Experimental validation indicated that freshly collected LUAD tissues showed significantly high mRNA expression of YKT6. IHC and TMA verified increased YKT6 protein level in LUAD. Knockdown of YKT6 inhibited cell proliferation and promoted apoptosis, with mitigated capability of migration and invasion. The top ten hub genes screened by PPI network were highly expressed in LUAD, and significantly associated with poor prognosis. GO and KEGG analyses showed that YKT6-related genes were mainly involved in cell cycle. CONCLUSION Elevated YKT6 expression is related to poor prognosis of LUAD patients. YKT6 can serve as a novel biomarker for LUAD diagnosis and prognosis. Cell proliferation, migration and invasion was impaired with increased apoptosis upon YKT6 silencing in lung cancer cells. In summary, this study comprehensively uncovered that YKT6 could be identified as a potential prognostic and diagnostic biomarker in LUAD.
Collapse
Affiliation(s)
- Liming Zhang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
- Department of Thoracic Surgery, Weifang Second People's Hospital, Weifang, Shandong, 261041, P.R. China
| | - Shaoqiang Wang
- Department of Thoracic Surgery, Weifang People's Hospital, Weifang, Shandong, 261000, P.R. China
| | - Lina Wang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China.
| |
Collapse
|
3
|
Zhang Y, Fong KW, Mao F, Wang R, Allison DB, Napier D, He D, Liu J, Zhang Y, Chen J, Kong Y, Li C, Li G, Liu J, Li Z, Zhu H, Wang C, Liu X. Elevating PLK1 overcomes BETi resistance in prostate cancer via triggering BRD4 phosphorylation-dependent degradation in mitosis. Cell Rep 2024; 43:114431. [PMID: 38968071 PMCID: PMC11334074 DOI: 10.1016/j.celrep.2024.114431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 04/20/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024] Open
Abstract
Bromodomain-containing protein 4 (BRD4) has emerged as a promising therapeutic target in prostate cancer (PCa). Understanding the mechanisms of BRD4 stability could enhance the clinical response to BRD4-targeted therapy. In this study, we report that BRD4 protein levels are significantly decreased during mitosis in a PLK1-dependent manner. Mechanistically, we show that BRD4 is primarily phosphorylated at T1186 by the CDK1/cyclin B complex, recruiting PLK1 to phosphorylate BRD4 at S24/S1100, which are recognized by the APC/CCdh1 complex for proteasome pathway degradation. We find that PLK1 overexpression lowers SPOP mutation-stabilized BRD4, consequently rendering PCa cells re-sensitized to BRD4 inhibitors. Intriguingly, we report that sequential treatment of docetaxel and JQ1 resulted in significant inhibition of PCa. Collectively, the results support that PLK1-phosphorylated BRD4 triggers its degradation at M phase. Sequential treatment of docetaxel and JQ1 overcomes BRD4 accumulation-associated bromodomain and extra-terminal inhibitor (BETi) resistance, which may shed light on the development of strategies to treat PCa.
Collapse
Affiliation(s)
- Yanquan Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.
| | - Ka-Wing Fong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Fengyi Mao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Ruixin Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Derek B Allison
- Pathology & Laboratory Medicine, University of Kentucky, Lexington, KY 40508, USA
| | - Dana Napier
- Biospecimen Procurement & Translational Pathology Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA
| | - Daheng He
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Jinpeng Liu
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Yeqing Zhang
- Department of Biology, College of Arts & Sciences, University of Kentucky, Lexington, KY 40506, USA
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Yifan Kong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Chaohao Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Guangbing Li
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Jinghui Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Zhiguo Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
4
|
Kong Y, Li C, Liu J, Wu S, Zhang M, Allison DB, Hassan F, He D, Wang X, Mao F, Zhang Q, Zhang Y, Li Z, Wang C, Liu X. Single-cell analysis identifies PLK1 as a driver of immunosuppressive tumor microenvironment in LUAD. PLoS Genet 2024; 20:e1011309. [PMID: 38885192 PMCID: PMC11182521 DOI: 10.1371/journal.pgen.1011309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
PLK1 (Polo-like kinase 1) plays a critical role in the progression of lung adenocarcinoma (LUAD). Recent studies have unveiled that targeting PLK1 improves the efficacy of immunotherapy, highlighting its important role in the regulation of tumor immunity. Nevertheless, our understanding of the intricate interplay between PLK1 and the tumor microenvironment (TME) remains incomplete. Here, using genetically engineered mouse model and single-cell RNA-seq analysis, we report that PLK1 promotes an immunosuppressive TME in LUAD, characterized with enhanced M2 polarization of tumor associated macrophages (TAM) and dampened antigen presentation process. Mechanistically, elevated PLK1 coincides with increased secretion of CXCL2 cytokine, which promotes M2 polarization of TAM and diminishes expression of class II major histocompatibility complex (MHC-II) in professional antigen-presenting cells. Furthermore, PLK1 negatively regulates MHC-II expression in cancer cells, which has been shown to be associated with compromised tumor immunity and unfavorable patient outcomes. Taken together, our results reveal PLK1 as a novel modulator of TME in LUAD and provide possible therapeutic interventions.
Collapse
Affiliation(s)
- Yifan Kong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Chaohao Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Sai Wu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Min Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Derek B. Allison
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, United States of America
| | - Faisal Hassan
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, United States of America
| | - Daheng He
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Xinyi Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Fengyi Mao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Qiongsi Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Yanquan Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Zhiguo Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
5
|
Zhao G, Wang Y, Zhou J, Ma P, Wang S, Li N. Pan-cancer analysis of polo-like kinase family genes reveals polo-like kinase 1 as a novel oncogene in kidney renal papillary cell carcinoma. Heliyon 2024; 10:e29373. [PMID: 38644836 PMCID: PMC11033160 DOI: 10.1016/j.heliyon.2024.e29373] [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: 10/08/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Polo-like kinases (PLKs) are a kinase class of serine/threonine with five members that play crucial roles in cell cycle regulation. However, their biological functions, regulation, and expression remain unclear. This study revealed the molecular properties, oncogenic role, and clinical significance of PLK genes in pan-cancers, particularly in kidney renal papillary cell carcinoma (KIRP). METHODS We evaluated the mutation landscape, expression level, and prognostic values of PLK genes using bioinformatics analyses and explored the association between the expression level of PLK genes and tumor microenvironment (TME), immune subtype, cancer immunotherapy, tumor stemness, and drug sensitivity. Finally, we verified the prognostic value in patients with KIRP through univariate and multivariate analyses and nomogram construction. RESULTS PLK genes are extensively altered in pan-cancer, which may contribute to tumorigenesis. These genes are aberrantly expressed in some types of cancer, with PLK1 being overexpressed in 31 cancers. PLK expression is closely associated with the prognosis of various cancers. The expression level of PLK genes is related with sensitivity to diverse drugs and cancer immunity as well as cancer immunotherapy. Importantly, we verified that PLK1 was overexpressed in KIRP tissues and could be an unfavorable prognostic biomarker in patients with KIRP. Hence, PLK1 may serve as an oncogenic gene in KIRP and should be explored in future studies. CONCLUSIONS Our study comprehensively reports the molecular characteristics and biological functions of PLK family gens across human cancers and recommends further investigation of these genes as potential biomarkers and therapeutic targets, especially in KIRP.
Collapse
Affiliation(s)
| | | | - Jiawei Zhou
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peiwen Ma
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| |
Collapse
|
6
|
Raab M, Kostova I, Peña‐Llopis S, Fietz D, Kressin M, Aberoumandi SM, Ullrich E, Becker S, Sanhaji M, Strebhardt K. Rescue of p53 functions by in vitro-transcribed mRNA impedes the growth of high-grade serous ovarian cancer. Cancer Commun (Lond) 2024; 44:101-126. [PMID: 38140698 PMCID: PMC10794014 DOI: 10.1002/cac2.12511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The cellular tumor protein p53 (TP53) is a tumor suppressor gene that is frequently mutated in human cancers. Among various cancer types, the very aggressive high-grade serous ovarian carcinoma (HGSOC) exhibits the highest prevalence of TP53 mutations, present in >96% of cases. Despite intensive efforts to reactivate p53, no clinical drug has been approved to rescue p53 function. In this study, our primary objective was to administer in vitro-transcribed (IVT) wild-type (WT) p53-mRNA to HGSOC cell lines, primary cells, and orthotopic mouse models, with the aim of exploring its impact on inhibiting tumor growth and dissemination, both in vitro and in vivo. METHODS To restore the activity of p53, WT p53 was exogenously expressed in HGSOC cell lines using a mammalian vector system. Moreover, IVT WT p53 mRNA was delivered into different HGSOC model systems (primary cells and patient-derived organoids) using liposomes and studied for proliferation, cell cycle progression, apoptosis, colony formation, and chromosomal instability. Transcriptomic alterations induced by p53 mRNA were analyzed using RNA sequencing in OVCAR-8 and primary HGSOC cells, followed by ingenuity pathway analysis. In vivo effects on tumor growth and metastasis were studied using orthotopic xenografts and metastatic intraperitoneal mouse models. RESULTS Reactivation of the TP53 tumor suppressor gene was explored in different HGSOC model systems using newly designed IVT mRNA-based methods. The introduction of WT p53 mRNA triggered dose-dependent apoptosis, cell cycle arrest, and potent long-lasting inhibition of HGSOC cell proliferation. Transcriptome analysis of OVCAR-8 cells upon mRNA-based p53 reactivation revealed significant alterations in gene expression related to p53 signaling, such as apoptosis, cell cycle regulation, and DNA damage. Restoring p53 function concurrently reduces chromosomal instability within the HGSOC cells, underscoring its crucial contribution in safeguarding genomic integrity by moderating the baseline occurrence of double-strand breaks arising from replication stress. Furthermore, in various mouse models, treatment with p53 mRNA reduced tumor growth and inhibited tumor cell dissemination in the peritoneal cavity in a dose-dependent manner. CONCLUSIONS The IVT mRNA-based reactivation of p53 holds promise as a potential therapeutic strategy for HGSOC, providing valuable insights into the molecular mechanisms underlying p53 function and its relevance in ovarian cancer treatment.
