1
|
Goleij P, Pourali G, Raisi A, Ravaei F, Golestan S, Abed A, Razavi ZS, Zarepour F, Taghavi SP, Ahmadi Asouri S, Rafiei M, Mousavi SM, Hamblin MR, Talei S, Sheida A, Mirzaei H. Role of Non-coding RNAs in the Response of Glioblastoma to Temozolomide. Mol Neurobiol 2025; 62:1726-1755. [PMID: 39023794 DOI: 10.1007/s12035-024-04316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/16/2024] [Indexed: 07/20/2024]
Abstract
Chemotherapy and radiotherapy are widely used in clinical practice across the globe as cancer treatments. Intrinsic or acquired chemoresistance poses a significant problem for medical practitioners and researchers, causing tumor recurrence and metastasis. The most dangerous kind of malignant brain tumor is called glioblastoma multiforme (GBM) that often recurs following surgery. The most often used medication for treating GBM is temozolomide chemotherapy; however, most patients eventually become resistant. Researchers are studying preclinical models that accurately reflect human disease and can be used to speed up drug development to overcome chemoresistance in GBM. Non-coding RNAs (ncRNAs) have been shown to be substantial in regulating tumor development and facilitating treatment resistance in several cancers, such as GBM. In this work, we mentioned the mechanisms of how different ncRNAs (microRNAs, long non-coding RNAs, circular RNAs) can regulate temozolomide chemosensitivity in GBM. We also address the role of these ncRNAs encapsulated inside secreted exosomes.
Collapse
Affiliation(s)
- Pouya Goleij
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahin Golestan
- Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atena Abed
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Sadat Razavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Zarepour
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Sahand Talei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hamed Mirzaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
2
|
Nikhil K, Shah K. The significant others of aurora kinase a in cancer: combination is the key. Biomark Res 2024; 12:109. [PMID: 39334449 PMCID: PMC11438406 DOI: 10.1186/s40364-024-00651-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
AURKA is predominantly famous as an essential mitotic kinase. Recent findings have also established its critical role in a plethora of other biological processes including ciliogenesis, mitochondrial dynamics, neuronal outgrowth, DNA replication and cell cycle progression. AURKA overexpression in numerous cancers is strongly associated with poor prognosis and survival. Still no AURKA-targeted drug has been approved yet, partially because of the associated collateral toxicity and partly due to its limited efficacy as a single agent in a wide range of tumors. Mechanistically, AURKA overexpression allows it to phosphorylate numerous pathological substrates promoting highly aggressive oncogenic phenotypes. Our review examines the most recent advances in AURKA regulation and focuses on 33 such direct cancer-specific targets of AURKA and their associated oncogenic signaling cascades. One of the common themes that emerge is that AURKA is often involved in a feedback loop with its substrates, which could be the decisive factor causing its sustained upregulation and hyperactivation in cancer cells, an Achilles heel not exploited before. This dynamic interplay between AURKA and its substrates offers potential opportunities for targeted therapeutic interventions. By targeting these substrates, it may be possible to disrupt this feedback loop to effectively reverse AURKA levels, thereby providing a promising avenue for developing safer AURKA-targeted therapeutics. Additionally, exploring the synergistic effects of AURKA inhibition with its other oncogenic and/or tumor-suppressor targets could provide further opportunities for developing effective combination therapies against AURKA-driven cancers, thereby maximizing its potential as a critical drug target.
Collapse
Affiliation(s)
- Kumar Nikhil
- Department of Chemistry, Purdue University Institute for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India.
| | - Kavita Shah
- Department of Chemistry, Purdue University Institute for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
3
|
Qi Y, Rezaeian AH, Wang J, Huang D, Chen H, Inuzuka H, Wei W. Molecular insights and clinical implications for the tumor suppressor role of SCF FBXW7 E3 ubiquitin ligase. Biochim Biophys Acta Rev Cancer 2024; 1879:189140. [PMID: 38909632 PMCID: PMC11390337 DOI: 10.1016/j.bbcan.2024.189140] [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: 04/12/2024] [Revised: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
FBXW7 is one of the most well-characterized F-box proteins, serving as substrate receptor subunit of SKP1-CUL1-F-box (SCF) E3 ligase complexes. SCFFBXW7 is responsible for the degradation of various oncogenic proteins such as cyclin E, c-MYC, c-JUN, NOTCH, and MCL1. Therefore, FBXW7 functions largely as a major tumor suppressor. In keeping with this notion, FBXW7 gene mutations or downregulations have been found and reported in many types of malignant tumors, such as endometrial, colorectal, lung, and breast cancers, which facilitate the proliferation, invasion, migration, and drug resistance of cancer cells. Therefore, it is critical to review newly identified FBXW7 regulation and tumor suppressor function under physiological and pathological conditions to develop effective strategies for the treatment of FBXW7-altered cancers. Since a growing body of evidence has revealed the tumor-suppressive activity and role of FBXW7, here, we updated FBXW7 upstream and downstream signaling including FBXW7 ubiquitin substrates, the multi-level FBXW7 regulatory mechanisms, and dysregulation of FBXW7 in cancer, and discussed promising cancer therapies targeting FBXW7 regulators and downstream effectors, to provide a comprehensive picture of FBXW7 and facilitate the study in this field.
Collapse
Affiliation(s)
- Yihang Qi
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jingchao Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daoyuan Huang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
4
|
Luo Y, Feng X, Lang W, Xu W, Wang W, Mei C, Ye L, Zhu S, Wang L, Zhou X, Zeng H, Ma L, Ren Y, Jin J, Xu R, Huang G, Tong H. Ectopic expression of the transcription factor ONECUT3 drives a complex karyotype in myelodysplastic syndromes. J Clin Invest 2024; 134:e172468. [PMID: 38386414 PMCID: PMC11014670 DOI: 10.1172/jci172468] [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: 05/26/2023] [Accepted: 02/20/2024] [Indexed: 02/24/2024] Open
Abstract
Chromosomal instability is a prominent biological feature of myelodysplastic syndromes (MDS), with over 50% of patients with MDS harboring chromosomal abnormalities or a complex karyotype (CK). Despite this observation, the mechanisms underlying mitotic and chromosomal defects in MDS remain elusive. In this study, we identified ectopic expression of the transcription factor ONECUT3, which is associated with CKs and poorer survival outcomes in MDS. ONECUT3-overexpressing cell models exhibited enrichment of several notable pathways, including signatures of sister chromosome exchange separation and mitotic nuclear division with the upregulation of INCENP and CDCA8 genes. Notably, dysregulation of chromosome passenger complex (CPC) accumulation, besides the cell equator and midbody, during mitotic phases consequently caused cytokinesis failure and defective chromosome segregation. Mechanistically, the homeobox (HOX) domain of ONECUT3, serving as the DNA binding domain, occupied the unique genomic regions of INCENP and CDCA8 and transcriptionally activated these 2 genes. We identified a lead compound, C5484617, that functionally targeted the HOX domain of ONECUT3, inhibiting its transcriptional activity on downstream genes, and synergistically resensitized MDS cells to hypomethylating agents. This study revealed that ONECUT3 promoted chromosomal instability by transcriptional activation of INCENP and CDCA8, suggesting potential prognostic and therapeutic roles for targeting high-risk MDS patients with a CK.
Collapse
Affiliation(s)
- Yingwan Luo
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaomin Feng
- Department of Cell Systems and Anatomy, Department of Pathology and Laboratory Medicine, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
| | - Wei Lang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weihong Xu
- Stanford Genome Technology Center, Palo Alto, California, USA
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Nansha District, Guangzhou, China
| | - Wei Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chen Mei
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Ye
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuanghong Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lu Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinping Zhou
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huimin Zeng
- Department of Cell Systems and Anatomy, Department of Pathology and Laboratory Medicine, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
- Department of Pediatrics, Peking University People’s Hospital, Beijing, China
| | - Liya Ma
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanling Ren
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Rongzhen Xu
- Department of Hematology, The Second Affiliated Hospital, School of Medicine
| | - Gang Huang
- Department of Cell Systems and Anatomy, Department of Pathology and Laboratory Medicine, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, and
- Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Kouroukli O, Bravou V, Giannitsas K, Tzelepi V. Tissue-Based Diagnostic Biomarkers of Aggressive Variant Prostate Cancer: A Narrative Review. Cancers (Basel) 2024; 16:805. [PMID: 38398199 PMCID: PMC10887410 DOI: 10.3390/cancers16040805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Prostate cancer (PC) is a common malignancy among elderly men, characterized by great heterogeneity in its clinical course, ranging from an indolent to a highly aggressive disease. The aggressive variant of prostate cancer (AVPC) clinically shows an atypical pattern of disease progression, similar to that of small cell PC (SCPC), and also shares the chemo-responsiveness of SCPC. The term AVPC does not describe a specific histologic subtype of PC but rather the group of tumors that, irrespective of morphology, show an aggressive clinical course, dictated by androgen receptor (AR) indifference. AR indifference represents an adaptive response to androgen deprivation therapy (ADT), driven by epithelial plasticity, an inherent ability of tumor cells to adapt to their environment by changing their phenotypic characteristics in a bi-directional way. The molecular profile of AVPC entails combined alterations in the tumor suppressor genes retinoblastoma protein 1 (RB1), tumor protein 53 (TP53), and phosphatase and tensin homolog (PTEN). The understanding of the biologic heterogeneity of castration-resistant PC (CRPC) and the need to identify the subset of patients that would potentially benefit from specific therapies necessitate the development of prognostic and predictive biomarkers. This review aims to discuss the possible pathophysiologic mechanisms of AVPC development and the potential use of emerging tissue-based biomarkers in clinical practice.
Collapse
Affiliation(s)
- Olga Kouroukli
- Department of Pathology, Evaggelismos General Hospital, 10676 Athens, Greece
| | - Vasiliki Bravou
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, 26504 Patras, Greece;
| | | | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
6
|
Naseem Y, Zhang C, Zhou X, Dong J, Xie J, Zhang H, Agboyibor C, Bi Y, Liu H. Inhibitors Targeting the F-BOX Proteins. Cell Biochem Biophys 2023; 81:577-597. [PMID: 37624574 DOI: 10.1007/s12013-023-01160-1] [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] [Accepted: 08/04/2023] [Indexed: 08/26/2023]
Abstract
F-box proteins are involved in multiple cellular processes through ubiquitylation and consequent degradation of targeted substrates. Any significant mutation in F-box protein-mediated proteolysis can cause human malformations. The various cellular processes F-box proteins involved include cell proliferation, apoptosis, invasion, angiogenesis, and metastasis. To target F-box proteins and their associated signaling pathways for cancer treatment, researchers have developed thousands of F-box inhibitors. The most advanced inhibitor of FBW7, NVD-BK M120, is a powerful P13 kinase inhibitor that has been proven to bring about apoptosis in cancerous human lung cells by disrupting levels of the protein known as MCL1. Moreover, F-box Inhibitors have demonstrated their efficacy for treating certain cancers through targeting particular mutated proteins. This paper explores the key studies on how F-box proteins act and their contribution to malignancy development, which fabricates an in-depth perception of inhibitors targeting the F-box proteins and their signaling pathways that eventually isolate the most promising approach to anti-cancer treatments.
Collapse
Affiliation(s)
- Yalnaz Naseem
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Chaofeng Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Xinyi Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Jianshu Dong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China.
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| | - Jiachong Xie
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Huimin Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Clement Agboyibor
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - YueFeng Bi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China.
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| | - Hongmin Liu
- Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, China.
