1
|
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
|
2
|
Chen S, Leng P, Guo J, Zhou H. FBXW7 in breast cancer: mechanism of action and therapeutic potential. J Exp Clin Cancer Res 2023; 42:226. [PMID: 37658431 PMCID: PMC10474666 DOI: 10.1186/s13046-023-02767-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/18/2023] [Indexed: 09/03/2023] Open
Abstract
Breast cancer is one of the frequent tumors that seriously endanger the physical and mental well-being in women. F-box and WD repeat domain-containing 7 (FBXW7) is a neoplastic repressor. Serving as a substrate recognition element for ubiquitin ligase, FBXW7 participates in the ubiquitin-proteasome system and is typically in charge of the ubiquitination and destruction of crucial oncogenic proteins, further performing a paramount role in cell differentiation, apoptosis and metabolic processes. Low levels of FBXW7 cause abnormal stability of pertinent substrates, mutations and/or deletions in the FBXW7 gene have been reported to correlate with breast cancer malignant progression and chemoresistance. Given the lack of an effective solution to breast cancer's clinical drug resistance dilemma, elucidating FBXW7's mechanism of action could provide a theoretical basis for targeted drug exploration. Therefore, in this review, we focused on FBXW7's role in a range of breast cancer malignant behaviors and summarized the pertinent cellular targets, signaling pathways, as well as the mechanisms regulating FBXW7 expression. We also proposed novel perspectives for the exploitation of alternative therapies and specific tumor markers for breast cancer by therapeutic strategies aiming at FBXW7.
Collapse
Affiliation(s)
- Siyu Chen
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosisand, Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology , Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Leng
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosisand, Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology , Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlin Guo
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosisand, Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology , Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Hao Zhou
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosisand, Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology , Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
3
|
Kim DE, Byeon HE, Kim DH, Kim SG, Yim H. Plk2-mediated phosphorylation and translocalization of Nrf2 activates anti-inflammation through p53/Plk2/p21 cip1 signaling in acute kidney injury. Cell Biol Toxicol 2023; 39:1509-1529. [PMID: 35842499 PMCID: PMC10425522 DOI: 10.1007/s10565-022-09741-1] [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] [Received: 04/01/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022]
Abstract
The Plk2 is a cellular stress-responsive factor that is induced in response to oxidative stress. However, the roles of Plk2 in acute kidney injury (AKI) have not been clarified. We previously found that Plk2 is an interacting factor of Nrf2 in response to cellular stress, since Plk2 is upregulated in the Nrf2-dependent network. Here, we show that the levels of p53, Plk2, p21cip1, and chromatin-bound Nrf2 were all upregulated in kidney tissues of mice or NRK52E cells treated with either cisplatin or methotrexate. Upregulation of Plk2 by p53 led to an increase of Nrf2 in both soluble and chromatin fractions in cisplatin-treated NRK52E cells. Consistently, depletion of Plk2 suppressed the levels of Nrf2. Of note, Plk2 directly phosphorylated Nrf2 at Ser40, which facilitated its interaction with p21cip1 and translocation into the nuclei for the activation of anti-oxidative and anti-inflammatory factors in response to AKI. Together, these findings suggest that Plk2 may serve as an anti-oxidative and anti-inflammatory regulator through the phosphorylation and activation of Nrf2 to protect kidney cells from kidney toxicants and that Plk2 and Nrf2 therefore work cooperatively for the protection and survival of kidney cells from harmful stresses.
Collapse
Affiliation(s)
- Da-Eun Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Gyeonggi-do, Korea
| | - Hye Eun Byeon
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Gyeonggi-do, Korea
| | - Dae-Hoon Kim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Gyeonggi-do, Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, 10326, Gyeonggi-Do, Korea.
- College of Pharmacy, Seoul National University, Gwanakro 599, Seoul, 08826, Korea.