Collapse
Affiliation(s)
- Monika Raab
- Department of GynecologyMedical SchoolGoethe‐UniversityFrankfurt am MainGermany
| | - Izabela Kostova
- Department of GynecologyMedical SchoolGoethe‐UniversityFrankfurt am MainGermany
| | - Samuel Peña‐Llopis
- Translational Genomics in Solid TumorsWest German Cancer CenterUniversity HospitalEssenGermany
- German Cancer Consortium (DKTK)EssenGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Daniela Fietz
- Histology and EmbryologyInstitute for Veterinary AnatomyGiessenGermany
| | - Monika Kressin
- Department of GynecologyMedical SchoolGoethe‐UniversityFrankfurt am MainGermany
- Histology and EmbryologyInstitute for Veterinary AnatomyGiessenGermany
| | - Seyed Mohsen Aberoumandi
- Histology and EmbryologyInstitute for Veterinary AnatomyGiessenGermany
- Franfurt Cancer Institute (FCI)Goethe UniversityFrankfurt am MainGermany
- German Cancer Consortium (DKTK), Partner site Frankfurt/Mainz, a partnership between DKFZ and University Hospital FrankfurtFrankfurt am MainGermany
| | - Evelyn Ullrich
- Franfurt Cancer Institute (FCI)Goethe UniversityFrankfurt am MainGermany
- German Cancer Consortium (DKTK), Partner site Frankfurt/Mainz, a partnership between DKFZ and University Hospital FrankfurtFrankfurt am MainGermany
- Experimental ImmunologyDepartment for Children and Adolescents MedicineUniversity Hospital FrankfurtGoethe UniversityFrankfurt am MainGermany
| | - Sven Becker
- Department of GynecologyMedical SchoolGoethe‐UniversityFrankfurt am MainGermany
| | - Mourad Sanhaji
- Department of GynecologyMedical SchoolGoethe‐UniversityFrankfurt am MainGermany
| | - Klaus Strebhardt
- Department of GynecologyMedical SchoolGoethe‐UniversityFrankfurt am MainGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
7
|
Li C, Allison DB, He D, Mao F, Wang X, Rychahou P, Imam IA, Kong Y, Zhang Q, Zhang Y, Liu J, Wang R, Rao X, Wu S, Evers BM, Shao Q, Wang C, Li Z, Liu X. Phosphorylation of AHR by PLK1 promotes metastasis of LUAD via DIO2-TH signaling. PLoS Genet 2023; 19:e1011017. [PMID: 37988371 PMCID: PMC10662729 DOI: 10.1371/journal.pgen.1011017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/13/2023] [Indexed: 11/23/2023] Open
Abstract
Metastasis of lung adenocarcinoma (LUAD) is a major cause of death in patients. Aryl hydrocarbon receptor (AHR), an important transcription factor, is involved in the initiation and progression of lung cancer. Polo-like kinase 1 (PLK1), a serine/threonine kinase, acts as an oncogene promoting the malignancy of multiple cancer types. However, the interaction between these two factors and their significance in lung cancer remain to be determined. In this study, we demonstrate that PLK1 phosphorylates AHR at S489 in LUAD, leading to epithelial-mesenchymal transition (EMT) and metastatic events. RNA-seq analyses reveal that type 2 deiodinase (DIO2) is responsible for EMT and enhanced metastatic potential. DIO2 converts tetraiodothyronine (T4) to triiodothyronine (T3), activating thyroid hormone (TH) signaling. In vitro and in vivo experiments demonstrate that treatment with T3 or T4 promotes the metastasis of LUAD, whereas depletion of DIO2 or a deiodinase inhibitor disrupts this property. Taking together, our results identify the AHR phosphorylation by PLK1 and subsequent activation of DIO2-TH signaling as mechanisms leading to LUAD metastasis. These findings can inform possible therapeutic interventions for this event.
Collapse
Affiliation(s)
- Chaohao Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Derek B. Allison
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, United States of America
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Daheng He
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Fengyi Mao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Xinyi Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Piotr Rychahou
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Surgery, University of Kentucky, Lexington, Kentucky, United States of America
| | - Ibrahim A. Imam
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky, United States of America
| | - Yifan Kong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Qiongsi Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Yanquan Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jinghui Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Ruixin Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Xiongjian Rao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Sai Wu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - B. Mark Evers
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Surgery, University of Kentucky, Lexington, Kentucky, United States of America
| | - Qing Shao
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky, United States of America
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Zhiguo Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, United States of America
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
8
|
Kong Y, Li C, Liu J, Zhang M, Allison DB, Hassan F, He D, Wang X, Mao F, Zhang Q, Zhang Y, Li Z, Wu S, Wang C, Liu X. Single-cell analysis characterizes PLK1 as a catalyst of an immunosuppressive tumor microenvironment in LUAD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551692. [PMID: 37577553 PMCID: PMC10418276 DOI: 10.1101/2023.08.02.551692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
PLK1 (Polo-like kinase 1) plays a critical role in the progression of lung adenocarcinoma (LUAD). Recent studies have unveiled that targeting PLK1 improves the efficacy of immunotherapy, highlighting its important role in the regulation of tumor immunity. Nevertheless, our understanding of the intricate interplay between PLK1 and the tumor microenvironment (TME) remains incomplete. Here, using genetically engineered mouse model and single-cell RNA-seq analysis, we report that PLK1 promotes an immunosuppressive TME in LUAD, characterized with enhanced M2 polarization of tumor associated macrophages (TAM) and dampened antigen presentation process. Mechanistically, elevated PLK1 coincides with increased secretion of CXCL2 cytokine, which promotes M2 polarization of TAM and diminishes expression of class II major histocompatibility complex (MHC-II) in professional antigen-presenting cells. Furthermore, PLK1 negatively regulates MHC-II expression in cancer cells, which has been shown to be associated with compromised tumor immunity and unfavorable patient outcomes. Taken together, our results reveal PLK1 as a novel modulator of TME in LUAD and provide possible therapeutic interventions.
Collapse
|
9
|
Li C, Allison DB, He D, Mao F, Wang X, Rychahou P, Imam IA, Kong Y, Zhang Q, Zhang Y, Liu J, Wang R, Rao X, Wu S, Shao Q, Wang C, Li Z, Liu X. Phosphorylation of AHR by PLK1 promotes metastasis of LUAD via DIO2-TH signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.31.551298. [PMID: 37577647 PMCID: PMC10418090 DOI: 10.1101/2023.07.31.551298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Metastasis of Lung adenocarcinoma (LUAD) is a major cause of death in patients. Aryl hydrocarbon receptor (AHR) is an important transcription factor involved in the initiation and progression of lung cancer. Polo-like kinase 1 (PLK1), a serine/threonine kinase, is an oncogene that promotes the malignancy of multiple cancer types. Nonetheless, the interaction between these two factors and significance in lung cancer remains to be determined. Here, we demonstrate that PLK1 phosphorylates AHR at S489 in LUAD, which leads to epithelial-mesenchymal transition (EMT) and metastatic events. RNA-seq analyses show that type 2 deiodinase (DIO2) is responsible for EMT and enhanced metastatic potential. DIO2 converts tetraiodothyronine (T4) to triiodothyronine (T3), which then activates thyroid hormone signaling. In vitro and in vivo experiments demonstrate that treatment with T3 or T4 promotes the metastasis of LUAD, whereas depletion of DIO2 or deiodinase inhibitor disrupts this property. Taken together, our results identify the phosphorylation of AHR by PLK1 as a mechanism leading to the progression of LUAD and provide possible therapeutic interventions for this event.
Collapse
|
10
|
Kong Y, Allison DB, Zhang Q, He D, Li Y, Mao F, Li C, Li Z, Zhang Y, Wang J, Wang C, Brainson CF, Liu X. The kinase PLK1 promotes the development of <i>Kras</i>/<i>Tp53</i>-mutant lung adenocarcinoma through transcriptional activation of the receptor RET. Sci Signal 2022; 15:eabj4009. [PMID: 36194647 PMCID: PMC9737055 DOI: 10.1126/scisignal.abj4009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increased abundance of polo-like kinase 1 (PLK1) is observed in various tumor types, particularly in lung adenocarcinoma (LUAD). Here, we found that PLK1 accelerated the progression of LUAD through a mechanism that was independent of its role in mediating mitotic cell division. Analysis of human tumor databases revealed that increased PLK1 abundance in LUAD correlated with mutations in KRAS and p53, with tumor stage, and with reduced survival in patients. In a mouse model of KRAS<sup>G12D</sup>-driven, p53-deficient LUAD, PLK1 overexpression increased tumor burden, decreased tumor cell differentiation, and reduced animal survival. PLK1 overexpression in cultured cells and mice indirectly increased the expression of the gene encoding the receptor tyrosine kinase RET by phosphorylating the transcription factor TTF-1. Signaling by RET and mutant KRAS in these tumors converged to activate the mitogen-activated protein kinase (MAPK) pathway. Pharmacological inhibition of the MAPK pathway kinase MEK combined with inhibition of either RET or PLK1 markedly suppressed tumor growth. Our findings show that PLK1 can amplify MAPK signaling and reveal a potential target for stemming progression in lung cancers with high PLK1 abundance.
Collapse
Affiliation(s)
- Yifan Kong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Derek B. Allison
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA,Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Qiongsi Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Daheng He
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Yuntong Li
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Fengyi Mao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Chaohao Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Zhiguo Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Yanquan Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Jianlin Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Christine F. Brainson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA,Corresponding author.
| |
Collapse
|
11
|
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: 17] [Impact Index Per Article: 5.7] [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
|
12
|
Wu Q, Jiang S, Cheng T, Xu M, Lu B. A Novel Pyroptosis-related Prognostic Model for Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:770301. [PMID: 34869364 PMCID: PMC8634647 DOI: 10.3389/fcell.2021.770301] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second most lethal malignant tumor because of its significant heterogeneity and complicated molecular pathogenesis. Novel prognostic biomarkers are urgently needed because no effective and reliable prognostic biomarkers currently exist for HCC patients. Increasing evidence has revealed that pyroptosis plays a role in the occurrence and progression of malignant tumors. However, the relationship between pyroptosis-related genes (PRGs) and HCC patient prognosis remains unclear. In this study, 57 PRGs were obtained from previous studies and GeneCards. The gene expression profiles and clinical data of HCC patients were acquired from public data portals. Least absolute shrinkage and selection operator (LASSO) Cox regression analysis was performed to establish a risk model using TCGA data. Additionally, the risk model was further validated in an independent ICGC dataset. Our results showed that 39 PRGs were significantly differentially expressed between tumor and normal liver tissues in the TCGA cohort. Functional analysis confirmed that these PRGs were enriched in pyroptosis-related pathways. According to univariate Cox regression analysis, 14 differentially expressed PRGs were correlated with the prognosis of HCC patients in the TCGA cohort. A risk model integrating two PRGs was constructed to classify the patients into different risk groups. Poor overall survival was observed in the high-risk group of both TCGA (p < 0.001) and ICGC (p < 0.001) patients. Receiver operating characteristic curves demonstrated the accuracy of the model. Furthermore, the risk score was confirmed as an independent prognostic indicator via multivariate Cox regression analysis (TCGA cohort: HR = 3.346, p < 0.001; ICGC cohort: HR = 3.699, p < 0.001). Moreover, the single-sample gene set enrichment analysis revealed different immune statuses between high- and low-risk groups. In conclusion, our new pyroptosis-related risk model has potential application in predicting the prognosis of HCC patients.