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
7
|
Ji Y, Zhang W, Shen K, Su R, Liu X, Ma Z, Liu B, Hu C, Xue Y, Xin Z, Yang Y, Li A, Jiang Z, Jing N, Zhu HH, Dong L, Zhu Y, Dong B, Pan J, Wang Q, Xue W. The ELAVL3/MYCN positive feedback loop provides a therapeutic target for neuroendocrine prostate cancer. Nat Commun 2023; 14:7794. [PMID: 38016952 PMCID: PMC10684895 DOI: 10.1038/s41467-023-43676-3] [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: 04/06/2023] [Accepted: 11/16/2023] [Indexed: 11/30/2023] Open
Abstract
Neuroendocrine prostate cancer is a rapidly progressive and lethal disease characterized by early visceral metastasis, poor prognosis, and limited treatment options. Uncovering the oncogenic mechanisms could lead to the discovery of potential therapeutic avenues. Here, we demonstrate that the RNA-binding protein ELAVL3 is specifically upregulated in neuroendocrine prostate cancer and that overexpression of ELAVL3 alone is sufficient to induce the neuroendocrine phenotype in prostate adenocarcinoma. Mechanistically, ELAVL3 is transcriptionally regulated by MYCN and subsequently binds to and stabilizes MYCN and RICTOR mRNA. Moreover, ELAVL3 is shown to be released in extracellular vesicles and induce neuroendocrine differentiation of adenocarcinoma cells via an intercellular mechanism. Pharmacological inhibition of ELAVL3 with pyrvinium pamoate, an FDA-approved drug, effectively suppresses tumor growth, reduces metastatic risk, and improves survival in neuroendocrine prostate cancer mouse models. Our results identify ELAVL3 as a critical regulator of neuroendocrine differentiation in prostate cancer and propose a drug repurposing strategy for targeted therapies.
Collapse
Affiliation(s)
- Yiyi Ji
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Weiwei Zhang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Kai Shen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Ruopeng Su
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Xinyu Liu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zehua Ma
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Bo Liu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Cong Hu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yizheng Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zhixiang Xin
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yi Yang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Ang Li
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zhou Jiang
- Department of Pathology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Na Jing
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Dong
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yinjie Zhu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Baijun Dong
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Jiahua Pan
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Qi Wang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China.
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
8
|
Imamura J, Ganguly S, Muskara A, Liao RS, Nguyen JK, Weight C, Wee CE, Gupta S, Mian OY. Lineage plasticity and treatment resistance in prostate cancer: the intersection of genetics, epigenetics, and evolution. Front Endocrinol (Lausanne) 2023; 14:1191311. [PMID: 37455903 PMCID: PMC10349394 DOI: 10.3389/fendo.2023.1191311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Androgen deprivation therapy is a cornerstone of treatment for advanced prostate cancer, and the development of castrate-resistant prostate cancer (CRPC) is the primary cause of prostate cancer-related mortality. While CRPC typically develops through a gain in androgen receptor (AR) signaling, a subset of CRPC will lose reliance on the AR. This process involves genetic, epigenetic, and hormonal changes that promote cellular plasticity, leading to AR-indifferent disease, with neuroendocrine prostate cancer (NEPC) being the quintessential example. NEPC is enriched following treatment with second-generation anti-androgens and exhibits resistance to endocrine therapy. Loss of RB1, TP53, and PTEN expression and MYCN and AURKA amplification appear to be key drivers for NEPC differentiation. Epigenetic modifications also play an important role in the transition to a neuroendocrine phenotype. DNA methylation of specific gene promoters can regulate lineage commitment and differentiation. Histone methylation can suppress AR expression and promote neuroendocrine-specific gene expression. Emerging data suggest that EZH2 is a key regulator of this epigenetic rewiring. Several mechanisms drive AR-dependent castration resistance, notably AR splice variant expression, expression of the adrenal-permissive 3βHSD1 allele, and glucocorticoid receptor expression. Aberrant epigenetic regulation also promotes radioresistance by altering the expression of DNA repair- and cell cycle-related genes. Novel therapies are currently being developed to target these diverse genetic, epigenetic, and hormonal mechanisms promoting lineage plasticity-driven NEPC.
Collapse
Affiliation(s)
- Jarrell Imamura
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Shinjini Ganguly
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Andrew Muskara
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Ross S. Liao
- Glickman Urologic Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jane K. Nguyen
- Glickman Urologic Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Christopher Weight
- Glickman Urologic Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Christopher E. Wee
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Shilpa Gupta
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Omar Y. Mian
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
9
|
Urabe F, Yamamoto Y, Kimura T. miRNAs in prostate cancer: Intercellular and extracellular communications. Int J Urol 2022; 29:1429-1438. [PMID: 36122303 DOI: 10.1111/iju.15043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/25/2022] [Indexed: 12/23/2022]
Abstract
Prostate cancer is the most prevalent male cancer in Western Europe and North America. Although new drugs were recently approved, clinical challenges such as accurately predicting and screening drug-resistant prostate cancer remain. microRNAs are short noncoding RNA molecules that participate in gene regulation at the post-transcriptional level by targeting messenger RNAs. There is accumulating evidence that intracellular microRNAs play important roles as promoters or inhibitors of prostate cancer progression. Additionally, recent studies showed that microRNAs are encapsulated in extracellular vesicles and shuttled into the extracellular space. Transfer of extracellular microRNAs contributes to intercellular communication between prostate cancer cells and components of the tumor microenvironment, which can promote prostate cancer progression. Furthermore, due to their encapsulation in extracellular vesicles, extracellular microRNAs can be stably present in body fluids which contain high levels of RNase. Thus, circulating microRNAs have great potential as noninvasive diagnostic and prognostic biomarkers for prostate cancer. Here, we summarize the roles of intracellular and extracellular microRNAs in prostate cancer progression and discuss the potential of microRNA-based therapeutics as a novel treatment strategy for prostate cancer.
Collapse
Affiliation(s)
- Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yusuke Yamamoto
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Wang F, Zhang H, Wang H, Qiu T, He B, Yang Q. Combination of AURKA inhibitor and HSP90 inhibitor to treat breast cancer with AURKA overexpression and TP53 mutations. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:180. [PMID: 36071247 DOI: 10.1007/s12032-022-01777-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022]
Abstract
Breast cancer is the most common cancer among women worldwide. Researches show that Aurora kinase A (AURKA) is highly expressed in approximately 73% of breast cancer patients, which induces drug resistance in breast cancer patients and decreases the median survival time. AURKA regulates spindle assembly, centrosome maturation, and chromosome alignment. AURKA overexpression affects the occurrence and development of breast cancer. Besides AURKA overexpression, heat shock protein 90 (HSP90) maintains the survival and proliferation of tumor cells by stabilizing the structure of oncoproteins, including P53 mutants (mtP53). TP53 mutations accounted for approximately 13%, 40%, 80%, 33%, 71%, and 82% of luminal A, Luminal B, Luminal C, normal basal-like, HER2-amplified, and basal-like breast cancers, respectively. TP53 mutation can aggravate cell genome instability and enhance the invasion, migration, and resistance of cancer cell. This review describes the research status of AURKA and HSP90 in breast cancer, summarizes the structure, function, and the chaperone cycle of HSP90, elaborates the interrelation between HSP90, mtP53, P53, and AURKA, and proposes the combination of HSP90 inhibitor and AURKA inhibitor to treat breast cancer. Targeting AURKA and HSP90 to treat cancer with AURKA overexpression and TP53 mutations will help improve the specificity and efficiency of breast cancer treatment and solve the problem of drug resistance.
Collapse
Affiliation(s)
- Fuping Wang
- Beijing Key Laboratory of Resistant Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100000, China
| | - Haotian Zhang
- Beijing Key Laboratory of Resistant Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100000, China
| | - Haitao Wang
- Department of Hematology, Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100000, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Binghong He
- Beijing Key Laboratory of Resistant Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100000, China
| | - Qiong Yang
- Beijing Key Laboratory of Resistant Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100000, China.
| |
Collapse
|
11
|
Burgess E, Livasy C, Trufan S, Zhu J, O'connor H, Hartman A, Clark P, Grigg C, Raghavan D. Clinical outcomes associated with expression of aurora kinase and p53 family members in muscle‑invasive bladder cancer. Mol Clin Oncol 2022; 16:102. [PMID: 35463214 PMCID: PMC9022081 DOI: 10.3892/mco.2022.2535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/02/2022] [Indexed: 11/21/2022] Open
Abstract
Biomarkers are needed in muscle-invasive bladder cancer (MIBC). We previously reported that high tumor aurora kinase (AURK) A expression identifies patients with MIBC with poor prognosis. Aberrant p53 expression has also been associated with poor outcomes in MIBC, though to the best of our knowledge, co-expression rates of p53 and aurora kinases have not been previously described in MIBC. As aurora kinase and p53 family members may co-regulate each other, the present study investigated whether tumor p53 or p63 protein expression influenced the prognostic value of AURKA in a pilot study of 50 patients with MIBC treated with curative intent. Immunohistochemistry for AURKA, AURKB, p53 and p63 were performed on archival pre-treatment tumor specimens and correlated with clinical outcomes in patients with MIBC who received neoadjuvant chemotherapy (NAC) prior to cystectomy. Baseline p53 [hazard ratio (HR) 1.46; 95% confidence interval (CI)=0.55-3.9; P=0.448) and p63 (HR 2.02; 95% CI=0.51-8.1; P=0.313) protein expression did not predict for overall survival (OS). Low p53 protein expression did not correlate with high AURKA (φ=0.190) or AURKB (φ=0.075) expression. However, in tumors with low p53 expression (n=17), the presence of either high AURKA or AURKB expression levels predicted an increased risk for relapse (HR 27.1; 95% CI=2.7-270.1; P=0.005) and mortality (HR 14.9; 95% CI=2.3-95.6; P=0.004) compared to tumors with both low AURKA and AURKB levels. The relationship between p63 and AURKA/B expression levels was not tested due to the prevalence (80%) of high p63 expression in the present cohort. In tumors with low AURKA expression, p53 status did not predict for OS (HR 0.62; 95% CI 0.2-3.2; P=0.572). In multivariable analysis, only high baseline AURKA expression predicted for inferior OS (HR 4.9; 95% CI 1.7-14.1; P=0.003). To the best of our knowledge, the present study was the first to report co-expression of p53 and aurora kinase family members in MIBC, and although wild-type p53 may regulate the aurora kinases in preclinical models, the adverse prognostic value of tumor AURKA overexpression was independent from baseline tumor p53 protein expression in the present cohort. AURKA remains an important prognostic biomarker in patients with MIBC and warrants further evaluation in prospective studies to validate whether baseline AURKA can identify patients that are unlikely to benefit from standard of care with NAC.
Collapse
Affiliation(s)
- Earle Burgess
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA
| | - Chad Livasy
- Carolinas Pathology Group, Charlotte, NC 28203, USA
| | - Sally Trufan
- Department of Cancer Biostatistics, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA
| | - Jason Zhu
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA
| | - Hazel O'connor
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA
| | | | - Peter Clark
- Department of Urology, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA
| | - Claud Grigg
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA
| | - Derek Raghavan
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA
| |
Collapse
|
12
|
Role of MicroRNAs in Neuroendocrine Prostate Cancer. Noncoding RNA 2022; 8:ncrna8020025. [PMID: 35447888 PMCID: PMC9029336 DOI: 10.3390/ncrna8020025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 11/25/2022] Open
Abstract
Therapy-induced neuroendocrine prostate cancer (t-NEPC/NEPC) is an aggressive variant of prostate cancer (PCa) that frequently emerges in castration-resistant prostate cancer (CRPC) under the selective pressure of androgen receptor (AR)-targeted therapies. This variant is extremely aggressive, metastasizes to visceral organs, tissues, and bones despite low serum PSA, and is associated with poor survival rates. It arises via a reversible trans-differentiation process, referred to as ‘neuroendocrine differentiation’ (NED), wherein PCa cells undergo a lineage switch and exhibit neuroendocrine features, characterized by the expression of neuronal markers such as enolase 2 (ENO2), chromogranin A (CHGA), and synaptophysin (SYP). The molecular and cellular mechanisms underlying NED in PCa are complex and not clearly understood, which contributes to a lack of effective molecular biomarkers for diagnosis and therapy of this variant. NEPC is thought to derive from prostate adenocarcinomas by clonal evolution. A characteristic set of genetic alterations, such as dual loss of retinoblastoma (RB1) and tumor protein (TP53) tumor suppressor genes and amplifications of Aurora kinase A (AURKA), NMYC, and EZH2, has been reported to drive NEPC. Recent evidence suggests that microRNAs (miRNAs) are important epigenetic players in driving NED in advanced PCa. In this review, we highlight the role of miRNAs in NEPC. These studies emphasize the diverse role that miRNAs play as oncogenes and tumor suppressors in driving NEPC. These studies have unveiled the important role of cellular processes such as the EMT and cancer stemness in determining NED in PCa. Furthermore, miRNAs are involved in intercellular communication between tumor cells and stromal cells via extracellular vesicles/exosomes that contribute to lineage switching. Recent studies support the promising potential of miRNAs as novel diagnostic biomarkers and therapeutic targets for NEPC.