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Gyeonggi-do, Korea.
| |
Collapse
|
4
|
Di Fiore R, Suleiman S, Drago-Ferrante R, Subbannayya Y, Suleiman S, Vasileva-Slaveva M, Yordanov A, Pentimalli F, Giordano A, Calleja-Agius J. The Role of FBXW7 in Gynecologic Malignancies. Cells 2023; 12:1415. [PMID: 37408248 DOI: 10.3390/cells12101415] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
The F-Box and WD Repeat Domain Containing 7 (FBXW7) protein has been shown to regulate cellular growth and act as a tumor suppressor. This protein, also known as FBW7, hCDC4, SEL10 or hAGO, is encoded by the gene FBXW7. It is a crucial component of the Skp1-Cullin1-F-box (SCF) complex, which is a ubiquitin ligase. This complex aids in the degradation of many oncoproteins, such as cyclin E, c-JUN, c-MYC, NOTCH, and MCL1, via the ubiquitin-proteasome system (UPS). The FBXW7 gene is commonly mutated or deleted in numerous types of cancer, including gynecologic cancers (GCs). Such FBXW7 mutations are linked to a poor prognosis due to increased treatment resistance. Hence, detection of the FBXW7 mutation may possibly be an appropriate diagnostic and prognostic biomarker that plays a central role in determining suitable individualized management. Recent studies also suggest that, under specific circumstances, FBXW7 may act as an oncogene. There is mounting evidence indicating that the aberrant expression of FBXW7 is involved in the development of GCs. The aim of this review is to give an update on the role of FBXW7 as a potential biomarker and also as a therapeutic target for novel treatments, particularly in the management of GCs.
Collapse
Affiliation(s)
- Riccardo Di Fiore
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Sherif Suleiman
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | | | - Yashwanth Subbannayya
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Sarah Suleiman
- Whipps Cross Hospital, Barts Health NHS Trust, Leytonstone, London E11 1NR, UK
| | - Mariela Vasileva-Slaveva
- Department of Breast Surgery, "Dr. Shterev" Hospital, 1330 Sofia, Bulgaria
- Research Institute, Medical University Pleven, 5800 Pleven, Bulgaria
- Bulgarian Breast and Gynecological Cancer Association, 1784 Sofia, Bulgaria
| | - Angel Yordanov
- Department of Gynecological Oncology, Medical University Pleven, 5800 Pleven, Bulgaria
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University "Giuseppe DeGennaro", 70010 Casamassima, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Jean Calleja-Agius
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| |
Collapse
|
5
|
Jiménez-Izquierdo R, Morrugares R, Suanes-Cobos L, Correa-Sáez A, Garrido-Rodríguez M, Cerero-Tejero L, Khan OM, de la Luna S, Sancho R, Calzado MA. FBXW7 tumor suppressor regulation by dualspecificity tyrosine-regulated kinase 2. Cell Death Dis 2023; 14:202. [PMID: 36934104 PMCID: PMC10024693 DOI: 10.1038/s41419-023-05724-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/20/2023]
Abstract
FBXW7 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 main tumor suppressor due to its ability to control proteasome-mediated degradation of several oncoproteins such as c-Jun, c-Myc, Cyclin E1, mTOR, and Notch1-IC. FBXW7 inactivation in human cancers results from a somatic mutation or downregulation of its protein levels. This work describes a novel regulatory mechanism for FBXW7 dependent on the serine/threonine protein kinase DYRK2. We show that DYRK2 interacts with and phosphorylates FBXW7 resulting in its proteasome-mediated degradation. DYRK2-dependent FBXW7 destabilization is independent of its ubiquitin ligase activity. The functional analysis demonstrates the existence of DYRK2-dependent regulatory mechanisms for key FBXW7 substrates. Finally, we provide evidence indicating that DYRK2-dependent regulation of FBXW7 protein accumulation contributes to cytotoxic effects in response to chemotherapy agents such as Doxorubicin or Paclitaxel in colorectal cancer cell lines and to BET inhibitors in T-cell acute lymphoblastic leukemia cell lines. Altogether, this work reveals a new regulatory axis, DYRK2/FBXW7, which provides an understanding of the role of these two proteins in tumor progression and DNA damage responses.
Collapse
Affiliation(s)
- Rafael Jiménez-Izquierdo
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Rosario Morrugares
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Lucía Suanes-Cobos
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Alejandro Correa-Sáez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Martín Garrido-Rodríguez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Laura Cerero-Tejero
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Omar M Khan
- Hamad Bin Khalifa University, College of Health and Life Sciences Qatar Foundation, Education City, Doha, Qatar
| | - Susana de la Luna
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08003, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Rocío Sancho
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, SE10 9RT, UK
- Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Marco A Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain.