Collapse
Affiliation(s)
- Qianqian Wu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China.,Department of Medicine, Nantong University, Nantong, China
| | - Sutian Jiang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China.,Department of Medicine, Nantong University, Nantong, China
| | - Tong Cheng
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China.,Department of Medicine, Nantong University, Nantong, China
| | - Manyu Xu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China.,Department of Medicine, Nantong University, Nantong, China
| | - Bing Lu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
13
|
Ren Y, Deng R, Zhang Q, Li J, Han B, Ye P. Bioinformatics analysis of key genes in triple negative breast cancer and validation of oncogene PLK1. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1637. [PMID: 33490149 PMCID: PMC7812170 DOI: 10.21037/atm-20-6873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Breast cancer is the most common malignancy in women. Triple-negative breast cancer (TNBC) refers to a special subtype that is deficient in the expression of estrogen (ER), progesterone (PR), and human epidermal growth factor receptor 2 (HER-2). In this study, a variety of bioinformatics analysis tools were used to screen Hub genes related to the occurrence and development of triple negative breast cancer, and their biological functions were analyzed. Methods Gene Expression Omnibus (GEO) breast cancer microarray data GSE62931 was selected as the research object. The differentially expressed genes (DEGs) were screened and the protein-protein interaction (PPI) network of DEGs was constructed using bioinformatics tools. The Hub genes were also screened. The Gene Ontology (GO) knowledgebase and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for biological enrichment analysis. The Gene Expression Profiling Interactive Analysis (GEPIA) online tool was used to verify the expression of the screened genes and patient survival. The effects of polo-like kinase 1 (PLK1) on the proliferation, invasion, migration, and dryness of breast cancer cells were verified using cell counting kit 8 (CCK-8), transwell migration assays, scratch tests, and clone formation tests. An animal model of subcutaneous xenotransplantation of breast cancer was established to evaluate the effect of PLK1 on the proliferation of breast cancer. Results A total of 824 DEGs were screened by GSE62931 microarray data; 405 of which were up-regulated and 419 of which were down-regulated. Functional enrichment analysis showed that these DEGs were mainly enriched in cancer-related pathways and were primarily involved in biological processes (BP) such as cell and mitotic division. From the Hub gene screening, PLK1 was further identified as the Hub gene associated with TNBC. Cell and animal experiments indicated that PLK1 promotes the proliferation, invasion, migration, and clone formation of breast cancer cells. Conclusions Gene chip combined with bioinformatics methods can effectively analyze the DEGs related to the occurrence and development of breast cancer, and the screening of PLK1 can provide theoretical guidance for further research on the molecular mechanism of breast cancer and the screening of molecular markers.
Collapse
Affiliation(s)
- Yi Ren
- Breast Department, Xuzhou Cancer Hospital/Xuzhou Hospital Affiliated to Jiangsu University, Xuzhou, China
| | - Rong Deng
- Department of General Surgery, Jiangsu Cancer Hospital, Nanjing, China
| | - Qian Zhang
- Breast Department, Xuzhou Cancer Hospital/Xuzhou Hospital Affiliated to Jiangsu University, Xuzhou, China
| | - Jing Li
- Breast Department, Xuzhou Cancer Hospital/Xuzhou Hospital Affiliated to Jiangsu University, Xuzhou, China
| | - Baosan Han
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Ye
- Shanghai Institute for Minimally Invasive Therapy, School of Medical Instrument & Food Engineering, University of Shanghai for Science & Technology, Shanghai, China
| |
Collapse
|
14
|
Yabuno Y, Uchihashi T, Sasakura T, Shimizu H, Naito Y, Fukushima K, Ota K, Kogo M, Nojima H, Yabuta N. Clathrin heavy chain phosphorylated at T606 plays a role in proper cell division. Cell Cycle 2019; 18:1976-1994. [PMID: 31272276 PMCID: PMC6681784 DOI: 10.1080/15384101.2019.1637201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/13/2019] [Accepted: 06/24/2019] [Indexed: 10/26/2022] Open
Abstract
Clathrin regulates mitotic progression, in addition to membrane trafficking. However, the detailed regulatory mechanisms of clathrin during mitosis remain elusive. Here, we demonstrate novel regulation of clathrin during mitotic phase of the cell cycle. Clathrin heavy chain (CHC) was phosphorylated at T606 by its association partner cyclin G-associated kinase (GAK). This phosphorylation was required for proper cell proliferation and tumor growth of cells implanted into nude mice. Immunofluorescence analysis showed that the localization of CHC-pT606 signals changed during mitosis. CHC-pT606 signals localized in the nucleus and at the centrosome during interphase, whereas CHC signals were mostly cytoplasmic. Co-immunoprecipitation suggested that CHC formed a complex with GAK and polo-like kinase 1 (PLK1). Depletion of GAK using siRNA induced metaphase arrest and aberrant localization of CHC-pT606, which abolished Kiz-pT379 (as a phosphorylation target of PLK1) signals on chromatin at metaphase. Taken together, we propose that the GAK_CHC-pT606_PLK1_Kiz-pT379 axis plays a role in proliferation of cancer cells.
Collapse
Affiliation(s)
- Yusuke Yabuno
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Toshihiro Uchihashi
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Towa Sasakura
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hiroyuki Shimizu
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoko Naito
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Division of Cancer Cell Regulation, Aichi Cancer Center Research Institute, Aichi, Japan
| | - Kohshiro Fukushima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kaori Ota
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Mikihiko Kogo
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hiroshi Nojima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Norikazu Yabuta
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
15
|
Feng Y, Lin J, Liu Y, Tang Y, Zhou Y, Zhong M. Investigation of expressions of PDK1, PLK1 and c-Myc in diffuse large B-cell lymphoma. Int J Exp Pathol 2019; 100:32-40. [PMID: 30912195 PMCID: PMC6463398 DOI: 10.1111/iep.12307] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/07/2019] [Accepted: 01/20/2019] [Indexed: 12/12/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma. Because the prognosis of DLBCL patients varies considerably, there is an urgent need to identify novel prognostic factors. In this study, we investigated the expression levels of the signalling enzyme 3-phosphoinositide-dependent protein kinase-1 (PDK1), the cell cycle regulatory enzyme Polo-like kinase 1 (PLK1) and the transcription factor (c-Myc) in DLBCL tissues and evaluated their clinical and prognostic significance. PDK1, PLK1 and c-Myc were detected by immunohistochemical staining of paraffin-embedded specimens from 152 DLBCL and 48 lymphadenitis patients. Expression levels were correlated with clinicopathological factors. PDK1, PLK1 and c-Myc were more commonly expressed in DLBCL specimens than in lymphadenitis specimens, and the expression of each protein correlated positively with that of the other two molecules. High PDK1, PLK1 and c-Myc expression, high international prognostic index score, high lactate dehydrogenase levels and late Ann Arbor stage were shown to correlate with shorter overall survival time. A multivariate Cox regression model showed that high expression levels of PLK1 and c-Myc were independent prognostic factors for DLBCL. Our findings indicate that PLK1 and c-Myc expression might be promising predictive biomarkers for DLBCL patients.
Collapse
Affiliation(s)
- Yuhua Feng
- Department of OncologyXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Jinguan Lin
- Department of Daytime ChemotherapyHunan Cancer HospitalXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Yiping Liu
- Department of OncologyXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Youhong Tang
- Department of OncologyXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yangying Zhou
- Department of OncologyXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Meizuo Zhong
- Department of OncologyXiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
16
|
Ganapathy S, Liu J, Xiong R, Yu T, Makriyannis A, Chen C. Chronic low dose arsenic exposure preferentially perturbs mitotic phase of the cell cycle. Genes Cancer 2018; 10:39-51. [PMID: 30899418 PMCID: PMC6420791 DOI: 10.18632/genesandcancer.185] [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: 01/25/2023] Open
Abstract
Environmental pollution is a big challenge for human survival. Arsenic compounds are well-known biohazard, the exposure of which is closely linked to onsets of various human diseases, particularly cancers. Upon chronically exposing to arsenic compounds, genomic integrity is often disrupted, leading to tumor development. However, the underlying mechanisms by which chronic, low dose arsenic exposure targets genetic stability to initiate carcinogenesis still remain not fully understood. In this study, human lung epithelial BEAS-2B cells and keratinocytes were treated with 0.5 μM of sodium arsenite for one month (designated as BEAS-2B-SA cells or keratinocytes-SA), and its effect on cell cycle responses was analyzed. After being arrested in mitotic phase of the cell cycle by nocodazole treatment, BEAS-2B-SA cells or keratinocytes-SA were delayed to enter next cytokinesis. The lagging exit of the cells from mitosis was accompanied by a sustained Plk1 phosphorylation, which led to a persistent activation of the mitotic regulators BubR1 and Cdc27. As the result, cyclin B1 (clnB1) degradation was attenuated. BEAS-2B-SA cells or keratinocytes-SA also expressed a constitutively active Akt. The cytogenetic analysis showed an increased numbers of aneuploidy in these cells. The suppression of Akt reversed the aberrant expressions of the mitotic regulators, delay of mitotic exit as well as chromosomal aberrations. Our findings suggest that a long-term exposure to low dose sodium arsenite aberrantly retains the catenation of mitosis, which facilitates establishing genetic instability and predisposes the cells to tumorigenesis.