Collapse
|
13
|
Fan J, Bellon M, Ju M, Zhao L, Wei M, Fu L, Nicot C. Clinical significance of FBXW7 loss of function in human cancers. Mol Cancer 2022; 21:87. [PMID: 35346215 PMCID: PMC8962602 DOI: 10.1186/s12943-022-01548-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/22/2022] [Indexed: 12/13/2022] Open
Abstract
FBXW7 (F-Box and WD Repeat Domain Containing 7) (also referred to as FBW7 or hCDC4) is a component of the Skp1-Cdc53 / Cullin-F-box-protein complex (SCF/β-TrCP). As a member of the F-box protein family, FBXW7 serves a role in phosphorylation-dependent ubiquitination and proteasome degradation of oncoproteins that play critical role(s) in oncogenesis. FBXW7 affects many regulatory functions involved in cell survival, cell proliferation, tumor invasion, DNA damage repair, genomic instability and telomere biology. This thorough review of current literature details how FBXW7 expression and functions are regulated through multiple mechanisms and how that ultimately drives tumorigenesis in a wide array of cell types. The clinical significance of FBXW7 is highlighted by the fact that FBXW7 is frequently inactivated in human lung, colon, and hematopoietic cancers. The loss of FBXW7 can serve as an independent prognostic marker and is significantly correlated with the resistance of tumor cells to chemotherapeutic agents and poorer disease outcomes. Recent evidence shows that genetic mutation of FBXW7 differentially affects the degradation of specific cellular targets resulting in a distinct and specific pattern of activation/inactivation of cell signaling pathways. The clinical significance of FBXW7 mutations in the context of tumor development, progression, and resistance to therapies as well as opportunities for targeted therapies is discussed.
Collapse
Affiliation(s)
- Jingyi Fan
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.,Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.,Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Marcia Bellon
- Department of Pathology and Laboratory Medicine, Center for Viral Pathogenesis, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Mingyi Ju
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.,Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.,Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.,Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
| | - Christophe Nicot
- Department of Pathology and Laboratory Medicine, Center for Viral Pathogenesis, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
14
|
Merkens L, Sailer V, Lessel D, Janzen E, Greimeier S, Kirfel J, Perner S, Pantel K, Werner S, von Amsberg G. Aggressive variants of prostate cancer: underlying mechanisms of neuroendocrine transdifferentiation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:46. [PMID: 35109899 PMCID: PMC8808994 DOI: 10.1186/s13046-022-02255-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
Prostate cancer is a hormone-driven disease and its tumor cell growth highly relies on increased androgen receptor (AR) signaling. Therefore, targeted therapy directed against androgen synthesis or AR activation is broadly used and continually improved. However, a subset of patients eventually progresses to castration-resistant disease. To date, various mechanisms of resistance have been identified including the development of AR-independent aggressive variant prostate cancer based on neuroendocrine transdifferentiation (NED). Here, we review the highly complex processes contributing to NED. Genetic, epigenetic, transcriptional aberrations and posttranscriptional modifications are highlighted and the potential interplay of the different factors is discussed. Background Aggressive variant prostate cancer (AVPC) with traits of neuroendocrine differentiation emerges in a rising number of patients in recent years. Among others, advanced therapies targeting the androgen receptor axis have been considered causative for this development. Cell growth of AVPC often occurs completely independent of the androgen receptor signal transduction pathway and cells have mostly lost the typical cellular features of prostate adenocarcinoma. This complicates both diagnosis and treatment of this very aggressive disease. We believe that a deeper understanding of the complex molecular pathological mechanisms contributing to transdifferentiation will help to improve diagnostic procedures and develop effective treatment strategies. Indeed, in recent years, many scientists have made important contributions to unravel possible causes and mechanisms in the context of neuroendocrine transdifferentiation. However, the complexity of the diverse molecular pathways has not been captured completely, yet. This narrative review comprehensively highlights the individual steps of neuroendocrine transdifferentiation and makes an important contribution in bringing together the results found so far.
Collapse
Affiliation(s)
- Lina Merkens
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Verena Sailer
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany
| | - Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Ella Janzen
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sarah Greimeier
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jutta Kirfel
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany
| | - Sven Perner
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany.,Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,Mildred Scheel Cancer Career Center Hamburg HaTRiCs4, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gunhild von Amsberg
- Department of Hematology and Oncology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,Martini-Klinik, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
15
|
Lee C, Chen Y, Hernandez E, Pong R, Ma S, Hofstad M, Kapur P, Zhau H, Chung LWK, Lai C, Lin H, Lee M, Raj GV, Hsieh J. The central role of Sphingosine kinase 1 in the development of neuroendocrine prostate cancer (NEPC): A new targeted therapy of NEPC. Clin Transl Med 2022; 12:e695. [PMID: 35184376 PMCID: PMC8858611 DOI: 10.1002/ctm2.695] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/09/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Neuroendocrine prostate cancer (NEPC) is often diagnosed as a sub-type from the castration-resistant prostate cancer (CRPC) recurred from the second generation of anti-androgen treatment and is a rapidly progressive fatal disease. The molecular mechanisms underlying the trans-differentiation from CRPC to NEPC are not fully characterized, which hampers the development of effective targeted therapy. METHODS Bioinformatic analyses were conducted to determine the clinical correlation of sphingosine kinase 1 (SphK1) in CRPC progression. To investigate the transcriptional regulation SphK1 and neuroendocrine (NE) transcription factor genes, both chromosome immunoprecipitation and luciferase reporter gene assays were performed. To demonstrate the role of SphK1 in NEPC development, neurosphere assay was carried out along with several biomarkers determined by quantitative PCR and western blot. Furthermore, in vivo NEPC xenograft models and patient-derived xenograft (PDX) model were employed to determine the effect of SphK1 inhibitors and target validation. RESULTS Significant prevalence of SphK1 in NEPC development is observed from clinical datasets. SphK1 is transcriptionally repressed by androgen receptor-RE1-silencing transcription factor (REST) complex. Furthermore, sphingosine 1-phosphate produced by SphK1 can modulate REST protein turnover via MAPK signaling pathway. Also, decreased REST protein levels enhance the expression of NE markers in CRPC, enabling the transition to NEPC. Finally, specific SphK1 inhibitors can effectively inhibit the growth of NEPC tumors and block the REST protein degradation in PDX. CONCLUSIONS SphK1 plays a central role in NEPC development, which offers a new target for this lethal cancer using clinically approved SphK1 inhibitors.
Collapse
Affiliation(s)
- Cheng‐Fan Lee
- Department of UrologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of Biochemistry and Molecular BiologyCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Yu‐An Chen
- Department of UrologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Elizabeth Hernandez
- Department of UrologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Rey‐Chen Pong
- Department of UrologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Shihong Ma
- Department of UrologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Mia Hofstad
- Department of UrologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Payal Kapur
- Urology and PathologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Haiyen Zhau
- Uro‐Oncology ResearchDepartment of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Leland WK Chung
- Uro‐Oncology ResearchDepartment of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Chih‐Ho Lai
- Department of Microbiology and ImmunologyGraduate Institute of Biomedical SciencesCollege of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Ho Lin
- Department of Life SciencesNational Chung Hsing UniversityTaichungTaiwan
| | - Ming‐Shyue Lee
- Department of Biochemistry and Molecular BiologyCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Ganesh V Raj
- Department of UrologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Jer‐Tsong Hsieh
- Department of UrologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
16
|
Slabáková E, Kahounová Z, Procházková J, Souček K. Regulation of Neuroendocrine-like Differentiation in Prostate Cancer by Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040075. [PMID: 34940756 PMCID: PMC8704250 DOI: 10.3390/ncrna7040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients’ expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.
Collapse
|
17
|
Hao J, Peng Q, Wang K, Yu G, Pan Y, Du X, Hu N, Zhang X, Qin Y, Li H. Antitumor Effect of Lenvatinib Combined with Alisertib in Hepatocellular Carcinoma by Targeting the DNA Damage Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6613439. [PMID: 34337035 PMCID: PMC8324353 DOI: 10.1155/2021/6613439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 06/12/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022]
Abstract
METHODS Immunohistochemical staining, sequencing, and genetic analysis of liver cancer tissues were performed. The antitumor efficacy of single-agent or combination treatment was measured by cell counting kit-8 assay and colony formation assays. Their antiproliferative and apoptosis activity is evaluated by cell cycle analyses and wound healing assays. The DNA-related proteins were also measured by Western blotting and immunohistochemical staining. The HepG2 xenograft model was used to detect the effects of lenvatinib-alisertib on the antitumor activity. RESULTS AURKA was found to be upregulated in HCC tissues (77.3%, 17/22). Combined alisertib and lenvatinib treatment significantly enhanced the inhibition of proliferation and migration in HepG2 and Hep3B cell lines compared to single-agent treatments (all Ps < 0.01). Alisertib alone or in combination with lenvatinib demonstrated a significant increase in the percentage of super-G2 cells (lenvatinib 1 μM vs. lenvatinib 1 μM + alisertib 0.1 μM 8.84 ± 0.84 vs. 34.0 ± 1.54, P < 0.001). Discontinuous spindles and missegregated chromosomes in HCC cells treated with alisertib in combination with lenvatinib were observed. We further revealed that combined treatment inhibited the expression of DNA damage pathway proteins compared to those of single-agent treatments. In nude mice, combined administration of alisertib combined with lenvatinib significantly enhanced the suppression of tumor growth and induced apoptosis (all Ps < 0.01). CONCLUSIONS Our findings provide evidence for the possible use of alisertib in combination with lenvatinib in the treatment of HCC for better therapeutic outcomes.