- Hospital Universitario Reina Sofía, Córdoba, Spain.
| |
Collapse
|
6
|
Weier AK, Homrich M, Ebbinghaus S, Juda P, Miková E, Hauschild R, Zhang L, Quast T, Mass E, Schlitzer A, Kolanus W, Burgdorf S, Gruß OJ, Hons M, Wieser S, Kiermaier E. Multiple centrosomes enhance migration and immune cell effector functions of mature dendritic cells. J Cell Biol 2022; 221:e202107134. [PMID: 36214847 PMCID: PMC9555069 DOI: 10.1083/jcb.202107134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 03/01/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
Centrosomes play a crucial role during immune cell interactions and initiation of the immune response. In proliferating cells, centrosome numbers are tightly controlled and generally limited to one in G1 and two prior to mitosis. Defects in regulating centrosome numbers have been associated with cell transformation and tumorigenesis. Here, we report the emergence of extra centrosomes in leukocytes during immune activation. Upon antigen encounter, dendritic cells pass through incomplete mitosis and arrest in the subsequent G1 phase leading to tetraploid cells with accumulated centrosomes. In addition, cell stimulation increases expression of polo-like kinase 2, resulting in diploid cells with two centrosomes in G1-arrested cells. During cell migration, centrosomes tightly cluster and act as functional microtubule-organizing centers allowing for increased persistent locomotion along gradients of chemotactic cues. Moreover, dendritic cells with extra centrosomes display enhanced secretion of inflammatory cytokines and optimized T cell responses. Together, these results demonstrate a previously unappreciated role of extra centrosomes for regular cell and tissue homeostasis.
Collapse
Affiliation(s)
- Ann-Kathrin Weier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Mirka Homrich
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Stephanie Ebbinghaus
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Pavel Juda
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Eliška Miková
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Robert Hauschild
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Lili Zhang
- Life and Medical Sciences Institute, Quantitative Systems Biology, University of Bonn, Bonn, Germany
| | - Thomas Quast
- Life and Medical Sciences Institute, Molecular Immunology and Cell Biology, University of Bonn, Bonn, Germany
| | - Elvira Mass
- Life and Medical Sciences Institute, Developmental Biology of the Immune System, University of Bonn, Bonn, Germany
| | - Andreas Schlitzer
- Life and Medical Sciences Institute, Quantitative Systems Biology, University of Bonn, Bonn, Germany
| | - Waldemar Kolanus
- Life and Medical Sciences Institute, Molecular Immunology and Cell Biology, University of Bonn, Bonn, Germany
| | - Sven Burgdorf
- Life and Medical Sciences Institute, Cellular Immunology, University of Bonn, Bonn, Germany
| | - Oliver J. Gruß
- Institute of Genetics, University of Bonn, Bonn, Germany
| | - Miroslav Hons
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Stefan Wieser
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Castelldefels, Spain
| | - Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
7
|
Hänle-Kreidler S, Richter KT, Hoffmann I. The SCF-FBXW7 E3 ubiquitin ligase triggers degradation of histone 3 lysine 4 methyltransferase complex component WDR5 to prevent mitotic slippage. J Biol Chem 2022; 298:102703. [PMID: 36395886 PMCID: PMC9764181 DOI: 10.1016/j.jbc.2022.102703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
During prolonged mitotic arrest induced by antimicrotubule drugs, cell fate decision is determined by two alternative pathways, one leading to cell death and the other inducing premature escape from mitosis by mitotic slippage. FBWX7, a member of the F-box family of proteins and substrate-targeting subunit of the SKP1-CUL1-F-Box E3 ubiquitin ligase complex, promotes mitotic cell death and prevents mitotic slippage, but molecular details underlying these roles for FBWX7 are unclear. In this study, we report that WDR5 (WD-repeat containing protein 5), a component of the mixed lineage leukemia complex of histone 3 lysine 4 methyltransferases, is a substrate of FBXW7. We determined by coimmunoprecipitation experiments and in vitro binding assays that WDR5 interacts with FBXW7 in vivo and in vitro. SKP1-CUL1-F-Box-FBXW7 mediates ubiquitination of WDR5 and targets it for proteasomal degradation. Furthermore, we find that WDR5 depletion counteracts FBXW7 loss of function by reducing mitotic slippage and polyploidization. In conclusion, our data elucidate a new mechanism in mitotic cell fate regulation, which might contribute to prevent chemotherapy resistance in patients after antimicrotubule drug treatment.