Collapse
Affiliation(s)
| | - Jian Liu
- The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Sheng, P.R. China
| | - Rui Xiong
- The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Sheng, P.R. China
| | - Tianqi Yu
- The Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | | | - Changyan Chen
- The Center for Drug Discovery, Northeastern University, Boston, MA, USA
| |
Collapse
|
17
|
Wen D, Wu J, Wang L, Fu Z. SUMOylation Promotes Nuclear Import and Stabilization of Polo-like Kinase 1 to Support Its Mitotic Function. Cell Rep 2017; 21:2147-2159. [PMID: 29166606 PMCID: PMC5728694 DOI: 10.1016/j.celrep.2017.10.085] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/22/2017] [Accepted: 10/23/2017] [Indexed: 02/08/2023] Open
Abstract
As a pivotal mitotic regulator, polo-like kinase 1 (PLK1) is under highly coordinated and multi-layered regulation. However, the pathways that control PLK1's activity and function have just begun to be elucidated. PLK1 has recently been shown to be functionally modulated by post-translational modifications (PTMs), including phosphorylation and ubiquitination. Herein, we report that SUMOylation plays an essential role in regulating PLK1's mitotic function. We found that Ubc9 was recruited to PLK1 upon initial phosphorylation and activation by CDK1/cyclin B. By in vivo and in vitro SUMOylation assays, PLK1 was identified as a physiologically relevant small ubiquitin-related modifier (SUMO)-targeted protein, preferentially modified by SUMO-1. We further showed that K492 on PLK1 is essential for SUMOylation. SUMOylation causes PLK1's nuclear import and significantly increases its protein stability, both of which are critical for normal mitotic progression and genomic integrity. Our findings suggest that SUMOylation is an important regulatory mechanism governing PLK1's mitotic function.
Collapse
Affiliation(s)
- Donghua Wen
- Department of Human and Molecular Genetics, Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| | - Jianguo Wu
- Department of Human and Molecular Genetics, Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| | - Lei Wang
- Department of Human and Molecular Genetics, Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| | - Zheng Fu
- Department of Human and Molecular Genetics, Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA.
| |
Collapse
|
18
|
Ma X, Wang L, Huang D, Li Y, Yang D, Li T, Li F, Sun L, Wei H, He K, Yu F, Zhao D, Hu L, Xing S, Liu Z, Li K, Guo J, Yang Z, Pan X, Li A, Shi Y, Wang J, Gao P, Zhang H. Polo-like kinase 1 coordinates biosynthesis during cell cycle progression by directly activating pentose phosphate pathway. Nat Commun 2017; 8:1506. [PMID: 29138396 PMCID: PMC5686148 DOI: 10.1038/s41467-017-01647-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 10/05/2017] [Indexed: 12/28/2022] Open
Abstract
Two hallmarks for cancer cells are the accelerated cell cycle progression as well as the altered metabolism, however, how these changes are coordinated to optimize the growth advantage for cancer cells are still poorly understood. Here we identify that Polo-like kinase 1 (Plk1), a key regulator for cell mitosis, plays a critical role for biosynthesis in cancer cells through activating pentose phosphate pathway (PPP). We find that Plk1 interacts with and directly phosphorylates glucose-6-phosphate dehydrogenase (G6PD). By activating G6PD through promoting the formation of its active dimer, Plk1 increases PPP flux and directs glucose to the synthesis of macromolecules. Importantly, we further demonstrate that Plk1-mediated activation of G6PD is critical for its role to promote cell cycle progression and cancer cell growth. Collectively, these findings establish a critical role for Plk1 in regulating biosynthesis in cancer cells, exemplifying how cell cycle progression and metabolic reprogramming are coordinated for cancer progression.
Collapse
Affiliation(s)
- Xiaoyu Ma
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Lin Wang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - De Huang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Yunyan Li
- High Magnetic Field Laboratory, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, Anhui, 230031, China
| | - Dongdong Yang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Tingting Li
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Fudong Li
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Linchong Sun
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Haoran Wei
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Kun He
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Fazhi Yu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Debiao Zhao
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Lan Hu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Songge Xing
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Zhaoji Liu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Kui Li
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Jing Guo
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Zhenye Yang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Xin Pan
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Ailing Li
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Yunyu Shi
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Junfeng Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, Anhui, 230031, China.
| | - Ping Gao
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
| | - Huafeng Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
19
|
The Emerging Role of Polo-Like Kinase 1 in Epithelial-Mesenchymal Transition and Tumor Metastasis. Cancers (Basel) 2017; 9:cancers9100131. [PMID: 28953239 PMCID: PMC5664070 DOI: 10.3390/cancers9100131] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/22/2017] [Accepted: 09/25/2017] [Indexed: 12/31/2022] Open
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase that plays a key role in the regulation of the cell cycle. PLK1 is overexpressed in a variety of human tumors, and its expression level often correlates with increased cellular proliferation and poor prognosis in cancer patients. It has been suggested that PLK1 controls cancer development through multiple mechanisms that include canonical regulation of mitosis and cytokinesis, modulation of DNA replication, and cell survival. However, emerging evidence suggests novel and previously unanticipated roles for PLK1 during tumor development. In this review, we will summarize the recent advancements in our understanding of the oncogenic functions of PLK1, with a focus on its role in epithelial-mesenchymal transition and tumor invasion. We will further discuss the therapeutic potential of these functions.
Collapse
|
20
|
Kumar S, Sharma G, Chakraborty C, Sharma AR, Kim J. Regulatory functional territory of PLK-1 and their substrates beyond mitosis. Oncotarget 2017; 8:37942-37962. [PMID: 28415805 PMCID: PMC5514964 DOI: 10.18632/oncotarget.16290] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/03/2017] [Indexed: 12/04/2022] Open
Abstract
Polo-like kinase 1 (PLK-1) is a well-known (Ser/Thr) mitotic protein kinase and is considered as a proto-oncogene. As hyper-activation of PLK-1 is broadly associated with poor prognosis and cancer progression, it is one of the most extensively studied mitotic kinases. During mitosis, PLK-1 regulates various cell cycle events, such as spindle pole maturation, chromosome segregation and cytokinesis. However, studies have demonstrated that the role of PLK-1 is not only restricted to mitosis, but PLK-1 can also regulate other vital events beyond mitosis, including transcription, translation, ciliogenesis, checkpoint adaptation and recovery, apoptosis, chromosomes dynamics etc. Recent reviews have tried to define the regulatory role of PLK-1 during mitosis progression and tumorigenesis, but its' functional role beyond mitosis is still largely unexplored. PLK-1 can regulate the activity of many proteins that work outside of its conventional territory. The dysregulation of these proteins can cause diseases such as Alzheimer's disease, tumorigenesis etc. and may also lead to drug resistance. Thus, in this review, we discussed the versatile role of PLK-1 and tried to collect data to validate its' functional role in cell cycle regulation apart from mitosis.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Garima Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Chiranjib Chakraborty
- Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Ashish Ranjan Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| |
Collapse
|
21
|
Qin T, Chen F, Zhuo X, Guo X, Yun T, Liu Y, Zhang C, Lai L. Discovery of Novel Polo-Like Kinase 1 Polo-Box Domain Inhibitors to Induce Mitotic Arrest in Tumor Cells. J Med Chem 2016; 59:7089-96. [PMID: 27425654 DOI: 10.1021/acs.jmedchem.6b00261] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Polo-like kinase 1(Plk1) is vital for cell mitosis and has been identified as anticancer target. Its polo-box domain (PBD) mediates substrate binding, blocking of which may offer selective Plk1 inhibition compared to kinase domain inhibitors. Although several PBD inhibitors were reported, most of them suffer from low cell activity. Here, we report the discovery of novel inhibitors to induce mitotic arrest in HeLa cells by virtual screening with Plk1 PBD and cellular activity testing. Of the 81 compounds tested in the cell assay, 10 molecules with diverse chemical scaffolds are potent to induce mitotic arrest of HeLa at low micromolar concentrations. The best compound induces mitotic arrest of HeLa cells with an EC50 of 4.4 μM. The cellular active inhibitors showed binding to Plk1 PBD and compete with PBD substrate in microscale thermophoresis analysis.
Collapse
Affiliation(s)
- Tan Qin
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University , Beijing, 100871, China
| | - Fangjin Chen
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University , Beijing, 100871, China
- Center for Quantitative Biology, Peking University , Beijing, 100871, China
| | - Xiaolong Zhuo
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University , Beijing, 100871, China
| | - Xiao Guo
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University , Beijing, 100871, China
| | - Taikangxiang Yun
- Center for Quantitative Biology, Peking University , Beijing, 100871, China
| | - Ying Liu
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University , Beijing, 100871, China
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University , Beijing, 100871, China
| | - Luhua Lai
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University , Beijing, 100871, China
- Center for Quantitative Biology, Peking University , Beijing, 100871, China
| |
Collapse
|
22
|
Patel H, Stavrou I, Shrestha RL, Draviam V, Frame MC, Brunton VG. Kindlin1 regulates microtubule function to ensure normal mitosis. J Mol Cell Biol 2016; 8:338-48. [PMID: 26993041 PMCID: PMC4991666 DOI: 10.1093/jmcb/mjw009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/13/2015] [Accepted: 11/08/2015] [Indexed: 11/14/2022] Open
Abstract
Loss of Kindlin 1 (Kin1) results in the skin blistering disorder Kindler Syndrome (KS), whose symptoms also include skin atrophy and reduced keratinocyte proliferation. Kin1 binds to integrins to modulate their activation and more recently it has been shown to regulate mitotic spindles and cell survival in a Plk1-dependent manner. Here we report that short-term Kin1 deletion in mouse skin results in impaired mitosis, which is associated with reduced acetylated tubulin (ac-tub) levels and cell proliferation. In cells, impaired mitosis and reduced ac-tub levels are also accompanied by reduced microtubule stability, all of which are rescued by HDAC6 inhibition. The ability of Kin1 to regulate HDAC6-dependent cellular ac-tub levels is dependent on its phosphorylation by Plk1. Taken together, these data define a novel role for Kin1 in microtubule acetylation and stability and offer a mechanistic insight into how certain KS phenotypes, such as skin atrophy and reduced cell proliferation, arise.
Collapse
Affiliation(s)
- Hitesh Patel
- Edinburgh Cancer Research Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Ifigeneia Stavrou
- Edinburgh Cancer Research Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Roshan L Shrestha
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Viji Draviam
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK Present address: School of Biological and Chemical Sciences, Queen Mary University of London, London, E11 4NS, UK
| | - Margaret C Frame
- Edinburgh Cancer Research Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Valerie G Brunton
- Edinburgh Cancer Research Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| |
Collapse
|
23
|
Kumar S, Sharma AR, Sharma G, Chakraborty C, Kim J. PLK-1: Angel or devil for cell cycle progression. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1865:190-203. [PMID: 26899266 DOI: 10.1016/j.bbcan.2016.02.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/01/2016] [Accepted: 02/16/2016] [Indexed: 12/31/2022]
Abstract
PLK-1 is a key player in the eukaryotic cell cycle. Cell cycle progression is precisely controlled by cell cycle regulatory kinases. PLK-1 is a mitotic kinase that actively regulates the G2/M transition, mitosis, mitotic exit, and cytokinesis. During cell cycle progression, PLK-1 controls various events related to the cell cycle maturation, directly and/or indirectly. On the contrary, aberrant expression of PLK-1 is strongly associated with tumorigenesis and its poor prognosis. The misexpression of PLK-1 causes the abnormalities including aneuploidy, mitotic defects, leading to tumorigenesis through inhibiting the p53 and pRB genes. Therefore, we reviewed the role of PLK-1 in the cell cycle progression and in the tumorigenesis either as a cell cycle regulator or on an attractive anti-cancer drug target.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo 200-704, Republic of Korea.
| | - Ashish Ranjan Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo 200-704, Republic of Korea.
| | - Garima Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo 200-704, Republic of Korea.
| | - Chiranjib Chakraborty
- Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida 203201, India.