Collapse
Affiliation(s)
- Jianwen Hao
- Department of Radiology, Tianjin Chest Hospital, Tianjin 300350, China
| | - Qizhen Peng
- Department of Radiology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, 523000 Guangdong, China
| | - Keruo Wang
- Department of Diagnostics, Tianjin Medical University, Tianjin 300070, China
| | - Ge Yu
- Department of Hepatobiliary, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Clinical Research Center for Cancer, Tianjin 300070, China
| | - Yi Pan
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Clinical Research Center for Cancer, Tianjin 300070, China
| | - Xiaoling Du
- Department of Diagnostics, Tianjin Medical University, Tianjin 300070, China
| | - Na Hu
- Department of Diagnostics, Tianjin Medical University, Tianjin 300070, China
| | - Xuening Zhang
- Department of Radiology Second Hospital of Tianjin Medical University, Tianjin 300070, China
| | - Yu Qin
- Department of Diagnostics, Tianjin Medical University, Tianjin 300070, China
| | - Huikai Li
- Department of Hepatobiliary, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Clinical Research Center for Cancer, Tianjin 300070, China
| |
Collapse
|
18
|
Wang J, Li T, Wang B. Exosomal transfer of miR‑25‑3p promotes the proliferation and temozolomide resistance of glioblastoma cells by targeting FBXW7. Int J Oncol 2021; 59:64. [PMID: 34278448 PMCID: PMC8295027 DOI: 10.3892/ijo.2021.5244] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
Intrinsic or acquired resistance to temozolomide (TMZ) is a frequent occurrence in patients with glioblastoma (GBM). Accumulating evidence has indicated that the exosomal transfer of proteins and RNAs may confer TMZ resistance to recipient cells; however, the potential molecular mechanisms are not fully understood. Thus, the aim of the present study was to elucidate the possible role of exosomal microRNAs (miRNAs/miRs) in the acquired resistance to TMZ in GBM. A TMZ-resistant GBM cell line (A172R) was used, and exosomes derived from A172R cells were extracted. Exosomal miR-25-3p was identified as a miRNA associated with TMZ resistance. The potential functions of exosomal miR-25-3p were evaluated by reverse transcription-quantitative PCR, as well as cell viability, colony formation and soft agar assay, flow cytometry, western blot analysis, BrdU incorporation assay, tumor xenograft formation, luciferase reporter assay and RNA immunoprecipitation. It was found that A172R-derived exosomes promoted the proliferation and TMZ resistance of sensitive GBM cells. Moreover, miR-25-3p epxression was upregulated in the exosomes of A172R cells and in serum samples of patients with GBM treated with TMZ. The depletion of exosomal miR-25-3p partially abrogated the effects induced by the transfer of exosomes from A172R cells. By contrast, miR-25-3p overexpression facilitated the proliferation and TMZ resistance of sensitive GBM cells. F-box and WD repeat domain-containing-7 (FBXW7) was identified as a direct target of miR-25-3p. FBXW7 knockdown promoted the proliferation and TMZ resistance of GBM cells. Furthermore, the exosomal transfer of miR-25-3p promoted c-Myc and cyclin E expression by downregulating FBXW7. Our results provided a novel insight into exosomal microRNAs in acquired TMZ resistance of GBM cells. Besides, exosomal miR-25-3p might be a potential prognostic marker for GBM patients.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Neurosurgery, Henan Provincial People's Hospital, Henan Provincial Cerebrovascular Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Tianxiao Li
- Department of Intervention Therapy, Henan Provincial People's Hospital, Henan Provincial Cerebrovascular Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Bin Wang
- Department of Neurosurgery, Henan Provincial People's Hospital, Henan Provincial Cerebrovascular Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
19
|
Beltran H, Demichelis F. Therapy considerations in neuroendocrine prostate cancer: what next? Endocr Relat Cancer 2021; 28:T67-T78. [PMID: 34111024 PMCID: PMC8289743 DOI: 10.1530/erc-21-0140] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022]
Abstract
Lineage plasticity and histologic transformation to small cell neuroendocrine prostate cancer (NEPC) is an increasingly recognized mechanism of treatment resistance in advanced prostate cancer. This is associated with aggressive clinical features and poor prognosis. Recent work has identified genomic, epigenomic, and transcriptome changes that distinguish NEPC from prostate adenocarcinoma, pointing to new mechanisms and therapeutic targets. Treatment-related NEPC arises clonally from prostate adenocarcinoma during the course of disease progression, retaining early genomic events and acquiring new molecular features that lead to tumor proliferation independent of androgen receptor activity, and ultimately demonstrating a lineage switch from a luminal prostate cancer phenotype to a small cell neuroendocrine carcinoma. Identifying the subset of prostate tumors most vulnerable to lineage plasticity and developing strategies for earlier detection and intervention for patients with NEPC may ultimately improve prognosis. Clinical trials focused on drug targeting of the lineage plasticity process and/or NEPC will require careful patient selection. Here, we review emerging targets and discuss biomarker considerations that may be informative for the design of future clinical studies.
Collapse
Affiliation(s)
- Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| |
Collapse
|
20
|
Sankhe K, Prabhu A, Khan T. Design strategies, SAR, and mechanistic insight of Aurora kinase inhibitors in cancer. Chem Biol Drug Des 2021; 98:73-93. [PMID: 33934503 DOI: 10.1111/cbdd.13850] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/18/2021] [Accepted: 04/18/2021] [Indexed: 12/11/2022]
Abstract
Aurora kinases (AURKs) are serine/threonine protein kinases that play a critical role during cell proliferation. Three isoforms of AURKs reported in mammals include AURKA, AURKB, AURKC, and all share a similar C-terminal catalytic domain with differences in their subcellular location, substrate specificity, and function. Recent research reports indicate an elevated expression of these kinases in several cancer types highlighting their role as oncogenes in tumorigenesis. Inhibition of AURKs is an attractive strategy to design potent inhibitors modulating this target. The last few years have witnessed immense research in the development of AURK inhibitors with few FDA approvals. The current clinical therapeutic regime in cancer is associated with severe side-effects and emerging resistance to existing drugs. This has been the key driver of research initiatives toward designing more potent drugs that can potentially circumvent the emerging resistance. This review is a comprehensive summary of recent research on AURK inhibitors and presents the development of scaffolds, their synthetic schemes, structure-activity relationships, biological activity, and enzyme inhibition potential. We hope to provide the reader with an array of scaffolds that can be selected for further research work and mechanistic studies in the development of new AURK inhibitors.
Collapse
Affiliation(s)
- Kaksha Sankhe
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- Department of Pharmaceutical Chemistry and Quality Assurance, Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry and Quality Assurance, Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
21
|
Aurora Kinase B Inhibition: A Potential Therapeutic Strategy for Cancer. Molecules 2021; 26:molecules26071981. [PMID: 33915740 PMCID: PMC8037052 DOI: 10.3390/molecules26071981] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
Aurora kinase B (AURKB) is a mitotic serine/threonine protein kinase that belongs to the aurora kinase family along with aurora kinase A (AURKA) and aurora kinase C (AURKC). AURKB is a member of the chromosomal passenger protein complex and plays a role in cell cycle progression. Deregulation of AURKB is observed in several tumors and its overexpression is frequently linked to tumor cell invasion, metastasis and drug resistance. AURKB has emerged as an attractive drug target leading to the development of small molecule inhibitors. This review summarizes recent findings pertaining to the role of AURKB in tumor development, therapy related drug resistance, and its inhibition as a potential therapeutic strategy for cancer. We discuss AURKB inhibitors that are in preclinical and clinical development and combination studies of AURKB inhibition with other therapeutic strategies.
Collapse
|
22
|
Samaržija I. Post-Translational Modifications That Drive Prostate Cancer Progression. Biomolecules 2021; 11:247. [PMID: 33572160 PMCID: PMC7915076 DOI: 10.3390/biom11020247] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
While a protein primary structure is determined by genetic code, its specific functional form is mostly achieved in a dynamic interplay that includes actions of many enzymes involved in post-translational modifications. This versatile repertoire is widely used by cells to direct their response to external stimuli, regulate transcription and protein localization and to keep proteostasis. Herein, post-translational modifications with evident potency to drive prostate cancer are explored. A comprehensive list of proteome-wide and single protein post-translational modifications and their involvement in phenotypic outcomes is presented. Specifically, the data on phosphorylation, glycosylation, ubiquitination, SUMOylation, acetylation, and lipidation in prostate cancer and the enzymes involved are collected. This type of knowledge is especially valuable in cases when cancer cells do not differ in the expression or mutational status of a protein, but its differential activity is regulated on the level of post-translational modifications. Since their driving roles in prostate cancer, post-translational modifications are widely studied in attempts to advance prostate cancer treatment. Current strategies that exploit the potential of post-translational modifications in prostate cancer therapy are presented.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
23
|
Tang JF, Li GL, Zhang T, Du YM, Huang SY, Ran JH, Li J, Chen DL. Homoharringtonine inhibits melanoma cells proliferation in vitro and vivo by inducing DNA damage, apoptosis, and G2/M cell cycle arrest. Arch Biochem Biophys 2021; 700:108774. [PMID: 33548212 DOI: 10.1016/j.abb.2021.108774] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 01/03/2023]
Abstract
Homoharringtonine (HHT), an approved anti-leukemic alkaloid, has been reported effectively in many types of tumor cells. However, its effect on melanoma cells has not been investigated. And the anti-melanoma mechanism of HHT is still unknown. In this study, we detected the effects of HHT on two melanoma cell lines (A375 and B16F10) and on the A375 xenograft mouse model. HHT significantly inhibited the proliferation of melanoma cells as investigated by the CCK8 method, cell cloning assay, and EdU experiment. HHT induced A375 and B16F10 cells DNA damage, apoptosis, and G2/M cell cycle arrest as proved by TdT-mediated dUTP Nick-End Labeling (TUNEL) and flow cytometry assay. Additionally, the loss of mitochondrial membrane potential in HHT-treated cells were visualized by JC-1 fluorescent staining. For the molecule mechanism study, western blotting results indicated the protein expression levels of ATM, P53, p-P53, p-CHK2, γ-H2AX, PARP, cleaved-PARP, cleaved caspase-3, cleaved caspase-9, Bcl-2, Bax, Aurka, p-Aurka, Plk1, p-Plk1, Cdc25c, CDK1, cyclin B1, and Myt1 were regulated by HHT. And the relative mRNA expression level of Aurka, Plk1, Cdc25c, CDK1, cyclin B1, and Myt1 were ascertained by q-PCR assay. The results in vivo experiment showed that HHT can slow down the growth rate of tumors. At the same time, the protein expression levels in vivo were consistent with that in vitro. Collectively, our study provided evidence that HHT could be considered an effective anti-melanoma agent by inducing DNA damage, apoptosis, and cell cycle arrest.
Collapse
Affiliation(s)
- Jia-Feng Tang
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Chongqing Three Gorges Medical College, Chongqing, Wanzhou, PR China
| | - Guo-Li Li
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Chongqing Three Gorges Medical College, Chongqing, Wanzhou, PR China
| | - Tao Zhang
- Neuroscience Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, PR China; Chongqing Three Gorges Medical College, Chongqing, Wanzhou, PR China
| | - Yu-Mei Du
- College of Public Health and Management, Chongqing Medical University, Chongqing, PR China
| | - Shi-Ying Huang
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China
| | - Jian-Hua Ran
- Neuroscience Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, PR China
| | - Jing Li
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China.
| | - Di-Long Chen
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Chongqing Three Gorges Medical College, Chongqing, Wanzhou, PR China.
| |
Collapse
|
24
|
Wang Y, Yang Y, Gan Z, Zhao C, Lv C, Zhang Y, Zhao X. Role of AURKA in the hypothalamus-pituitary-testicular axis in Tibetan sheep from Tianzhu. Gen Comp Endocrinol 2021; 300:113617. [PMID: 32950578 DOI: 10.1016/j.ygcen.2020.113617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 11/20/2022]
Abstract
The mitosis-associated protein aurora kinase A (AURKA) regulates the maturation of germ cells. We have previously reported using transcriptome analysis that AURKA is expressed in yak testes. Although Tibetan sheep possess an immense economic value, their reproductive rate is low. Herein, the expression and functions of AURKA in the hypothalamus-pituitary-testicular (HPT) axis in Tibetan sheep from Tianzhu were investigated. The cDNA sequence of sheep AURKA was cloned and bioinformatics techniques were used to predict its structure. Tissue expression of AURKA was determined by qPCR, immunoblotting, immunostaining, and immunohistochemistry. The AURKA coding sequence was found to be 1218 bp in length, encoding a 405-amino acid polypeptide chain. Furthermore, the highest sequence similarity of AURKA with the corresponding sequence in other species was seen in goat and cattle; the least degree of similarity was seen in the domestic cat. In addition, AURKA expression was elevated in the testes compared to that in the hypothalamus and pituitary (p < 0.01). Moreover, AURKA was mainly localized in the hypothalamic paraventricular nucleus (magnocellular), chromophobe cells of the pituitary, and spermatogenic cells of the testis. These results indicated that AURKA might participate in sheep reproductive regulation, thus providing a reference for the study of AURKA function in the reproductive process of Tibetan sheep from Tianzhu.