Collapse
Affiliation(s)
- Simon Hänle-Kreidler
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, Heidelberg, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Kai T. Richter
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, Heidelberg, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Ingrid Hoffmann
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, Heidelberg, Germany,For correspondence: Ingrid Hoffmann
| |
Collapse
|
8
|
Zhang C, Ni C, Lu H. Polo-Like Kinase 2: From Principle to Practice. Front Oncol 2022; 12:956225. [PMID: 35898867 PMCID: PMC9309260 DOI: 10.3389/fonc.2022.956225] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 11/21/2022] Open
Abstract
Polo-like kinase (PLK) 2 is an evolutionarily conserved serine/threonine kinase that shares the n-terminal kinase catalytic domain and the C-terminal Polo Box Domain (PBD) with other members of the PLKs family. In the last two decades, mounting studies have focused on this and tried to clarify its role in many aspects. PLK2 is essential for mitotic centriole replication and meiotic chromatin pairing, synapsis, and crossing-over in the cell cycle; Loss of PLK2 function results in cell cycle disorders and developmental retardation. PLK2 is also involved in regulating cell differentiation and maintaining neural homeostasis. In the process of various stimuli-induced stress, including oxidative and endoplasmic reticulum, PLK2 may promote survival or apoptosis depending on the intensity of stimulation and the degree of cell damage. However, the role of PLK2 in immunity to viral infection has been studied far less than that of other family members. Because PLK2 is extensively and deeply involved in normal physiological functions and pathophysiological mechanisms of cells, its role in diseases is increasingly being paid attention to. The effect of PLK2 in inhibiting hematological tumors and fibrotic diseases, as well as participating in neurodegenerative diseases, has been gradually recognized. However, the research results in solid organ tumors show contradictory results. In addition, preliminary studies using PLK2 as a disease predictor and therapeutic target have yielded some exciting and promising results. More research will help people better understand PLK2 from principle to practice.
Collapse
Affiliation(s)
- Chuanyong Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Chuangye Ni
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Hao Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
- *Correspondence: Hao Lu,
| |
Collapse
|
9
|
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: 57] [Impact Index Per Article: 28.5] [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
|
10
|
Shu L, Chen A, Li L, Yao L, He Y, Xu J, Gu W, Li Q, Wang K, Zhang T, Liu G. NRG1 regulates Fra-1 transcription and metastasis of triple-negative breast cancer cells via the c-Myc ubiquitination as manipulated by ERK1/2-mediated Fbxw7 phosphorylation. Oncogene 2022; 41:907-919. [PMID: 34992218 DOI: 10.1038/s41388-021-02142-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 11/14/2021] [Accepted: 11/29/2021] [Indexed: 11/09/2022]
Abstract
Neuregulin 1 (NRG1), an EGF family member, is expressed in most breast cancers. It promotes breast cancer growth and metastasis in HER2 receptor expressing breast cancer. However, its role in triple-negative breast cancer (TNBC) has not been extensively investigated. In this study, we observed that NRG1 knockdown resulted in the suppression of TNBC cells (MDA-MB-231 cell and MDA-MB-468 cell) metastasis and downregulation of Fra-1 (FOS-like 1, AP-1 transcription factor subunit, which is an overexpressed transcription factor in TNBC and acts as a coordinator of metastasis). In addition, the transcriptional regulation of Fra-1 by NRG1 was mediated by ERK1/2-induced recruitment of c-Myc (MYC proto-oncogene, transcription factor) to the promoter of Fra-1. Furthermore, c-Myc was targeted by an E3 ligase Fbxw7 and its ubiquitination and degradation by Fbxw7 was regulated by NRG1 expression and ERK1/2-mediated Fbxw7 phosphorylation that results in the dissociation and nuclear import of c-Myc. Taken together, the results of our study demonstrated that NRG1 regulates the Fra-1 expression to coordinate the TNBC metastasis via the novel ERK1/2-Fbxw7-c-Myc pathway and targeting NRG1 expression could be a potential therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Le Shu
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui Province, People's Republic of China.