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo 200-704, Republic of Korea.
| |
Collapse
|
24
|
Wu J, Ivanov AI, Fisher PB, Fu Z. Polo-like kinase 1 induces epithelial-to-mesenchymal transition and promotes epithelial cell motility by activating CRAF/ERK signaling. eLife 2016; 5:e10734. [PMID: 27003818 PMCID: PMC4811775 DOI: 10.7554/elife.10734] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 03/03/2016] [Indexed: 12/22/2022] Open
Abstract
Polo-like kinase 1 (PLK1) is a key cell cycle regulator implicated in the development of various cancers, including prostate cancer. However, the functions of PLK1 beyond cell cycle regulation remain poorly characterized. Here, we report that PLK1 overexpression in prostate epithelial cells triggers oncogenic transformation. It also results in dramatic transcriptional reprogramming of the cells, leading to epithelial-to-mesenchymal transition (EMT) and stimulation of cell migration and invasion. Consistently, PLK1 downregulation in metastatic prostate cancer cells enhances epithelial characteristics and inhibits cell motility. The signaling mechanisms underlying the observed cellular effects of PLK1 involve direct PLK1-dependent phosphorylation of CRAF with subsequent stimulation of the MEK1/2-ERK1/2-Fra1-ZEB1/2 signaling pathway. Our findings highlight novel non-canonical functions of PLK1 as a key regulator of EMT and cell motility in normal prostate epithelium and prostate cancer. This study also uncovers a previously unanticipated role of PLK1 as a potent activator of MAPK signaling.
Collapse
Affiliation(s)
- Jianguo Wu
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, United States
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, United States
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, United States
| | - Andrei I Ivanov
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, United States
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, United States
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, United States
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, United States
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, United States
| | - Zheng Fu
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, United States
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, United States
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, United States
| |
Collapse
|
25
|
Li J, Wang R, Schweickert PG, Karki A, Yang Y, Kong Y, Ahmad N, Konieczny SF, Liu X. Plk1 inhibition enhances the efficacy of gemcitabine in human pancreatic cancer. Cell Cycle 2016; 15:711-9. [PMID: 26890815 PMCID: PMC4845940 DOI: 10.1080/15384101.2016.1148838] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 01/21/2023] Open
Abstract
Gemcitabine is the standard-of-care for chemotherapy in patients with pancreatic adenocarcinoma and it can directly incorporate into DNA or inhibit ribonucleotide reductase to prevent DNA replication and, thus, tumor cell growth. Most pancreatic tumors, however, develop resistance to gemcitabine. Polo-like kinase 1 (Plk1), a critical regulator in many cell cycle events, is significantly elevated in human pancreatic cancer. In this study, we show that Plk1 is required for the G1/S transition and that inhibition of Plk1 significantly reduces the DNA synthesis rate in human pancreatic cancer cells. Furthermore, the combined effect of a specific Plk1 inhibitor GSK461364A with gemcitabine was examined. We show that inhibition of Plk1 significantly potentiates the anti-neoplastic activity of gemcitabine in both cultured pancreatic cancer cells and Panc1-derived orthotopic pancreatic cancer xenograft tumors. Overall, our study demonstrates that co-targeting Plk1 can significantly enhance the efficacy of gemcitabine, offering a promising new therapeutic option for the treatment of gemcitabine-resistant human pancreatic cancer.
Collapse
Affiliation(s)
- Jie Li
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Ruixin Wang
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | | | - Anju Karki
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Yi Yang
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Yifan Kong
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI, USA
| | - Stephen F. Konieczny
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
- Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
- Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
26
|
Bose P, Grant S. Rational Combinations of Targeted Agents in AML. J Clin Med 2015; 4:634-664. [PMID: 26113989 PMCID: PMC4470160 DOI: 10.3390/jcm4040634] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/06/2015] [Indexed: 12/20/2022] Open
Abstract
Despite modest improvements in survival over the last several decades, the treatment of AML continues to present a formidable challenge. Most patients are elderly, and these individuals, as well as those with secondary, therapy-related, or relapsed/refractory AML, are particularly difficult to treat, owing to both aggressive disease biology and the high toxicity of current chemotherapeutic regimens. It has become increasingly apparent in recent years that coordinated interruption of cooperative survival signaling pathways in malignant cells is necessary for optimal therapeutic results. The modest efficacy of monotherapy with both cytotoxic and targeted agents in AML testifies to this. As the complex biology of AML continues to be elucidated, many “synthetic lethal” strategies involving rational combinations of targeted agents have been developed. Unfortunately, relatively few of these have been tested clinically, although there is growing interest in this area. In this article, the preclinical and, where available, clinical data on some of the most promising rational combinations of targeted agents in AML are summarized. While new molecules should continue to be combined with conventional genotoxic drugs of proven efficacy, there is perhaps a need to rethink traditional philosophies of clinical trial development and regulatory approval with a focus on mechanism-based, synergistic strategies.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Internal Medicine, Virginia Commonwealth University and VCU Massey Cancer Center Center, 1201 E Marshall St, MMEC 11-213, P.O. Box 980070, Richmond, VA 23298, USA; E-Mail:
| | - Steven Grant
- Departments of Internal Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, Human and Molecular Genetics and the Institute for Molecular Medicine, Virginia Commonwealth University and VCU Massey Cancer Center, 401 College St, P.O. Box 980035, Richmond, VA 23298, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-804-828-5211; Fax: +1-804-628-5920
| |
Collapse
|
27
|
Li Z, Lu Y, Ahmad N, Strebhardt K, Liu X. Low-dose arsenic-mediated metabolic shift is associated with activation of Polo-like kinase 1 (Plk1). Cell Cycle 2015; 14:3030-9. [PMID: 26292025 PMCID: PMC4825546 DOI: 10.1080/15384101.2015.1080397] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/26/2015] [Accepted: 08/02/2015] [Indexed: 10/23/2022] Open
Abstract
Arsenic is a well-established human carcinogen associated with cancers of the skin, liver, lung, kidney, and bladder. Although numerous carcinogenic pathways have been proposed, the molecular mechanisms underlying arsenic-associated cancer etiology are still elusive. The cellular responses to arsenic exposure are dose dependent. It was recently shown that low-dose arsenic leads to a metabolic shift from mitochondrial respiration to aerobic glycolysis via inactivation of tumor suppressor p53 and activation of NF-κB. However, how inactivation of p53, activation of NF-κB, and metabolic change are coordinated in response to low-dose arsenic exposure is still not completely understood. Polo-like kinase 1 (Plk1) is a well- documented regulator in many cell cycle-related events. Herein, we showed that low-dose arsenic leads to elevation of Plk1 in an NF-κB-dependent manner and that elevation of Plk1 contributes to the metabolic change from oxidative phosphorylation to glycolysis via activation of the PI3K/AKT/mTOR pathway. Furthermore, we showed that inhibition/depletion of Plk1 reverses low-dose arsenic-associated phenotypes, including enhanced cell proliferation, activation of the PI3K/AKT/mTOR pathway, and increased glycolysis. Finally, inhibition of the PI3K/AKT/mTOR pathway also antagonizes the enhanced glycolytic influx due to low-dose arsenic exposure. Our studies support the notion that Plk1 likely plays a critical role in cellular responses to low-dose arsenic.
Collapse
Affiliation(s)
- Zhiguo Li
- Department of Biochemistry; Purdue University; West Lafayette, IN USA
| | - Ying Lu
- Department of Biochemistry; Purdue University; West Lafayette, IN USA
- School of Public Health; Xinjiang Medical University; Urumqi, Xinjiang, China
| | - Nihal Ahmad
- Department of Dermatology; University of Wisconsin; Madison, WI USA
| | - Klaus Strebhardt
- Department of Obstetrics and Gynecology; J.W. Goethe University; Frankfurt, Germany
| | - Xiaoqi Liu
- Department of Biochemistry; Purdue University; West Lafayette, IN USA
- Center for Cancer Research; Purdue University; West Lafayette, IN USA
| |
Collapse
|
28
|
Bose P, Dai Y, Grant S. Histone deacetylase inhibitor (HDACI) mechanisms of action: emerging insights. Pharmacol Ther 2014; 143:323-336. [PMID: 24769080 PMCID: PMC4117710 DOI: 10.1016/j.pharmthera.2014.04.004] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 04/10/2014] [Indexed: 02/05/2023]
Abstract
Initially regarded as "epigenetic modifiers" acting predominantly through chromatin remodeling via histone acetylation, HDACIs, alternatively referred to as lysine deacetylase or simply deacetylase inhibitors, have since been recognized to exert multiple cytotoxic actions in cancer cells, often through acetylation of non-histone proteins. Some well-recognized mechanisms of HDACI lethality include, in addition to relaxation of DNA and de-repression of gene transcription, interference with chaperone protein function, free radical generation, induction of DNA damage, up-regulation of endogenous inhibitors of cell cycle progression, e.g., p21, and promotion of apoptosis. Intriguingly, this class of agents is relatively selective for transformed cells, at least in pre-clinical studies. In recent years, additional mechanisms of action of these agents have been uncovered. For example, HDACIs interfere with multiple DNA repair processes, as well as disrupt cell cycle checkpoints, critical to the maintenance of genomic integrity in the face of diverse genotoxic insults. Despite their pre-clinical potential, the clinical use of HDACIs remains restricted to certain subsets of T-cell lymphoma. Currently, it appears likely that the ultimate role of these agents will lie in rational combinations, only a few of which have been pursued in the clinic to date. This review focuses on relatively recently identified mechanisms of action of HDACIs, with particular emphasis on those that relate to the DNA damage response (DDR), and discusses synergistic strategies combining HDACIs with several novel targeted agents that disrupt the DDR or antagonize anti-apoptotic proteins that could have implications for the future use of HDACIs in patients with cancer.