Collapse
Affiliation(s)
- Yuanyuan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yang Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Ze Gan
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Caiying Zhao
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Chen Lv
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China.
| |
Collapse
|
25
|
Bhagirath D, Liston M, Patel N, Akoto T, Lui B, Yang TL, To DM, Majid S, Dahiya R, Tabatabai ZL, Saini S. MicroRNA determinants of neuroendocrine differentiation in metastatic castration-resistant prostate cancer. Oncogene 2020; 39:7209-7223. [PMID: 33037409 PMCID: PMC7718386 DOI: 10.1038/s41388-020-01493-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 02/08/2023]
Abstract
Therapy-induced neuroendocrine prostate cancer (NEPC), an extremely aggressive variant of castration-resistant prostate cancer (CRPC), is increasing in incidence with the widespread use of highly potent androgen receptor (AR)-pathway inhibitors (APIs) such as Enzalutamide (ENZ) and Abiraterone and arises via a reversible trans-differentiation process, referred to as neuroendocrine differentiation (NED). The molecular basis of NED is not completely understood leading to a lack of effective molecular markers for its diagnosis. Here, we demonstrate for the first time, that lineage switching to NE states is accompanied by key miRNA alterations including downregulation of miR-106a~363 cluster and upregulation of miR-301a and miR-375. To systematically investigate the key miRNAs alterations driving therapy-induced NED, we performed small RNA-NGS in a retrospective cohort of human metastatic CRPC clinical samples + PDX models with adenocarcinoma features (CRPC-adeno) vs those with neuroendocrine features (CRPC-NE). Further, with the application of machine learning algorithms to sequencing data, we trained a 'miRNA classifier' that could robustly classify 'CRPC-NE' from 'CRPC-Adeno' cases. The performance of classifier was validated in an additional cohort of mCRPC patients and publicly available PCa cohorts. Importantly, we demonstrate that miR-106a~363 cluster pleiotropically regulate cardinal nodal proteins instrumental in driving NEPC including Aurora Kinase A, N-Myc, E2F1 and STAT3. Our study has important clinical implications and transformative potential as our 'miRNA classifier' can be used as a molecular tool to stratify mCRPC patients into those with/without NED and guide treatment decisions. Further, we identify novel miRNA NED drivers that can be exploited for NEPC therapeutic targeting.
Collapse
Affiliation(s)
- Divya Bhagirath
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Michael Liston
- Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, CA, USA
| | - Nikhil Patel
- Department of Pathology, Augusta University, Augusta, GA, USA
| | - Theresa Akoto
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Byron Lui
- Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, CA, USA
| | - Thao Ly Yang
- Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, CA, USA
| | - Dat My To
- Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, CA, USA
| | - Shahana Majid
- Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, CA, USA
| | - Rajvir Dahiya
- Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, CA, USA
| | - Z Laura Tabatabai
- Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, CA, USA
| | - Sharanjot Saini
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA.
| |
Collapse
|
26
|
Epigenetic modulations and lineage plasticity in advanced prostate cancer. Ann Oncol 2020; 31:470-479. [PMID: 32139297 DOI: 10.1016/j.annonc.2020.02.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer is the most common cancer and second leading cause of cancer-related death in American men. Antiandrogen therapies are part of the standard of therapeutic regimen for advanced or metastatic prostate cancers; however, patients who receive these treatments are more likely to develop castration-resistant prostate cancer (CRPC) or neuroendocrine prostate cancer (NEPC). In the development of CRPC or NEPC, numerous genetic signaling pathways have been under preclinical investigations and in clinical trials. Accumulated evidence shows that DNA methylation, chromatin integrity, and accessibility for transcriptional regulation still play key roles in prostate cancer initiation and progression. Better understanding of how epigenetic change regulates the progression of prostate cancer and the interaction between epigenetic and genetic modulators driving NEPC may help develop a better risk stratification and more effective treatment regimens for prostate cancer patients.
Collapse
|
27
|
Bang S, Kaur S, Kurokawa M. Regulation of the p53 Family Proteins by the Ubiquitin Proteasomal Pathway. Int J Mol Sci 2019; 21:E261. [PMID: 31905981 PMCID: PMC6981958 DOI: 10.3390/ijms21010261] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/24/2019] [Indexed: 12/25/2022] Open
Abstract
The tumor suppressor p53 and its homologues, p63 and p73, play a pivotal role in the regulation of the DNA damage response, cellular homeostasis, development, aging, and metabolism. A number of mouse studies have shown that a genetic defect in the p53 family could lead to spontaneous tumor development, embryonic lethality, or severe tissue abnormality, indicating that the activity of the p53 family must be tightly regulated to maintain normal cellular functions. While the p53 family members are regulated at the level of gene expression as well as post-translational modification, they are also controlled at the level of protein stability through the ubiquitin proteasomal pathway. Over the last 20 years, many ubiquitin E3 ligases have been discovered that directly promote protein degradation of p53, p63, and p73 in vitro and in vivo. Here, we provide an overview of such E3 ligases and discuss their roles and functions.
Collapse
Affiliation(s)
| | | | - Manabu Kurokawa
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA; (S.B.); (S.K.)
| |
Collapse
|
28
|
Lin M, Xu Y, Gao Y, Pan C, Zhu X, Wang ZW. Regulation of F-box proteins by noncoding RNAs in human cancers. Cancer Lett 2019; 466:61-70. [DOI: 10.1016/j.canlet.2019.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/11/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022]
|
29
|
The Crucial Role of CXCL8 and Its Receptors in Colorectal Liver Metastasis. DISEASE MARKERS 2019; 2019:8023460. [PMID: 31827643 PMCID: PMC6886345 DOI: 10.1155/2019/8023460] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022]
Abstract
CXCL8 (also known as IL-8) can produce different biological effects by binding to its receptors: CXCR1, CXCR2, and the Duffy antigen receptor for chemokines (DARC). CXCL8 and its receptors are associated with the development of various tumor types, especially colorectal cancer and its liver metastases. In addition to promoting angiogenesis, proliferation, invasion, migration, and the survival of colorectal cancer (CRC) cells, CXCL8 and its receptors have also been known to induce the epithelial-mesenchymal transition (EMT) of CRC cells, to help them to escape host immunosurveillance as well as to enhance resistance to anoikis, which promotes the formation of circulating tumor cells (CTCs) and their colonization of distant organs. In this paper, we will review the established roles of CXCL8 signaling in CRC and discuss the possible strategies of targeting CXCL8 signaling for overcoming CRC drug resistance and cancer progression, including direct targeting of CXCL8/CXCR1/2 or indirect targeting through the inhibition of CXCL8-CXCR1/2 signaling.
Collapse
|
30
|
Hu J, Han B, Huang J. Morphologic Spectrum of Neuroendocrine Tumors of the Prostate: An Updated Review. Arch Pathol Lab Med 2019; 144:320-325. [PMID: 31644322 DOI: 10.5858/arpa.2019-0434-ra] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
CONTEXT.— The incidence of neuroendocrine tumors of the prostate increases after hormonal therapy. Neuroendocrine tumors possess a broad spectrum of morphologic features and pose challenges in the pathologic diagnosis and clinical management of patients. OBJECTIVE.— To present a brief updated summary of neuroendocrine tumors of the prostate with an overview of their histopathologic and immunohistochemical profiles and differential diagnoses. DATA SOURCES.— Literature review, personal experience in the daily practice of pathologic diagnosis, and laboratory research. CONCLUSIONS.— Our understanding of neuroendocrine tumors of the prostate classification and diagnosis continues to evolve. These advances benefit the risk stratification and management of prostate cancer.
Collapse
Affiliation(s)
- Jing Hu
- From the Department of Pathology, Shandong University QiLu Hospital, Jinan, China (Drs Hu and Han); the Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, China (Dr Han); and the Department of Pathology, Duke University School of Medicine, Durham, North Carolina (Dr Huang)
| | - Bo Han
- From the Department of Pathology, Shandong University QiLu Hospital, Jinan, China (Drs Hu and Han); the Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, China (Dr Han); and the Department of Pathology, Duke University School of Medicine, Durham, North Carolina (Dr Huang)
| | - Jiaoti Huang
- From the Department of Pathology, Shandong University QiLu Hospital, Jinan, China (Drs Hu and Han); the Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, China (Dr Han); and the Department of Pathology, Duke University School of Medicine, Durham, North Carolina (Dr Huang)
| |
Collapse
|
31
|
Kuai X, Li L, Chen R, Wang K, Chen M, Cui B, Zhang Y, Li J, Zhu H, Zhou H, Huang J, Qin J, Wang Z, Wei W, Gao D. SCF FBXW7/GSK3β-Mediated GFI1 Degradation Suppresses Proliferation of Gastric Cancer Cells. Cancer Res 2019; 79:4387-4398. [PMID: 31289136 DOI: 10.1158/0008-5472.can-18-4032] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/24/2019] [Accepted: 07/03/2019] [Indexed: 11/16/2022]
Abstract
Gastric cancer is the third leading cause of cancer-related death worldwide. The regulatory mechanisms underlying gastric cancer cell proliferation are largely unclear. Here, we show that the transcription factor GFI1 is associated with advanced clinical gastric cancer progression and promoted gastric cancer cell proliferation partially through inhibition of gastrokine-2 (GKN2) transcription. GFI1 was a degrading substrate of FBXW7, whose loss was observed in gastric cancer. Mechanistically, GSK3β-mediated GFI1 S94/S98 phosphorylation triggered its interaction with FBXW7, resulting in SCFFBXW7-mediated ubiquitination and degradation. A nondegradable GFI1 S94A/S98A mutant was more potent in driving gastric cancer cell proliferation and tumorigenesis than wild-type GFI1. Overall, this study reveals the oncogenic role of GFI1 in gastric cancer and provides mechanistic insights into the tumor suppressor function of FBXW7. SIGNIFICANCE: These findings demonstrate the oncogenic role of the transcription factor GFI1 and the tumor suppressive function of FBXW7 in gastric cancer.
Collapse
Affiliation(s)
- Xiaoling Kuai
- Department of Gastroenterology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Long Li
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ran Chen
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kangjunjie Wang
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Min Chen
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Binghai Cui
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuxue Zhang
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | - Hongwen Zhu
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianfei Huang
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Jun Qin
- The Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhiwei Wang
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Daming Gao
- State Key Laboratory of Cell Biology, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
32
|
Tripathi V, Kaur E, Kharat SS, Hussain M, Damodaran AP, Kulshrestha S, Sengupta S. Abrogation of FBW7α-dependent p53 degradation enhances p53's function as a tumor suppressor. J Biol Chem 2019; 294:13224-13232. [PMID: 31346036 DOI: 10.1074/jbc.ac119.008483] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
The gene encoding the tumor suppressor p53 is mutated in most cancers. p53 expression is known to be tightly controlled by several E3 ligases. Here, we show that F-box and WD repeat domain-containing 7α (FBW7α), the substrate-recognition component of the SCFFBW7 multiprotein E3 ligase complex, targets both WT and tumor-derived mutants of p53 for proteasomal degradation in multiple human cancer cell lines (HCT116 and U2OS). We found that lack of FBW7α stabilizes p53 levels, thereby increasing its half-life. p53 ubiquitylation and subsequent degradation require the F-box and the C-terminal WD40 repeats in FBW7α. The polyubiquitylation of p53 occurred via Lys-48 linkage and involved phosphorylation on p53 at Ser-33 and Ser-37 by glycogen synthase kinase 3β (GSK3β) and DNA-dependent protein kinase (DNA-PK), respectively. These phosphorylation events created a phosphodegron that enhanced p53 binding to FBW7α, allowing for the attachment of polyubiquitin moieties at Lys-132 in p53. FBW7α-dependent p53 polyubiquitylation apparently occurred during and immediately after DNA double-strand breaks induced by either doxorubicin or ionizing radiation. Accordingly, in cells lacking FBW7α, p53 induction was enhanced after DNA damage. Phosphodegron-mediated polyubiquitylation of p53 on Lys-132 had functional consequences, with cells in which FBW7α-mediated p53 degradation was abrogated exhibiting enhancement of their tumorigenic potential. We conclude that p53, which previously has been reported to transactivate FBW7, is also targeted by the same E3 ligase for degradation, suggesting the presence of a regulatory feedback loop that controls p53 levels and functions during DNA damage.