| | - Ao Chen
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Linrui Li
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Lun Yao
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Yiduo He
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Jianbo Xu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Wei Gu
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui Province, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, Anhui Province, People's Republic of China
| | - Qiang Li
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui Province, People's Republic of China
- Department of Cell Biology, School of Biological Sciences, Bengbu Medical College, Bengbu, 233030, Anhui Province, People's Republic of China
| | - Kun Wang
- College of Biomedical Engineering, Hubei University of Medicine, Shiyan, 442000, Hubei Province, People's Republic of China
| | - Tongcun Zhang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, 430070, Hubei Province, People's Republic of China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Guoquan Liu
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui Province, People's Republic of China.
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, Anhui Province, People's Republic of China.
| |
Collapse
|
11
|
Korns J, Liu X, Takiar V. A review of Plks: Thinking outside the (polo) box. Mol Carcinog 2022; 61:254-263. [PMID: 35049091 DOI: 10.1002/mc.23388] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/19/2022]
Abstract
The polo-like kinase (Plk) family is comprised of five different members (Plk1-5), each with their own distinct functions. Plk family members participate in pivotal cell division processes as well as in non-mitotic roles. Importantly, Plk expression has been correlated with various disease states, including cancer. Multiples therapies, which primarily target Plk1, are currently being investigated alone or in combination with other agents for clinical use in different cancers. As the role of Plks in disease progression becomes more prominent, it is important to outline their functions as cell cycle regulators and more. This review summarizes the structure and both mitotic and non-mitotic functions of each of the five Plk family members, sequentially. Additionally, the proposed mechanisms for how Plks contribute to tumorigenesis and the therapeutics currently under investigation are outlined.
Collapse
Affiliation(s)
- Julianna Korns
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnat, Ohio, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Vinita Takiar
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnat, Ohio, USA.,Cincinnati VA Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
12
|
Wang CH, Lu TJ, Wang LK, Wu CC, Chen ML, Kuo CY, Shyu RY, Tsai FM. Tazarotene-induced gene 1 interacts with Polo-like kinase 2 and inhibits cell proliferation in HCT116 colorectal cancer cells. Cell Biol Int 2021; 45:2347-2356. [PMID: 34314079 DOI: 10.1002/cbin.11681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/20/2021] [Accepted: 07/24/2021] [Indexed: 01/05/2023]
Abstract
Tazarotene-induced gene 1 (TIG1) is considered to be a tumor suppressor gene that is highly expressed in normal or well-differentiated colon tissues, while downregulation of TIG1 expression occurs in poorly differentiated colorectal cancer (CRC) tissues. However, it is still unclear how TIG1 regulates the tumorigenesis of CRC. Polo-like kinases (Plks) are believed to play an important role in regulating the cell cycle. The performance of PLK2 in CRC is negatively correlated with the differentiation status of CRC tissues. Here, we found that PLK2 can induce the growth of CRC cells and that TIG1 can prevent PLK2 from promoting the proliferation of CRC cells. We also found that the expression of PLK2 in CRC cells was associated with low levels of Fbxw7 protein and increased expression of cyclin E1. When TIG1 was coexpressed with PLK2, the changes in Fbxw7/cyclin E1 levels induced by PLK2 were reversed. In contrast, silencing TIG1 promoted the proliferation of CRC, and when PLK2 was also silenced, the proliferation of CRC cells induced by TIG1 silencing was significantly inhibited. The above research results suggest that TIG1 can regulate the tumorigenesis of CRC by regulating the activity of PLK2.