Collapse
Affiliation(s)
- Prithviraj Bose
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yun Dai
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA; Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA; Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
29
|
Giráldez S, Herrero-Ruiz J, Mora-Santos M, Japón MÁ, Tortolero M, Romero F. SCF(FBXW7α) modulates the intra-S-phase DNA-damage checkpoint by regulating Polo like kinase-1 stability. Oncotarget 2014; 5:4370-83. [PMID: 24970797 PMCID: PMC4147330 DOI: 10.18632/oncotarget.2021] [Citation(s) in RCA: 19] [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: 03/28/2014] [Accepted: 05/26/2014] [Indexed: 12/22/2022] Open
Abstract
The intra-S-checkpoint is essential to control cell progression through S phase under normal conditions and in response to replication stress. When DNA lesions are detected, replication fork progression is blocked allowing time for repair to avoid genomic instability and the risk of cancer. DNA replication initiates at many origins of replication in eukaryotic cells, where a series of proteins form pre-replicative complexes (pre-RCs) that are activated to become pre-initiation complexes and ensure a single round of replication in each cell cycle. PLK1 plays an important role in the regulation of DNA replication, contributing to the regulation of pre-RCs formation by phosphorylating several proteins, under both normal and stress conditions. Here we report that PLK1 is ubiquitinated and degraded by SCFFBXW7α/proteasome. Moreover, we identified a new Cdc4 phosphodegron in PLK1, conserved from yeast to humans, whose mutation prevents PLK1 destruction. We established that endogenous SCFFBXW7α degrades PLK1 in the G1 and S phases of an unperturbed cell cycle and in S phase following UV irradiation. Furthermore, we showed that FBXW7α overexpression or UV irradiation prevented the loading of proteins onto chromatin to form pre-RCs and, accordingly, reduced cell proliferation. We conclude that PLK1 degradation mediated by SCFFBXW7α modulates the intra-S-phase checkpoint.
Collapse
Affiliation(s)
- Servando Giráldez
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| | - Joaquín Herrero-Ruiz
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| | - Mar Mora-Santos
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| | - Miguel Á. Japón
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Anatomía Patológica, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Maria Tortolero
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| | - Francisco Romero
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| |
Collapse
|
30
|
Abstract
Polo-like kinase-1 (Plk1) belongs to a family of serine-threonine kinases and plays a critical role in mitotic progression. Plk1 involves in the initiation of mitosis, centrosome maturation, bipolar spindle formation, and cytokinesis, well-reported as traditional functions of Plk1. In this review, we discuss the role of Plk1 during DNA damage response beyond the functions in mitotsis. When DNA is damaged in cells under various stress conditions, the checkpoint mechanism is activated to allow cells to have enough time for repair. When damage is repaired, cells progress continuously their division, which is called checkpoint recovery. If damage is too severe to repair, cells undergo apoptotic pathway. If damage is not completely repaired, cells undergo a process called checkpoint adaptation, and resume cell division cycle with damaged DNA. Plk1 targets and regulates many key factors in the process of damage response, and we deal with these subjects in this review.
Collapse
Affiliation(s)
- Sun-Yi Hyun
- Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330-714, Korea
| | - Hyo-In Hwan
- Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330-714, Korea
| | - Young-Joo Jang
- Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330-714, Korea
| |
Collapse
|
31
|
Chen P, Zhao X, Ma L. Downregulation of microRNA-100 correlates with tumor progression and poor prognosis in hepatocellular carcinoma. Mol Cell Biochem 2013; 383:49-58. [PMID: 23842624 DOI: 10.1007/s11010-013-1753-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 07/03/2013] [Indexed: 12/11/2022]
Abstract
Increasing evidence suggests that dysregulation of microRNAs is correlated with malignant transformation and tumor development. miR-100, a potential tumor suppressor, is downregulated by many human cancers. However, the expression and functions of miR-100 in hepatocellular carcinoma (HCC) are still unclear. The aim of this study was to detect the expression of miR-100 in HCC tissues and investigate its clinicopathological and prognostic significance. Also, the effects of miR-100 on growth and apoptosis of HCC cells and its potential molecular mechanisms were analyzed. Results showed that the expression level of miR-100 in HCC tissues was significantly lower than that in matched non-cancerous liver tissues. Also, low-miR-100 expression was observed to be significantly correlated with higher tumor grade, higher incidence of lymph node metastasis, advanced TNM stage and higher incidence of tumor recurrence in HCC patients. Multivariate survival analyses suggested that low-miR-100 expression was an independent prognostic factor for HCC patients (HR = 1.66, 95 % CI 1.32-2.82, P = 0.019). In addition, we found that upregulation of miR-100 could inhibit growth and increase apoptosis of HCC cells by downregulating polo-like kinase 1 (plk1). In HCC tissues, miR-100 expression was inversely correlated with the expression of plk1 protein (r = -0.418; P = 0.029). Therefore, downregulation of miR-100 was correlated with progressive pathological feature and poor prognosis in HCC patients, and miR-100 could function as a tumor suppressor by targeting plk1. miR-100 may serve as a prognostic marker and molecular therapeutic target in HCC.
Collapse
Affiliation(s)
- Ping Chen
- Department of Medical Oncology, The First People's Hospital of Yancheng, The Fourth Affiliated Hospital, Medical School of Nantong University, No. 14 Yuehe Road, Tinghu District, Yancheng, 224001, Jiangsu, People's Republic of China,
| | | | | |
Collapse
|
32
|
Gomes-da-Silva LC, Ramalho JS, Pedroso de Lima MC, Simões S, Moreira JN. Impact of anti-PLK1 siRNA-containing F3-targeted liposomes on the viability of both cancer and endothelial cells. Eur J Pharm Biopharm 2013; 85:356-64. [PMID: 23659854 DOI: 10.1016/j.ejpb.2013.04.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 04/03/2013] [Accepted: 04/09/2013] [Indexed: 01/31/2023]
Abstract
We have previously described the development of novel sterically stabilized F3-targeted pH-sensitive liposomes, which exhibited the ability to target both cancer and endothelial cells. Herein, the therapeutic potential of those liposomes was assessed upon encapsulation of a siRNA against a well-validated molecular target, PLK1. Treatment of prostate cancer (PC3) and angiogenic endothelial (HMEC-1) cells with F3-targeted liposomes containing anti-PLK1 siRNA resulted in a significant decrease in cell viability, which was mediated by a marked PLK1 silencing, both at the mRNA and protein levels. Furthermore, pre-treatment of PC3 cells with F3-targeted liposomes containing anti-PLK1 siRNA enabled a 3-fold reduction of paclitaxel IC50 and a 2.5-fold augment of the percentage of cancer cells in G2/mitosis arrest, which ultimately culminated in cell death. Overall, the F3-targeted nanocarrier containing an anti-PLK1 siRNA might constitute a valuable system for prostate cancer treatment, either applied in a single schedule or combined with conventional chemotherapy.
Collapse
Affiliation(s)
- Lígia C Gomes-da-Silva
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Portugal
| | | | | | | | | |
Collapse
|
33
|
Kim HJ, Cho JH, Kim JR. Downregulation of Polo-like kinase 1 induces cellular senescence in human primary cells through a p53-dependent pathway. J Gerontol A Biol Sci Med Sci 2013; 68:1145-56. [PMID: 23525475 DOI: 10.1093/gerona/glt017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Polo-like kinase 1 (PLK1) plays a key role in various stages of mitosis from entry into M phase to exit from mitosis. However, its role in cellular senescence remains to be determined. Therefore, the effects of PLK1 on cellular senescence in human primary cells were investigated. We found that expression of PLK1 decreased in human dermal fibroblasts and human umbilical vein endothelial cells under replicative senescence and premature senescence induced by adriamycin. PLK1 knockdown with PLK1 small interfering RNAs in young cells induced premature senescence. In contrast, upregulation of PLK1 in old cells partially reversed senescence phenotypes. Cellular senescence by PLK1 inhibition was observed in p16 knockdown cells but not in p53 knockdown cells. Our data suggest that PLK1 repression might result in cellular senescence in human primary cells via a p53-dependent pathway.
Collapse
Affiliation(s)
- Hee-Jin Kim
- These authors contributed equally to this work
| | | | | |
Collapse
|
34
|
Hou X, Li Z, Huang W, Li J, Staiger C, Kuang S, Ratliff T, Liu X. Plk1-dependent microtubule dynamics promotes androgen receptor signaling in prostate cancer. Prostate 2013; 73:1352-63. [PMID: 23661607 PMCID: PMC3720713 DOI: 10.1002/pros.22683] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 04/08/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND The androgen receptor (AR) signaling continues to be essential in castrate-resistant prostate cancer (CRPC). Taxel-based chemotherapy is the current standard treatment for CRPC patients. Unfortunately, almost all patients eventually develop resistance toward this chemotherapy. Significantly, it was recently found that the anti-tumor effect of paclitaxel in CRPC is due to its inhibition of AR activity via its inhibition of microtubule dynamics. Polo-like kinase 1 (Plk1), a critical regulator in many cell cycle events, is elevated in prostate cancer (PCa) and linked to tumor grades. Of note, we have previously shown that Plk1 phosphorylates CLIP-170 and p150(Glued) , two important regulators of microtubule dynamics. METHODS We compared paclitaxel-mediated phenotypes (inhibition of the AR signaling, decrease of microtubule dynamics and cell death) of PCa cells expressing different forms of CLIP-170 and p150(Glued) with different Plk1 phosphorylation states. RESULTS We show that Plk1 phosphorylation of CLIP-170 and p150(Glued) affects cellular responses to paclitaxel. Expression of Plk1-unphosphorylatable mutants of CLIP-170 and p150(Glued) results in increased paclitaxel-induced apoptosis, increased protein degradation of the AR, and decreased nuclear accumulation of the AR in response to androgen in prostate cancer cells. Finally, we show that cells expressing unphosphorylatable mutants of CLIP-170 have defective microtubule dynamics, thus providing a new mechanism to understand how Plk1-associated kinase activity promotes constitutive activation of AR signaling in CRPC. CONCLUSIONS Our data suggest that a combination of inhibition of Plk1 and paclitaxel might be a novel avenue for treatment of CRPC.