Collapse
Affiliation(s)
- Vivek Tripathi
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Ekjot Kaur
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Suhas Sampat Kharat
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Mansoor Hussain
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | | | - Swati Kulshrestha
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sagar Sengupta
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
33
|
Hickmann AK, Frick M, Hadaschik D, Battke F, Bittl M, Ganslandt O, Biskup S, Döcker D. Molecular tumor analysis and liquid biopsy: a feasibility investigation analyzing circulating tumor DNA in patients with central nervous system lymphomas. BMC Cancer 2019; 19:192. [PMID: 30823914 PMCID: PMC6397454 DOI: 10.1186/s12885-019-5394-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 02/20/2019] [Indexed: 02/06/2023] Open
Abstract
Background Central nervous system lymphomas (CNSL) is a devastating disease. Currently, a confirmatory biopsy is required prior to treatment. Objective Our investigation aims to prove the feasibility of a minimally-invasive diagnostic approach for the molecular characterization of CNSL. Methods Tissue biopsies from 6 patients with suspected CNSL were analyzed using a 649gene next-generation sequencing (NGS) tumor panel (tumor vs. reference tissue (EDTA-blood)). The individual somatic mutation pattern was used as a basis for the digital PCR analyzing circulating tumor DNA (ctDNA) from plasma and cerebrospinal fluid (CSF) samples, identifying one selected tumor mutation during this first step of the feasibility investigation. Results NGS-analysis of biopsy tissue revealed a specific somatic mutation pattern in all confirmed lymphoma samples (n = 5, NGS-sensitivity 100%) and none in the sample identified as normal brain tissue (NGS-specificity 100%). cfDNA-extraction was dependent on the extraction-kit used and feasible in 3 samples, in all of which somatic mutations were detectable (100%). Analysis of CSF-derived cfDNA was superior to plasma-derived cfDNA and routine microscopic analysis (lymphoma cells: n = 2, 40%). One patient showed a divergent molecular pattern, typical of Burkitt-Lymphoma (HIV+, serologic evidence of EBV-infection). Lumbar puncture was tolerated without complications, whereas biopsy caused 3 hemorrhages. Conclusions Our investigation provides evidence that analysis of cfDNA in central nervous system tumors is feasible using the described protocol. Molecular characterization of CNSL could be achieved by analysis of CSF-derived cfDNA. Knowledge of a tumor’s specific mutation pattern may allow initiation of targeted therapies, treatment surveillance and could lead to minimally-invasive diagnostics in the future. Electronic supplementary material The online version of this article (10.1186/s12885-019-5394-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne-Katrin Hickmann
- Department of Neurosurgery, Kantonsspital St. Gallen, Rorschacherstrasse 95, 9600, St. Gallen, Switzerland. .,Neurosurgical Department, Klinikum Stuttgart, Stuttgart, Germany.
| | - Maximilian Frick
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany
| | - Dirk Hadaschik
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany
| | - Florian Battke
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany
| | - Markus Bittl
- Neurosurgical Department, Klinikum Stuttgart, Stuttgart, Germany
| | - Oliver Ganslandt
- Neurosurgical Department, Klinikum Stuttgart, Stuttgart, Germany
| | - Saskia Biskup
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany.,Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Outpatient Clinic for Human Genetics, Tübingen, Germany
| | - Dennis Döcker
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany.,Outpatient Clinic for Human Genetics, Tübingen, Germany
| |
Collapse
|
34
|
FBXW7 in Cancer: What Has Been Unraveled Thus Far? Cancers (Basel) 2019; 11:cancers11020246. [PMID: 30791487 PMCID: PMC6406609 DOI: 10.3390/cancers11020246] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/14/2022] Open
Abstract
: The FBXW7 (F-box with 7 tandem WD40) protein encoded by the gene FBXW7 is one of the crucial components of ubiquitin ligase called Skp1-Cullin1-F-box (SCF) complex that aids in the degradation of many oncoproteins via the ubiquitin-proteasome system (UPS) thus regulating cellular growth. FBXW7 is considered as a potent tumor suppressor as most of its target substrates can function as potential growth promoters, including c-Myc, Notch, cyclin E, c-JUN, and KLF5. Its regulators include p53, C/EBP-δ, Numb, microRNAs, Pin 1, Hes-5, BMI1, Ebp2. Mounting evidence has indicated the involvement of aberrant expression of FBXW7 for tumorigenesis. Moreover, numerous studies have also shown its role in cancer cell chemosensitization, thereby demonstrating the importance of FBXW7 in the development of curative cancer therapy. This comprehensive review emphasizes on the targets, functions, regulators and expression of FBXW7 in different cancers and its involvement in sensitizing cancer cells to chemotherapeutic drugs.
Collapse
|
35
|
Puca L, Vlachostergios PJ, Beltran H. Neuroendocrine Differentiation in Prostate Cancer: Emerging Biology, Models, and Therapies. Cold Spring Harb Perspect Med 2019; 9:a030593. [PMID: 29844220 PMCID: PMC6360865 DOI: 10.1101/cshperspect.a030593] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although a de novo clinical presentation of small cell neuroendocrine carcinoma of the prostate is rare, a subset of patients previously diagnosed with prostate adenocarcinoma may develop neuroendocrine features in later stages of castration-resistant prostate cancer (CRPC) progression as a result of treatment resistance. Despite sharing clinical, histologic, and some molecular features with other neuroendocrine carcinomas, including small cell lung cancer, castration-resistant neuroendocrine prostate cancer (CRPC-NE) is clonally derived from prostate adenocarcinoma. CRPC-NE therefore retains early prostate cancer genomic alterations and acquires new molecular changes making them resistant to traditional CRPC therapies. This review focuses on recent advances in our understanding of CRPC-NE biology, the transdifferentiation/plasticity process, and development and characterization of relevant CRPC-NE preclinical models.
Collapse
Affiliation(s)
- Loredana Puca
- Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York 10021
- Division of Medical Oncology, Weill Cornell Medicine, New York, New York 10021
| | | | - Himisha Beltran
- Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York 10021
- Division of Medical Oncology, Weill Cornell Medicine, New York, New York 10021
| |
Collapse
|
36
|
Yeh CH, Bellon M, Nicot C. FBXW7: a critical tumor suppressor of human cancers. Mol Cancer 2018; 17:115. [PMID: 30086763 PMCID: PMC6081812 DOI: 10.1186/s12943-018-0857-2] [Citation(s) in RCA: 356] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) is involved in multiple aspects of cellular processes, such as cell cycle progression, cellular differentiation, and survival (Davis RJ et al., Cancer Cell 26:455-64, 2014; Skaar JR et al., Nat Rev Drug Discov 13:889-903, 2014; Nakayama KI and Nakayama K, Nat Rev Cancer 6:369-81, 2006). F-box and WD repeat domain containing 7 (FBXW7), also known as Sel10, hCDC4 or hAgo, is a member of the F-box protein family, which functions as the substrate recognition component of the SCF E3 ubiquitin ligase. FBXW7 is a critical tumor suppressor and one of the most commonly deregulated ubiquitin-proteasome system proteins in human cancer. FBXW7 controls proteasome-mediated degradation of oncoproteins such as cyclin E, c-Myc, Mcl-1, mTOR, Jun, Notch and AURKA. Consistent with the tumor suppressor role of FBXW7, it is located at chromosome 4q32, a genomic region deleted in more than 30% of all human cancers (Spruck CH et al., Cancer Res 62:4535-9, 2002). Genetic profiles of human cancers based on high-throughput sequencing have revealed that FBXW7 is frequently mutated in human cancers. In addition to genetic mutations, other mechanisms involving microRNA, long non-coding RNA, and specific oncogenic signaling pathways can inactivate FBXW7 functions in cancer cells. In the following sections, we will discuss the regulation of FBXW7, its role in oncogenesis, and the clinical implications and prognostic value of loss of function of FBXW7 in human cancers.
Collapse
Affiliation(s)
- Chien-Hung Yeh
- Department of Pathology and Laboratory Medicine, Center for Viral Pathogenesis, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Marcia Bellon
- Department of Pathology and Laboratory Medicine, Center for Viral Pathogenesis, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Christophe Nicot
- Department of Pathology and Laboratory Medicine, Center for Viral Pathogenesis, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
37
|
Chen JS, Yeh CN, Cheng CT, Yen CC, Chen YY, Huang SC, Chiang KC, Yeh TS, Chen SC, Chao TC, Yang MH, Chao Y. Role of PLK1 signaling pathway genes in gastrointestinal stromal tumors. Oncol Lett 2018; 16:3070-3082. [PMID: 30127898 PMCID: PMC6096274 DOI: 10.3892/ol.2018.9003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 05/14/2018] [Indexed: 02/06/2023] Open
Abstract
In previous studies by the authors, aurora kinase A (AURKA) was demonstrated as an independent poor prognostic marker for the recurrence of localized gastrointestinal stromal tumors (GISTs) and for the progression of advanced GISTs. In the present study, the prognostic effect of genes involved in cell cycle regulation in GISTs was further examined. Leading edge analysis in gene set enrichment analysis was used to identify the most common genes in the top 10 enriched gene sets of high-risk patients with GISTs in a Japanese study. The obtained gene list was uploaded to the Pathway Interaction Database to search for critical pathways. Selected genes within the pathway were subsequently verified through immunohistochemistry (IHC) in another cohort of patients. A total of 5 genes in 'PLK1 signaling events,' namely AURKA, polo-like kinase 1 (PLK1), cell division cycle 25C (CDC25C), budding uninhibited by benzimidazoles (BUB1), and targeting protein for Xklp2 (TPX2), were identified for subsequent study. Among the Japanese cohort, all 5 genes, except BUB1, were significant prognostic factors for poor recurrence-free survival (RFS). Among 141 patients enrolled for the IHC study, all 5 genes exhibited variable expression patterns. In the association study, only AURKA exhibited significant overexpression in non-gastric tumors. Although all 5 genes were considered as risk factors for poor RFS based on a univariate analysis, only the mitotic count and expression levels of CDC25C, BUB1, and TPX2 retained prognostic effects in the multivariate analysis. The PLK1 signaling pathway is crucial in the disease progression of GISTs. Genes within this pathway may serve as predictive markers for adjuvant therapy.