Collapse
Affiliation(s)
- Chun-Hua Wang
- Department of Dermatology, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tzung-Ju Lu
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Lu-Kai Wang
- Radiation Biology Core Laboratory, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chang-Chieh Wu
- Department of Surgery, Tri-Service General Hospital Keelung Branch, National Defense Medical Center, Keelung, Taiwan
| | - Mao-Liang Chen
- Department of Research, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Rong-Yaun Shyu
- Department of Internal Medicine, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| |
Collapse
|
13
|
Yang Y, Zhou X, Liu X, Song R, Gao Y, Wang S. Implications of FBXW7 in Neurodevelopment and Neurodegeneration: Molecular Mechanisms and Therapeutic Potential. Front Cell Neurosci 2021; 15:736008. [PMID: 34512273 PMCID: PMC8424092 DOI: 10.3389/fncel.2021.736008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/04/2021] [Indexed: 11/25/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) mediated protein degradation is crucial to maintain quantitive and functional homeostasis of diverse proteins. Balanced cellular protein homeostasis controlled by UPS is fundamental to normal neurological functions while impairment of UPS can also lead to some neurodevelopmental and neurodegenerative disorders. Functioning as the substrate recognition component of the SCF-type E3 ubiquitin ligase, FBXW7 is essential to multiple aspects of cellular processes via targeting a wide range of substrates for proteasome-mediated degradation. Accumulated evidence shows that FBXW7 is fundamental to neurological functions and especially implicated in neurodevelopment and the nosogenesis of neurodegeneration. In this review, we describe general features of FBXW7 gene and proteins, and mainly present recent findings that highlight the vital roles and molecular mechanisms of FBXW7 in neurodevelopment such as neurogenesis, myelination and cerebral vasculogenesis and in the pathogenesis of some typical neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease and Huntington’s disease. Additionally, we also provide a prospect on focusing FBXW7 as a potential therapeutic target to rescue neurodevelopmental and neurodegenerative impairment.
Collapse
Affiliation(s)
- Yu Yang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Xuan Zhou
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China.,Research Center for Quality of Life and Applied Psychology, School of Humanities and Management, Guangdong Medical University, Dongguan, China
| | - Xinpeng Liu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Ruying Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Yiming Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Shuai Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| |
Collapse
|
14
|
Roopasree OJ, Adivitiya, Chakraborty S, Kateriya S, Veleri S. Centriole is the pivot coordinating dynamic signaling for cell proliferation and organization during early development in the vertebrates. Cell Biol Int 2021; 45:2178-2197. [PMID: 34288241 DOI: 10.1002/cbin.11667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/12/2021] [Indexed: 11/07/2022]
Abstract
Vertebrates have an elaborate and functionally segmented body. It evolves from a single cell by systematic cell proliferation but attains a complex body structure with exquisite precision. This development requires two cellular events: cell cycle and ciliogenesis. For these events, the dynamic molecular signaling is converged at the centriole. The cell cycle helps in cell proliferation and growth of the body and is a highly regulated and integrated process. Its errors cause malignancies and developmental disorders. The cells newly proliferated are organized during organogenesis. For a cellular organization, dedicated signaling hubs are developed in the cells, and most often cilia are utilized. The cilium is generated from one of the centrioles involved in cell proliferation. The developmental signaling pathways hosted in cilia are essential for the elaboration of the body plan. The cilium's compartmental seclusion is ideal for noise-free molecular signaling and is essential for the precision of the body layout. The dysfunctional centrioles and primary cilia distort the development of body layout that manifest as serious developmental disorders. Thus, centriole has a dual role in the growth and cellular organization. It organizes dynamically expressed molecules of cell cycle and ciliogenesis and plays a balancing act to generate new cells and organize them during development. A putative master molecule may regulate and coordinate the dynamic gene expression at the centrioles. The convergence of many critical signaling components at the centriole reiterates the idea that centriole is a major molecular workstation involved in elaborating the structural design and complexity in vertebrates. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- O J Roopasree
- Agroprocessing Technology Division, CSIR-National Institute of Interdisciplinary Science and Technology, Thiruvananthapuram, Kerala 695019 and Academy of CSIR, Uttar Pradesh - 201002, India
| | - Adivitiya
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Soura Chakraborty
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Suneel Kateriya