Collapse
Affiliation(s)
- Xianzeng Hou
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907
- Department of Neurosurgery, Provincial Hospital affiliated to Shandong University, Jinan, China 250021
| | - Zhiguo Li
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907
| | - Weize Huang
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907
| | - Jiejie Li
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Christopher Staiger
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Tim Ratliff
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907
| |
Collapse
|
35
|
Bibi N, Parveen Z, Rashid S. Identification of potential Plk1 targets in a cell-cycle specific proteome through structural dynamics of kinase and Polo box-mediated interactions. PLoS One 2013; 8:e70843. [PMID: 23967120 PMCID: PMC3744538 DOI: 10.1371/journal.pone.0070843] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/23/2013] [Indexed: 01/15/2023] Open
Abstract
Polo like kinase 1 (Plk1) is a key player in orchestrating the wide variety of cell-cycle events ranging from centrosome maturation, mitotic entry, checkpoint recovery, transcriptional control, spindle assembly, mitotic progression, cytokinesis and DNA damage checkpoints recovery. Due to its versatile nature, Plk1 is considered an imperative regulator to tightly control the diverse aspects of the cell cycle network. Interactions among Plk1 polo box domain (PBD) and its putative binding proteins are crucial for the activation of Plk1 kinase domain (KD). To date, only a few substrate candidates have been characterized through the inclusion of both polo box and kinase domain-mediated interactions. Thus it became compelling to explore precise and specific Plk1 substrates through reassessment and extension of the structure-function paradigm. To narrow this apparently wide gap in knowledge, here we employed a thorough sequence search of Plk1 phosphorylation signature containing proteins and explored their structure-based features like conceptual PBD-binding capabilities and subsequent recruitment of KD directed phosphorylation to dissect novel targets of Plk1. Collectively, we identified 4,521 phosphodependent proteins sharing similarity to the consensus phosphorylation and PBD recognition motifs. Subsequent application of filters including similarity index, Gene Ontology enrichment and protein localization resulted in stringent pre-filtering of irrelevant candidates and isolated unique targets with well-defined roles in cell-cycle machinery and carcinogenesis. These candidates were further refined structurally using molecular docking and dynamic simulation assays. Overall, our screening approach enables the identification of several undefined cell-cycle associated functions of Plk1 by uncovering novel phosphorylation targets.
Collapse
Affiliation(s)
- Nousheen Bibi
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Zahida Parveen
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sajid Rashid
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
36
|
Deeraksa A, Pan J, Sha Y, Liu XD, Eissa NT, Lin SH, Yu-Lee LY. Plk1 is upregulated in androgen-insensitive prostate cancer cells and its inhibition leads to necroptosis. Oncogene 2013; 32:2973-83. [PMID: 22890325 PMCID: PMC3499666 DOI: 10.1038/onc.2012.309] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 05/29/2012] [Accepted: 06/10/2012] [Indexed: 12/18/2022]
Abstract
Castration-resistant prostate cancer (PCa) is refractory to hormone therapy and new strategies for treatment are urgently needed. We found that androgen-insensitive (AI) PCa cells, LNCaP-AI, are reprogrammed to upregulate the mitotic kinase Plk1 (Polo-like kinase 1) and other M-phase cell-cycle proteins, which may underlie AI PCa growth. In androgen-depleted media, LNCaP-AI cells showed exquisite sensitivity to growth inhibition by subnanomolar concentrations of a small molecule inhibitor of Plk1, BI2536, suggesting that these cells are dependent on Plk1 for growth. In contrast, the androgen-responsive parental LNCaP cells showed negligible responses to BI2536 treatment under the same condition. BI2536 treatment of LNCaP-AI cells resulted in an increase in cell death marker PARP-1 (polymerase-1) but did not activate caspase-3, an apoptosis marker, suggesting that the observed cell death was caspase-independent. BI2536-treated LNCaP-AI cells formed multinucleated giant cells that contain clusters of nuclear vesicles indicative of mitotic catastrophe. Live-cell time-lapse imaging revealed that BI2536-treated giant LNCaP-AI cells underwent necroptosis, as evidenced by 'explosive' cell death and partial reversal of cell death by a necroptosis inhibitor. Our studies suggest that LNCaP-AI cells underwent reprogramming in both their cell growth and cell death pathways, rendering them highly sensitive to Plk1 inhibition that induces necroptosis. Harnessing necroptosis through Plk1 inhibition may be explored for therapeutic intervention of castration-resistant PCa.
Collapse
Affiliation(s)
- Arpaporn Deeraksa
- Department of Medicine, Section of Immunology Allergy and Rheumatology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jing Pan
- Department of Medicine, Section of Immunology Allergy and Rheumatology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Youbao Sha
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Xian-De Liu
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - N Tony Eissa
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Sue-Hwa Lin
- Department of Molecular Pathology, UT Texas at M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Li-yuan Yu-Lee
- Department of Medicine, Section of Immunology Allergy and Rheumatology, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
37
|
Wissing MD, van Diest PJ, van der Wall E, Gelderblom H. Antimitotic agents for the treatment of patients with metastatic castrate-resistant prostate cancer. Expert Opin Investig Drugs 2013; 22:635-61. [PMID: 23586879 DOI: 10.1517/13543784.2013.789858] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Metastatic castrate-resistant prostate cancer (mCRPC) is the second deadliest cancer in men. The group of taxanes, which target microtubules of mitotic cells, is currently the only chemotherapy which has proven to increase overall survival in mCRPC patients. Other mitotic inhibitors are being explored for their clinical potential in mCRPC treatment. AREAS COVERED In this review, we summarize recent developments in the application of mitotic inhibitors for mCRPC from a clinical perspective. The four main groups of mitotic inhibitors currently being tested in clinical trials are microtubule-inhibitors, polo-like kinase 1 inhibitors, aurora kinase inhibitors and kinesin-spindle protein inhibitors. Compounds of these groups of inhibitors that are in clinical development for mCRPC are discussed. For this extensive overview, relevant literature was searched in PubMed and retrieved from clinicaltrials.gov and presentations at ASCO/AACR meetings. EXPERT OPINION In general, mitotic inhibitors are clinically well tolerated but exert limited antitumor activity compared to preclinical study results. However, efficacy of mitotic inhibitors is improving, either by personalizing treatment, by introducing more active compounds, by decreasing resistance of cancer cells against mitotic inhibitors or by using mitotic inhibitors in combination therapies.
Collapse
Affiliation(s)
- Michel D Wissing
- Department of Clinical Oncology, Leiden University Medical Center, Albinusdreef 2 K1-62, 2333ZA Leiden, The Netherlands
| | | | | | | |
Collapse
|
38
|
Wissing MD, Carducci MA, Gelderblom H, van Diest PJ. Tales of how great drugs were brought down by a flawed rationale--letter. Clin Cancer Res 2013; 19:1303. [PMID: 23393074 DOI: 10.1158/1078-0432.ccr-12-2695] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Dasmahapatra G, Patel H, Nguyen T, Attkisson E, Grant S. PLK1 inhibitors synergistically potentiate HDAC inhibitor lethality in imatinib mesylate-sensitive or -resistant BCR/ABL+ leukemia cells in vitro and in vivo. Clin Cancer Res 2013; 19:404-14. [PMID: 23204129 PMCID: PMC3548959 DOI: 10.1158/1078-0432.ccr-12-2799] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine whether Polo-like kinase 1 (PLK1) inhibitors (e.g., BI2536) and histone deacetylase (HDAC) inhibitors (e.g., vorinostat) interact synergistically in the BCR/ABL(+) leukemia cells sensitive or resistant to imatinib mesylate (IM) in vitro and in vivo. EXPERIMENTAL DESIGN K562 and LAMA84 cells sensitive or resistant to imatinib mesylate and primary CML cells were exposed to BI2536 and vorinostat. Effects on cell viability and signaling pathways were determined using flow cytometry, Western blotting, and gene transfection. K562 and BV173/E255K animal models were used to test in vivo efficacy. RESULTS Cotreatment with BI2536 and vorinostat synergistically induced cell death in parental or imatinib mesylate-resistant BCR/ABL(+) cells and primary CD34(+) bone marrow cells but was minimally toxic to normal cells. BI2536/vorinostat cotreatment triggered pronounced mitochondrial dysfunction, inhibition of p-BCR/ABL, caspase activation, PARP cleavage, reactive oxygen species (ROS) generation, and DNA damage (manifest by increased expression of γH2A.X, p-ATM, p-ATR), events attenuated by the antioxidant TBAP. PLK1 short hairpin RNA (shRNA) knockdown significantly increased HDACI lethality, whereas HDAC1-3 shRNA knockdown reciprocally increased BI2536-induced apoptosis. Genetic interruption of the DNA damage linker H1.2 partially but significantly reduced PLK1/HDAC inhibitor-mediated cell death, suggesting a functional role for DNA damage in lethality. Finally, BI2536/vorinostat cotreatment dramatically reduced tumor growth in both subcutaneous and systemic BCR/ABL(+) leukemia xenograft models and significantly enhanced animal survival. CONCLUSIONS These findings suggest that concomitant PLK1 and HDAC inhibition is active against imatinib mesylate-sensitive or refractory CML and ALL cells both in vitro and in vivo and that this strategy warrants further evaluation in the setting of BCR/ABL(+) leukemias.
Collapse
Affiliation(s)
- Girija Dasmahapatra
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Hiral Patel
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tri Nguyen
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Elisa Attkisson
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Steven Grant
- Division of Hematology/Oncology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
- Virginia Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
40
|
RSK promotes G2 DNA damage checkpoint silencing and participates in melanoma chemoresistance. Oncogene 2012; 32:4480-9. [PMID: 23108403 DOI: 10.1038/onc.2012.472] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 07/17/2012] [Accepted: 08/29/2012] [Indexed: 12/30/2022]
Abstract
The incidence of malignant melanoma is growing rapidly worldwide and there is still no effective therapy for metastatic disease. This type of cancer is highly resistant to conventional DNA-damaging chemotherapeutics, and intense research has been dedicated for understanding the molecular pathways underlying chemoresistance. The Ras/mitogen-activated protein kinase (MAPK) signalling pathway is often deregulated in melanoma, which frequently harbours activating mutations in NRAS or BRAF. Herein, we demonstrate that the MAPK-activated protein kinase RSK (p90 ribosomal S6 kinase) contributes to melanoma chemoresistance by altering their response to chemotherapeutic agents. We find that RSK phosphorylates checkpoint kinase 1 (Chk1) at an inhibitory site, Ser280, both in vitro and in vivo. Our results indicate that RSK is the predominant protein kinase operating downstream of mitogens and oncogenes of the Ras/MAPK pathway, and consistent with this, we find that RSK constitutively phosphorylates Chk1 in melanoma. We show that RSK inhibition increases Chk1 activity in response to DNA-damaging agents, suggesting that the Ras/MAPK pathway modulates Chk1 function and the response to DNA damage. Accordingly, we demonstrate that RSK promotes G2 DNA damage checkpoint silencing in a Chk1-dependent manner, and find that RSK inhibitors sensitize melanoma cells to DNA-damaging agents. Together, our results identify a novel link between the Ras/MAPK pathway and the DNA damage response, and suggest that RSK inhibitors may be used to modulate chemosensitivity, which is one of the major obstacles to melanoma treatment.