Collapse
Affiliation(s)
- Jen-Shi Chen
- Division of Hematology-Oncology, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C.,GIST Team, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Chun-Nan Yeh
- GIST Team, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C.,Department of Surgery, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Chi-Tung Cheng
- GIST Team, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C.,Department of Surgery, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C.,Institute of Biomedical Informatics, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - Chueh-Chuan Yen
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C.,Division of Medical Oncology, Center for Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan, R.O.C
| | - Yen-Yang Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan, R.O.C
| | - Shih-Chiang Huang
- GIST Team, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C.,Department of Pathology, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Kun-Chun Chiang
- Department of Surgery, Keelung Medical Center, Chang Gung Memorial Hospital and University, Keelung 204, Taiwan, R.O.C
| | - Ta-Sen Yeh
- GIST Team, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C.,Department of Surgery, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - San-Chi Chen
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C.,Division of Medical Oncology, Center for Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan, R.O.C
| | - Ta-Chung Chao
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C.,Division of Medical Oncology, Center for Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan, R.O.C
| | - Muh-Hwa Yang
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C.,Division of Medical Oncology, Center for Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan, R.O.C
| | - Yee Chao
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C.,Division of Medical Oncology, Center for Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan, R.O.C
| |
Collapse
|
38
|
Wasserman JK, AlGhamdi D, de Almeida JR, Stockley TL, Perez-Ordonez B. P53 Gene Mutation Identified by Next Generation Sequencing in Poorly Differentiated Neuroendocrine Carcinoma of the Nasal Cavity. Head Neck Pathol 2018; 13:516-522. [PMID: 29845478 PMCID: PMC6684699 DOI: 10.1007/s12105-018-0934-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/16/2018] [Indexed: 10/16/2022]
Abstract
Neuroendocrine carcinomas (NECs) are epithelial neoplasms showing morphologic, immunophenotypic or ultrastructural evidence of neuroendocrine differentiation. The 2017 WHO Classification of Head and Neck Tumours classifies NECs into well, moderately and poorly differentiated NECs according to light microscopic features, mitotic rate and presence of tumour necrosis. In this study, we performed next generation sequencing (NGS) using a targeted 161 cancer gene panel on a poorly differentiated NEC of the nasal cavity. The tumour was composed of large cells arranged in poorly formed glands and solid nests. The mitotic count rate was 30/10 HPFs and p53 protein was strongly expressed in all tumour cells. NGS identified a missense mutation, c.764T > G (p.Ile255Ser) in the TP53 gene with an allele frequency of 85%. This mutation results in an isoleucine to serine substitution and a non-functional protein. No other mutations were identified. These results suggest that TP53 mutations may drive oncogenesis in poorly differentiated NECs of the head and neck.
Collapse
Affiliation(s)
- Jason K. Wasserman
- 0000 0004 0474 0428grid.231844.8Laboratory Medicine Program, University Health Network, Toronto, ON Canada ,0000 0001 2157 2938grid.17063.33Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada ,0000 0004 0474 0428grid.231844.8Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| | - Doaa AlGhamdi
- 0000 0004 0474 0428grid.231844.8Laboratory Medicine Program, University Health Network, Toronto, ON Canada ,0000 0001 2157 2938grid.17063.33Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - John R. de Almeida
- 0000 0004 0474 0428grid.231844.8Department of Otolaryngology Head and Neck Surgery, Department of Surgical Oncology, University Health Network, Toronto, ON Canada
| | - Tracy L. Stockley
- 0000 0004 0474 0428grid.231844.8Laboratory Medicine Program, University Health Network, Toronto, ON Canada ,0000 0001 2157 2938grid.17063.33Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - Bayardo Perez-Ordonez
- 0000 0004 0474 0428grid.231844.8Laboratory Medicine Program, University Health Network, Toronto, ON Canada ,0000 0001 2157 2938grid.17063.33Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| |
Collapse
|
39
|
Chen R, Dong X, Gleave M. Molecular model for neuroendocrine prostate cancer progression. BJU Int 2018; 122:560-570. [DOI: 10.1111/bju.14207] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ruiqi Chen
- Department of Urologic Sciences; Vancouver Prostate Centre; University of British Columbia; Vancouver BC Canada
- Faculty of Medicine; University of Toronto; Toronto ON Canada
| | - Xuesen Dong
- Department of Urologic Sciences; Vancouver Prostate Centre; University of British Columbia; Vancouver BC Canada
| | - Martin Gleave
- Department of Urologic Sciences; Vancouver Prostate Centre; University of British Columbia; Vancouver BC Canada
| |
Collapse
|
40
|
Sárközy M, Kahán Z, Csont T. A myriad of roles of miR-25 in health and disease. Oncotarget 2018; 9:21580-21612. [PMID: 29765562 PMCID: PMC5940376 DOI: 10.18632/oncotarget.24662] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Small non-coding RNAs including microRNAs (miRNAs) have been recently recognized as important regulators of gene expression. MicroRNAs play myriads of roles in physiological processes as well as in the pathogenesis of a number of diseases by translational repression or mRNA destabilization of numerous target genes. The miR-106b-25 cluster is highly conserved in vertebrates and consists of three members including miR-106b, miR-93 and miR-25. MiR-106b and miR-93 share the same seed sequences; however, miR-25 has only a similar seed sequence resulting in different predicted target mRNAs. In this review, we specifically focus on the role of miR-25 in healthy and diseased conditions. Many of miR-25 target mRNAs are involved in biological processes such as cell proliferation, differentiation, and migration, apoptosis, oxidative stress, inflammation, calcium handling, etc. Therefore, it is no surprise that miR-25 has been reported as a key regulator of common cancerous and non-cancerous diseases. MiR-25 plays an important role in the pathogenesis of acute myocardial infarction, left ventricular hypertrophy, heart failure, diabetes mellitus, diabetic nephropathy, tubulointerstitial nephropathy, asthma bronchiale, cerebral ischemia/reperfusion injury, neurodegenerative diseases, schizophrenia, multiple sclerosis, etc. MiR-25 is also a well-described oncogenic miRNA playing a crucial role in the development of many tumor types including brain tumors, lung, breast, ovarian, prostate, thyroid, oesophageal, gastric, colorectal, hepatocellular cancers, etc. In this review, our aim is to discuss the translational therapeutic role of miR-25 in common diseased conditions based on relevant basic research and clinical studies.
Collapse
Affiliation(s)
- Márta Sárközy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Zsuzsanna Kahán
- Department of Oncotherapy, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Tamás Csont
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
41
|
Tang A, Gao K, Chu L, Zhang R, Yang J, Zheng J. Aurora kinases: novel therapy targets in cancers. Oncotarget 2017; 8:23937-23954. [PMID: 28147341 PMCID: PMC5410356 DOI: 10.18632/oncotarget.14893] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/17/2017] [Indexed: 02/06/2023] Open
Abstract
Aurora kinases, a family of serine/threonine kinases, consisting of Aurora A (AURKA), Aurora B (AURKB) and Aurora C (AURKC), are essential kinases for cell division via regulating mitosis especially the process of chromosomal segregation. Besides regulating mitosis, Aurora kinases have been implicated in regulating meiosis. The deletion of Aurora kinases could lead to failure of cell division and impair the embryonic development. Overexpression or gene amplification of Aurora kinases has been clarified in a number of cancers. And a growing number of studies have demonstrated that inhibition of Aurora kinases could potentiate the effect of chemotherapies. For the past decades, a series of Aurora kinases inhibitors (AKIs) developed effectively repress the progression and growth of many cancers both in vivo and in vitro, suggesting that Aurora kinases could be a novel therapeutic target. In this review, we'll first briefly present the structure, localization and physiological functions of Aurora kinases in mitosis, then describe the oncogenic role of Aurora kinases in tumorigenesis, we shall finally discuss the outcomes of AKIs combination with conventional therapy.
Collapse
Affiliation(s)
- Anqun Tang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Keyu Gao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Laili Chu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Rui Zhang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Jing Yang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China.,Department of Oncology, The First Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
42
|
Li W, Qian L, Lin J, Huang G, Hao N, Wei X, Wang W, Liang J. CD44 regulates prostate cancer proliferation, invasion and migration via PDK1 and PFKFB4. Oncotarget 2017; 8:65143-65151. [PMID: 29029419 PMCID: PMC5630319 DOI: 10.18632/oncotarget.17821] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 04/05/2017] [Indexed: 12/12/2022] Open
Abstract
Our recent studies have shown that CD44, a cell-surface protein with functions in many biologic processes, involved in glucose metabolism of prostate cancer cells. However, the molecular mechanisms of the regulation need to be further elucidated. In present study, LNCaP cells infected with lentivirus vector overexpressing CD44. The expression levels of key enzymes in glucose metabolism known as PDK1 and PFKFB4 were determined using QRT-PCR and western blot. PDK1 and PFKFB4 in LNCaP and PC3 cells were knocked down with shRNA respectively, and then cell proliferation, invasion and cell migration assay were performed. We found that overexpression of CD44 increased expression levels of PDK1 and PFKFB4 in LNCaP cells. Silencing of PDK1 and PFKFB4 could decrease cell proliferation, inhibit invasion and migration ability of prostate cancer cells. In addition, CD44 inhibitor could decrease glucose consumption and increase ROS levels of PC-3 cells significantly, as well as sensitize PC-3 cells to docetaxel. Taken together, CD44 could modulate aggressive phenotype of prostate cancer cells, by regulation of the expression of PDK1 and PFKFB4. CD44 may be a novel potential therapeutic target.
Collapse
Affiliation(s)
- Wei Li
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, NanNing, China
| | - Li Qian
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, NanNing, China
| | - Junhao Lin
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, NanNing, China
| | - Guihai Huang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, NanNing, China
| | - Nan Hao
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, NanNing, China
| | - Xiuwang Wei
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, NanNing, China
| | - Wei Wang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, NanNing, China
| | - Jianbo Liang
- Department of Urology, The People's Hospital of Guangxi Zhuang Autonomous Region, NanNing, China
| |
Collapse
|
43
|
Abstract
Metastatic castration-resistant prostate cancer (CRPC) is associated with substantial clinical, pathologic, and molecular heterogeneity. Most tumors remain driven by androgen receptor (AR) signaling, which has clinical implications for patient selection for AR-directed approaches. However, histologic and clinical resistance phenotypes can emerge after AR inhibition, in which the tumors become less dependent on the AR. In this review, we discuss prostate cancer variants including neuroendocrine (NEPC) and aggressive variant (AVPC) prostate cancers and their clinical implications. Improvements in the understanding of the biologic mechanisms and molecular features underlying prostate cancer variants may help prognostication and facilitate the development of novel therapeutic approaches for subclasses of patient with CRPC.
Collapse
Affiliation(s)
- Panagiotis J Vlachostergios
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, 413 East 69th Street 1412,, New York, NY, 10021, USA
| | - Loredana Puca
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, 413 East 69th Street 1412,, New York, NY, 10021, USA
| | - Himisha Beltran
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, 413 East 69th Street 1412,, New York, NY, 10021, USA.
| |
Collapse
|
44
|
Xu L, Liu H, Yan Z, Sun Z, Luo S, Lu Q. Inhibition of the Hedgehog signaling pathway suppresses cell proliferation by regulating the Gli2/miR-124/AURKA axis in human glioma cells. Int J Oncol 2017; 50:1868-1878. [PMID: 28393219 DOI: 10.3892/ijo.2017.3946] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/22/2017] [Indexed: 11/06/2022] Open
Abstract
Multiple lines of evidence indicate that aberrant activation of Hedgehog (Hh) signaling plays an important role in tumorigenesis in human glioma. However, the underlying molecular mechanism and crucial downstream targets of glioma-associated oncogene (Gli), a primary transcriptional regulator of Hh signaling, are not fully understood. Here, we report the identification of miR-124 as a novel downstream target of the transcriptional factor Gli2, which is important for proliferation and tumor growth in human glioma cells. Blockade of Hh signaling leads to a remarkable increase in miR-124 expression in glioma cells, whereas overexpression of Gli2 suppresses miR-124 expression by increasing the direct binding of Gli2 to the upstream region of the transcriptional start site for miR-124. Furthermore, we found that miR-124 potentially interacts with the 3'-UTR region of AURKA. Overexpression of miR-124 significantly decreased the expression of AURKA in glioma cells. In contrast, the loss of miR-124 led to the increased expression of AURKA mRNA and protein. In addition, cell proliferation and colony formation ability were significantly decreased following Gli2 knockdown in human glioma cells, while transfection with a miR-124 inhibitor rescued the proliferative ability of cells. These results demonstrate that miR-124 is an important downstream target gene of Hh signaling, and the Gli2/miR-124/AURKA axis is essential for the proliferation and growth of human glioma cells.