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Shobi Veleri
- Drug Safety Division, ICMR-National Institute of Nutrition, Hyderabad, 500007, India
| |
Collapse
|
15
|
Badarudeen B, Anand U, Mukhopadhyay S, Manna TK. Ubiquitin signaling in the control of centriole duplication. FEBS J 2021; 289:4830-4849. [PMID: 34115927 DOI: 10.1111/febs.16069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/22/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022]
Abstract
The centrosome plays an essential role in maintaining genetic stability, ciliogenesis and cell polarisation. The core of the centrosome is made up of two centrioles that duplicate precisely once during every cell cycle to generate two centrosomes that are required for bipolar spindle assembly and chromosome segregation. Abundance of centriole proteins at optimal levels and their recruitment to the centrosome are tightly regulated in time and space in order to restrict aberrant duplication of centrioles, a phenomenon that is observed in many cancers. Recent advances have conclusively shown that dedicated ubiquitin ligase-dependent protein degradation machineries are involved in governing centriole duplication. These studies revealed intricate mechanistic insights into how the ubiquitin ligases target different centriole proteins. In certain cases, a specific ubiquitin ligase targets a number of substrate proteins that co-regulate centriole assembly, prompting the possibility that substrate-targeting occurs during formation of the sub-centriolar structures. There are also instances where a specific centriole duplication protein is targeted by several ubiquitin ligases at different stages of the cell cycle, suggesting synchronised actions. Recent evidence also indicated a direct association of E3 ubiquitin ligase with the centrioles, supporting the notion that substrate-targeting occurs in the organelle itself. In this review, we highlight these advances by underlining the mechanisms of how different ubiquitin ligase machineries control centriole duplication and discuss our views on their coordination.
Collapse
Affiliation(s)
- Binshad Badarudeen
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, India
| | - Ushma Anand
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, India
| | - Swarnendu Mukhopadhyay
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, India
| | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, India
| |
Collapse
|
16
|
Zhang X, Wei C, Liang H, Han L. Polo-Like Kinase 4's Critical Role in Cancer Development and Strategies for Plk4-Targeted Therapy. Front Oncol 2021; 11:587554. [PMID: 33777739 PMCID: PMC7994899 DOI: 10.3389/fonc.2021.587554] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Polo-like kinases (Plks) are critical regulatory molecules during the cell cycle process. This family has five members: Plk1, 2, 3, 4, and 5. Plk4 has been identified as a master regulator of centriole replication, and its aberrant expression is closely associated with cancer development. In this review, we depict the DNA, mRNA, and protein structure of Plk4, and the regulation of Plk4 at a molecular level. Then we list the downstream targets of Plk4 and the hallmarks of cancer associated with these targets. The role of Plk4 in different cancers is also summarized. Finally, we review the inhibitors that target Plk4 in the hope of discovering effective anticancer drugs. From authors' perspective, Plk4 might represent a valuable tumor biomarker and critical target for cancer diagnosis and therapy.
Collapse
Affiliation(s)
| | | | | | - Lei Han
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
17
|
Cao F, Xia X, Fan Y, Liu Q, Song J, Zhang Q, Guo Y, Yao S. Knocking down of Polo-like kinase 2 inhibits cell proliferation and induced cell apoptosis in human glioma cells. Life Sci 2021; 270:119084. [PMID: 33482186 DOI: 10.1016/j.lfs.2021.119084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/06/2021] [Accepted: 01/14/2021] [Indexed: 12/13/2022]
Abstract
AIMS Polo-like kinase 2 (PLK2) belongs to a family of serine/threonine kinases, and it is involved in tumorigenesis. The present study aimed to explore the potential clinical significance of PLK2 in the development of gliomas. MAIN METHODS Immunohistochemistry (IHC) was performed to detect the expression of PLK2 in glioma tissues. Cell proliferation and apoptosis were determined by Cell Counting Kit 8 (CCK8) and flow cytometry analysis, respectively. KEY FINDINGS PLK2 expression gradually increased with the degree of glioma malignancy. High PLK2 expression was associated with a poor prognosis in glioma. Short hairpin RNAs targeting PLK2 (shPLK2) inhibited the viability and induced apoptosis of glioma cells, both in vitro and in vivo. Ring finger protein 180 (RNF180), an E3 ubiquitin ligase, interacted with PLK2 and induced the ubiquitination of PLK2. Overexpression of PLK2 in glioma cells significantly inhibited RNF180 upregulation-induced cell apoptosis. The expression level of RNF180 gradually decreased with the degree of glioma malignancy. SIGNIFICANCE Knocking down of PLK2 may suppress the glioma development through cancer cell proliferation inhibition and cell apoptosis promotion. Furthermore, RNF180 may mediate the ubiquitination of PLK2. The present findings may help improve the clinical management of glioma in the future.