Collapse
|
41
|
Iliuk A, Liu XS, Xue L, Liu X, Tao WA. Chemical visualization of phosphoproteomes on membrane. Mol Cell Proteomics 2012; 11:629-39. [PMID: 22593177 PMCID: PMC3434771 DOI: 10.1074/mcp.o112.018010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 05/10/2012] [Indexed: 11/06/2022] Open
Abstract
With new discoveries of important roles of phosphorylation on a daily basis, phospho-specific antibodies, as the primary tool for on-membrane detection of phosphoproteins, face enormous challenges. To address an urgent need for convenient and reliable analysis of phosphorylation events, we report a novel strategy for sensitive phosphorylation analysis in the Western blotting format. The chemical reagent, which we termed pIMAGO, is based on a multifunctionalized soluble nanopolymer and is capable of selectively binding to phosphorylated residues independent of amino acid microenvironment, thus offering great promise as a universal tool in biological analyses where the site of phosphorylation is not known or its specific antibody is not available. The specificity and sensitivity of the approach was first examined using a mixture of standard proteins. The method was then applied to monitor phosphorylation changes in in vitro kinase and phosphatase assays. Finally, to demonstrate the unique ability of pIMAGO to measure endogenous phosphorylation, we used it to visualize and determine the differences in phosphorylated proteins that interact with wild-type and kinase dead mutant of Polo-like kinase 1 during mitosis, the results of which were further confirmed by a quantitative phosphoproteomics experiment.
Collapse
Affiliation(s)
- Anton Iliuk
- From the Department of Biochemistry and Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - X. Shawn Liu
- From the Department of Biochemistry and Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - Liang Xue
- From the Department of Biochemistry and Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - Xiaoqi Liu
- From the Department of Biochemistry and Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - W. Andy Tao
- From the Department of Biochemistry and Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
42
|
Salvi M, Trashi E, Cozza G, Franchin C, Arrigoni G, Pinna LA. Investigation on PLK2 and PLK3 substrate recognition. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2012; 1824:1366-73. [PMID: 22828320 DOI: 10.1016/j.bbapap.2012.07.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 07/13/2012] [Accepted: 07/16/2012] [Indexed: 10/28/2022]
Abstract
Analyses of human phosphoproteome based on primary structure of the aminoacids surrounding the phosphor Ser/Thr suggest that a significant proportion of phosphosites is generated by a restricted number of acidophilic kinases, among which protein kinase CK2 plays a prominent role. Recently, new acidophilic kinases belonging to the Polo like kinase family have been characterized, with special reference to PLK1, PLK2, and PLK3 kinases. While some progress has been made in deciphering the PLK1-dependent phosphoproteome, very little is known about the targets of PLK2 and PLK3 kinases. In this report by using an in vitro approach, consisting of cell lysate phosphorylation, phosphoprotein separation by 2D gel electrophoresis and mass spectrometry, we describe the identification of new potential substrates of PLK2 and PLK3 kinases. We have identified and validated as in vitro PLK2 and PLK3 substrates HSP90, GRP-94, β-tubulin, calumenin, and 14-3-3 epsilon. The phosphosites generated by PLK3 in these proteins have been identified by mass spectrometry analysis to get new insights about PLKs specificity determinants. These latter have been further corroborated by an in silico analysis of the PLKs substrate binding region.
Collapse
Affiliation(s)
- M Salvi
- Department of Biomedical Sciences, University of Padova, Italy.
| | | | | | | | | | | |
Collapse
|
43
|
Seeger-Nukpezah T, Liebau MC, Höpker K, Lamkemeyer T, Benzing T, Golemis EA, Schermer B. The centrosomal kinase Plk1 localizes to the transition zone of primary cilia and induces phosphorylation of nephrocystin-1. PLoS One 2012; 7:e38838. [PMID: 22701722 PMCID: PMC3372538 DOI: 10.1371/journal.pone.0038838] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 05/11/2012] [Indexed: 01/13/2023] Open
Abstract
Polo-like kinase (Plk1) plays a central role in regulating the cell cycle. Plk1-mediated phosphorylation is essential for centrosome maturation, and for numerous mitotic events. Although Plk1 localizes to multiple subcellular sites, a major site of action is the centrosomes, which supports mitotic functions in control of bipolar spindle formation. In G0 or G1 untransformed cells, the centriolar core of the centrosome differentiates into the basal body of the primary cilium. Primary cilia are antenna-like sensory organelles dynamically regulated during the cell cycle. Whether Plk1 has a role in ciliary biology has never been studied. Nephrocystin-1 (NPHP1) is a ciliary protein; loss of NPHP1 in humans causes nephronophthisis (NPH), an autosomal-recessive cystic kidney disease. We here demonstrate that Plk1 colocalizes with nephrocystin-1 to the transition zone of primary cilia in epithelial cells. Plk1 co-immunoprecipitates with NPHP1, suggesting it is part of the nephrocystin protein complex. We identified a candidate Plk1 phosphorylation motif (D/E-X-S/T-φ-X-D/E) in nephrocystin-1, and demonstrated in vitro that Plk1 phosphorylates the nephrocystin N-terminus, which includes the specific PLK1 phosphorylation motif. Further, induced disassembly of primary cilia rapidly evoked Plk1 kinase activity, while small molecule inhibition of Plk1 activity or RNAi-mediated downregulation of Plk1 limited the first and second phase of ciliary disassembly. These data identify Plk1 as a novel transition zone signaling protein, suggest a function of Plk1 in cilia dynamics, and link Plk1 to the pathogenesis of NPH and potentially other cystic kidney diseases.
Collapse
Affiliation(s)
- Tamina Seeger-Nukpezah
- Department of Developmental Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Max C. Liebau
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of Pediatrics, University of Cologne, Cologne, Germany
| | - Katja Höpker
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Tobias Lamkemeyer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Erica A. Golemis
- Department of Developmental Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
44
|
Hikichi Y, Honda K, Hikami K, Miyashita H, Kaieda I, Murai S, Uchiyama N, Hasegawa M, Kawamoto T, Sato T, Ichikawa T, Cao S, Nie Z, Zhang L, Yang J, Kuida K, Kupperman E. TAK-960, a novel, orally available, selective inhibitor of polo-like kinase 1, shows broad-spectrum preclinical antitumor activity in multiple dosing regimens. Mol Cancer Ther 2012; 11:700-9. [PMID: 22188812 DOI: 10.1158/1535-7163.mct-11-0762] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine protein kinase involved in key processes during mitosis. Human PLK1 has been shown to be overexpressed in various human cancers, and elevated levels of PLK1 have been associated with poor prognosis, making it an attractive target for anticancer therapy. TAK-960 [4-[(9-cyclopentyl-7,7-difluoro-5-methyl-6-oxo-6,7,8,9-tetrahydro-5H-pyrimido[4,5-b][1,4]diazepin-2-yl)amino]-2-fluoro-5-methoxy-N-(1-methylpiperidin-4-yl) benzamide] is a novel, investigational, orally bioavailable, potent, and selective PLK1 inhibitor that has shown activity in several tumor cell lines, including those that express multidrug-resistant protein 1 (MDR1). Consistent with PLK1 inhibition, TAK-960 treatment caused accumulation of G(2)-M cells, aberrant polo mitosis morphology, and increased phosphorylation of histone H3 (pHH3) in vitro and in vivo. TAK-960 inhibited proliferation of multiple cancer cell lines, with mean EC(50) values ranging from 8.4 to 46.9 nmol/L, but not in nondividing normal cells (EC(50) >1,000 nmol/L). The mutation status of TP53 or KRAS and MDR1 expression did not correlate with the potency of TAK-960 in the cell lines tested. In animal models, oral administration of TAK-960 increased pHH3 in a dose-dependent manner and significantly inhibited the growth of HT-29 colorectal cancer xenografts. Treatment with once daily TAK-960 exhibited significant efficacy against multiple tumor xenografts, including an adriamycin/paclitaxel-resistant xenograft model and a disseminated leukemia model. TAK-960 has entered clinical evaluation in patients with advanced cancers.
Collapse
MESH Headings
- 4-Aminobenzoic Acid/chemistry
- 4-Aminobenzoic Acid/pharmacology
- Administration, Oral
- Animals
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- Azepines/chemistry
- Azepines/pharmacology
- Biological Availability
- Cell Cycle Checkpoints/drug effects
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Drugs, Investigational/chemistry
- Drugs, Investigational/pharmacokinetics
- Drugs, Investigational/pharmacology
- Female
- HT29 Cells
- Histones/metabolism
- Humans
- K562 Cells
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, Nude
- Mice, SCID
- Molecular Structure
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Neoplasms/pathology
- Phosphorylation/drug effects
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacokinetics
- Protein Kinase Inhibitors/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/metabolism
- Xenograft Model Antitumor Assays
- Polo-Like Kinase 1
Collapse
Affiliation(s)
- Yuichi Hikichi
- Takeda Pharmaceutical Company Ltd., 26-1 Muraoka Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Luo J, Liu X. Polo-like kinase 1, on the rise from cell cycle regulation to prostate cancer development. Protein Cell 2012; 3:182-97. [PMID: 22447658 PMCID: PMC4875424 DOI: 10.1007/s13238-012-2020-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Accepted: 02/04/2012] [Indexed: 01/19/2023] Open
Abstract
Polo-like kinase 1 (Plk1), a well-characterized member of serine/threonine kinases Plk family, has been shown to play pivotal roles in mitosis and cytokinesis in eukaryotic cells. Recent studies suggest that Plk1 not only controls the process of mitosis and cytokinesis, but also, going beyond those previously described functions, plays critical roles in DNA replication and Pten null prostate cancer initiation. In this review, we briefly summarize the functions of Plk1 in mitosis and cytokinesis, and then mainly focus on newly discovered functions of Plk1 in DNA replication and in Pten-null prostate cancer initiation. Furthermore, we briefly introduce the architectures of human and mouse prostate glands and the possible roles of Plk1 in human prostate cancer development. And finally, the newly chemotherapeutic development of small-molecule Plk1 inhibitors to target Plk1 in cancer treatment and their translational studies are also briefly reviewed.
Collapse
Affiliation(s)
- Jijing Luo
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907 USA
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907 USA
| |
Collapse
|