Collapse
Affiliation(s)
- Liyao Xu
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hua Liu
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhangming Yan
- MOE Key Laboratory of Bioinformatics, School of Life Science, Tsinghua University, Beijing 100084, P.R. China
| | - Zhirong Sun
- MOE Key Laboratory of Bioinformatics, School of Life Science, Tsinghua University, Beijing 100084, P.R. China
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Quqin Lu
- Department of Biostatistics and Epidemiology, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
45
|
Hoang DT, Iczkowski KA, Kilari D, See W, Nevalainen MT. Androgen receptor-dependent and -independent mechanisms driving prostate cancer progression: Opportunities for therapeutic targeting from multiple angles. Oncotarget 2017; 8:3724-3745. [PMID: 27741508 PMCID: PMC5356914 DOI: 10.18632/oncotarget.12554] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/29/2016] [Indexed: 12/25/2022] Open
Abstract
Despite aggressive treatment for localized cancer, prostate cancer (PC) remains a leading cause of cancer-related death for American men due to a subset of patients progressing to lethal and incurable metastatic castrate-resistant prostate cancer (CRPC). Organ-confined PC is treated by surgery or radiation with or without androgen deprivation therapy (ADT), while options for locally advanced and disseminated PC include radiation combined with ADT, or systemic treatments including chemotherapy. Progression to CRPC results from failure of ADT, which targets the androgen receptor (AR) signaling axis and inhibits AR-driven proliferation and survival pathways. The exact mechanisms underlying the transition from androgen-dependent PC to CRPC remain incompletely understood. Reactivation of AR has been shown to occur in CRPC despite depletion of circulating androgens by ADT. At the same time, the presence of AR-negative cell populations in CRPC has also been identified. While AR signaling has been proposed as the primary driver of CRPC, AR-independent signaling pathways may represent additional mechanisms underlying CRPC progression. Identification of new therapeutic strategies to target both AR-positive and AR-negative PC cell populations and, thereby, AR-driven as well as non-AR-driven PC cell growth and survival mechanisms would provide a two-pronged approach to eliminate CRPC cells with potential for synthetic lethality. In this review, we provide an overview of AR-dependent and AR-independent molecular mechanisms which drive CRPC, with special emphasis on the role of the Jak2-Stat5a/b signaling pathway in promoting castrate-resistant growth of PC through both AR-dependent and AR-independent mechanisms.
Collapse
Affiliation(s)
- David T Hoang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kenneth A Iczkowski
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Kilari
- Department of Medicine, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - William See
- Department of Urology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marja T Nevalainen
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pharmacology/Toxicology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
46
|
Wøyen AVT, Laczkó G, Høyer S, Hegyi L. The Missing Link in the Diagnostic Pathway of Prostate Cancer. Pathol Oncol Res 2017; 23:447-450. [PMID: 28054315 DOI: 10.1007/s12253-016-0183-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/29/2016] [Indexed: 11/28/2022]
Abstract
Prostate cancer is one of the most common cancers in the Western world. It is among the leading causes of cancer related death. While its incidence and survival increased significantly during the last few decades in Denmark, the mortality rate did not change for patients younger than 80 year old. Development of new techniques, such as multiparametric MRI, helps to increase the accuracy of diagnosis. However, a missing link in the diagnostic pathway may result in mistreatment if an acinar adenocarcinoma of prostate is transformed into a neuroendocrine phenotype such as small cell carcinoma.
Collapse
Affiliation(s)
| | - Gergely Laczkó
- Urologisk Afdeling, Regionshospitalet Viborg, Viborg, Denmark
| | - Søren Høyer
- Patologisk Institut, Aarhus Universitetshospital, Aarhus, Denmark
| | - Laszlo Hegyi
- Patologisk Institut, Regionshospitalet Viborg, Pakhusvej 1, 8800, Viborg, Denmark.
| |
Collapse
|
47
|
Xu J, Wu W, Wang J, Huang C, Wen W, Zhao F, Xu X, Pan X, Wang W, Zhu Q, Chen L. miR-367 promotes the proliferation and invasion of non-small cell lung cancer via targeting FBXW7. Oncol Rep 2016; 37:1052-1058. [PMID: 28000899 DOI: 10.3892/or.2016.5314] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/06/2016] [Indexed: 11/05/2022] Open
Abstract
The involvement of miR-367 in lung cancer development remains unclear. In the present study, we analyzed the expression of miR-367 in tumor and adjacent tissue samples from 113 patients with non-small cell lung cancer (NSCLC) utilizing real-time PCR. miR-367 expression was significantly upregulated in the cancer tissues compared with non-cancer controls. Based on the median value of the miR-367 expression level, we divided the NSCLC patients into miR-367 high-expression and miR-367 low-expression groups. Overexpression of miR-367 was correlated with a poorer prognosis of NSCLC patients Chi-square (χ2) test showed a significant statistical correlation between tumor size, tumor stage, metastasis and miR-367 expression. Additionally, miR-367 expression was found to be negatively correlated with FBXW7 expression. Based on the above correlations, we performed a series of functional experiments to further confirm the effect of miR-367 on NSCLC. Our results indicated that miR-367 may be involved in the development and progression of NSCLC by promoting proliferation and invasion and impeding apoptosis in NSCLC cells. Furthermore, FBXW7 was identified as a potential target of miR-367, and FBXW7 silencing partially compromised the invasive, proliferative and migratory capacities in the cells with low miR-367 expression. Thus, the miR-367/FBXW7 axis may be involved in the development and progression of NSCLC and may be valuable as a therapeutic target for the treatment of human NSCLC, especially cancers with high invasive potential.
Collapse
Affiliation(s)
- Jing Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Weibing Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jun Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chenjun Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Fei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xinfeng Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xianglong Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Quan Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
48
|
Xu X, Huang YH, Li YJ, Cohen A, Li Z, Squires J, Zhang W, Chen XF, Zhang M, Huang JT. Potential therapeutic effect of epigenetic therapy on treatment-induced neuroendocrine prostate cancer. Asian J Androl 2016; 19:686-693. [PMID: 27905327 PMCID: PMC5676429 DOI: 10.4103/1008-682x.191518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although adenocarcinomas of the prostate are relatively indolent, some patients with advanced adenocarcinomas show recurrence of treatment-induced neuroendocrine prostate cancer, which is highly aggressive and lethal. Detailed biological features of treatment-induced neuroendocrine prostate cancer have not been characterized owing to limited biopsies/resections and the lack of a cellular model. In this study, we used a unique cellular model (LNCaP/NE1.8) to investigate the potential role of cancer stem cells in treatment-induced neuroendocrine prostate cancer with acquired resistance to hormonal therapy and chemotherapy. We also studied the role of cancer stem cells in enhancing invasion in treatment-induced neuroendocrine prostate cancer cells that recurred after long-term androgen-ablation treatment. Using an in vitro system mimicking clinical androgen-ablation, our results showed that the neuroendocrine-like subclone NE1.8 cells were enriched with cancer stem cells. Compared to parental prostate adenocarcinoma LNCaP cells, NE1.8 cells are more resistant to androgen deprivation therapy and chemotherapeutic agents and show increased cancer cell invasiveness. Results from this study also suggest a potential epigenetic therapeutic strategy using suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, as a chemotherapeutic agent for therapy-resistant treatment-induced neuroendocrine prostate cancer cells to minimize the risk of prostate cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Xiang Xu
- School of Life Sciences, Anhui University, Hefei, China.,Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Yu-Hua Huang
- Department of Urology, The First Affiliated Hospital of SooChow University, Suzhou, China
| | - Yan-Jing Li
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Alexa Cohen
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Zhen Li
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Jill Squires
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Wei Zhang
- Department of Pathology, Beijing Hospital, Beijing, China
| | - Xu-Feng Chen
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Min Zhang
- School of Life Sciences, Anhui University, Hefei, China
| | - Jiao-Ti Huang
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, California, USA
| |
Collapse
|
49
|
Sasai K, Treekitkarnmongkol W, Kai K, Katayama H, Sen S. Functional Significance of Aurora Kinases-p53 Protein Family Interactions in Cancer. Front Oncol 2016; 6:247. [PMID: 27933271 PMCID: PMC5122578 DOI: 10.3389/fonc.2016.00247] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022] Open
Abstract
Aurora kinases play critical roles in regulating spindle assembly, chromosome segregation, and cytokinesis to ensure faithful segregation of chromosomes during mitotic cell division cycle. Molecular and cell biological studies have revealed that Aurora kinases, at physiological levels, orchestrate complex sequential cellular processes at distinct subcellular locations through functional interactions with its various substrates. Aberrant expression of Aurora kinases, on the other hand, cause defects in mitotic spindle assembly, checkpoint response activation, and chromosome segregation leading to chromosomal instability. Elevated expression of Aurora kinases correlating with chromosomal instability is frequently detected in human cancers. Recent genomic profiling of about 3000 human cancer tissue specimens to identify various oncogenic signatures in The Cancer Genome Atlas project has reported that recurrent amplification and overexpression of Aurora kinase-A characterize distinct subsets of human tumors across multiple cancer types. Besides the well-characterized canonical pathway interactions of Aurora kinases in regulating assembly of the mitotic apparatus and chromosome segregation, growing evidence also supports the notion that deregulated expression of Aurora kinases in non-canonical pathways drive transformation and genomic instability by antagonizing tumor suppressor and exacerbating oncogenic signaling through direct interactions with critical proteins. Aberrant expression of the Aurora kinases–p53 protein family signaling axes appears to be critical in the abrogation of p53 protein family mediated tumor suppressor pathways frequently deregulated during oncogenic transformation process. Recent findings reveal the existence of feedback regulatory loops in mRNA expression and protein stability of these protein families and their consequences on downstream effectors involved in diverse physiological functions, such as mitotic progression, checkpoint response pathways, as well as self-renewal and pluripotency in embryonic stem cells. While these investigations have focused on the functional consequences of Aurora kinase protein family interactions with wild-type p53 family proteins, those involving Aurora kinases and mutant p53 remain to be elucidated. This article presents a comprehensive review of studies on Aurora kinases–p53 protein family interactions along with a prospective view on the possible functional consequences of Aurora kinase–mutant p53 signaling pathways in tumor cells. Additionally, we also discuss therapeutic implications of these findings in Aurora kinases overexpressing subsets of human tumors.
Collapse
Affiliation(s)
- Kaori Sasai
- Department of Molecular Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Warapen Treekitkarnmongkol
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Kazuharu Kai
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Hiroshi Katayama
- Department of Molecular Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Subrata Sen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| |
Collapse
|
50
|
Guo Y, Ma J, Zheng Y, Li L, Gui X, Wang Q, Meng X, Shang H. HPV16 E6 upregulates Aurora A expression. Oncol Lett 2016; 12:1387-1393. [PMID: 27446442 PMCID: PMC4950527 DOI: 10.3892/ol.2016.4786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/13/2016] [Indexed: 12/26/2022] Open
Abstract
Overexpression of Aurora A kinase occurs in certain types of cancer, and therefore results in chromosome instability and phosphorylation-mediated ubiquitylation and degradation of p53 for tumorigenesis. The high-risk subtype human papillomavirus (HPV)16 early oncoprotein E6 is a major contributor inducing host cell immortalization and transformation through interaction with a number of cellular factors. In the present study, co-immunoprecipitation, glutathione S-transferase pull-down and immunostaining were used to show that HPV16 E6 and Aurora A bind to each other in vivo and in vitro. Western blotting and reverse transcription-polymerase chain reaction were used to reveal that HPV16 E6 inhibited cell apoptosis by stabilizing Aurora A expression. The present study may report a new mechanism for the involvement of HPV16 E6 in carcinogenesis, as HPV16 E6 elevates Aurora A expression and the latter may be a common target for oncogenic viruses that result in cell carcinogenesis.
Collapse
Affiliation(s)
- Yi Guo
- Department of Gynecology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jiaming Ma
- Department of Pain Management, Shengjing Hospital, Shenyang, Liaoning 110022, P.R. China
| | - Yahong Zheng
- Department of Gynecology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lu Li
- Department of Gynecology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaowei Gui
- Department of Gynecology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Qian Wang
- Key Laboratory of AIDS Immunology of Ministry of Health, Department of Laboratory Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiangkai Meng
- Department of Gynecology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hong Shang
- Key Laboratory of AIDS Immunology of Ministry of Health, Department of Laboratory Medicine, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|