Collapse
Affiliation(s)
- Fang Cao
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
| | - Xiangping Xia
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
| | - Yinchun Fan
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
| | - Qian Liu
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
| | - Jiancheng Song
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
| | - Qiang Zhang
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China..
| | - Shengtao Yao
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China.
| |
Collapse
|
18
|
Lin SF, Yeh CN, Huang YT, Chou TC, Wong RJ. Therapeutic inhibition of polo-like kinases in anaplastic thyroid cancer. Cancer Sci 2021; 112:803-814. [PMID: 33306266 PMCID: PMC7893987 DOI: 10.1111/cas.14769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022] Open
Abstract
Polo‐like kinases (PLKs) are potent regulators of cell proliferation and cell survival. Polo‐like kinases are potential targets in the treatment of anaplastic thyroid cancer (ATC), a rare but deadly disease. The therapeutic effects of volasertib, a PLK inhibitor, was evaluated for the treatment of ATC either alone or in combination with sorafenib. Volasertib decreased cell viability in three ATC cell lines (8505C, 8305C, and KAT18) in a dose‐dependent manner. Volasertib caused ATC cells to accumulate in G2/M phase, activated caspase‐3 activity, and induced apoptosis. Combination therapy using volasertib and sorafenib in ATC cells showed mostly synergistic effects. In vivo studies revealed that combination therapy of volasertib and sorafenib was effective in the treatment of 8505C xenografts. Single‐agent volasertib treatment was sufficient to retard 8305C tumor growth. No substantial morbidity was observed in animals that received either single‐agent or combination treatment. These preclinical findings suggest that volasertib could be an effective drug in treating ATC.
Collapse
Affiliation(s)
- Shu-Fu Lin
- Department of Internal Medicine, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan.,Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Nan Yeh
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Tung Huang
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ting-Chao Chou
- Laboratory of Preclinical Pharmacology Core, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Richard J Wong
- Head and Neck Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
19
|
Raab CA, Raab M, Becker S, Strebhardt K. Non-mitotic functions of polo-like kinases in cancer cells. Biochim Biophys Acta Rev Cancer 2021; 1875:188467. [PMID: 33171265 DOI: 10.1016/j.bbcan.2020.188467] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Inhibitors of mitotic protein kinases are currently being developed as non-neurotoxic alternatives of microtubule-targeting agents (taxanes, vinca alkaloids) which provide a substantial survival benefit for patients afflicted with different types of solid tumors. Among the mitotic kinases, the cyclin-dependent kinases, the Aurora kinases, the kinesin spindle protein and Polo-like kinases (PLKs) have emerged as attractive targets of cancer therapeutics. The functions of mammalian PLK1-5 are traditionally linked to the regulation of the cell cycle and to the stress response. Especially the key role of PLK1 and PLK4 in cellular growth and proliferation, their overexpression in multiple types of human cancer and their druggability, make them appealing targets for cancer therapy. Inhibitors for PLK1 and PLK4 are currently being tested in multiple cancer trials. The clinical success of microtubule-targeting agents is attributed not solely to the induction of a mitotic arrest in cancer cells, but also to non-mitotic effects like targeting intracellular trafficking on microtubules. This raises the question whether new cancer targets like PLK1 and PLK4 regulate critical non-mitotic functions in tumor cells. In this article we summarize the important roles of PLK1-5 for the regulation of non-mitotic signaling. Due to these functions it is conceivable that inhibitors for PLK1 or PLK4 can target interphase cells, which underscores their attractive potential as cancer drug targets. Moreover, we also describe the contribution of the tumor-suppressors PLK2, PLK3 and PLK5 to cancer cell signaling outside of mitosis. These observations highlight the urgent need to develop highly specific ATP-competitive inhibitors for PLK4 and for PLK1 like the 3rd generation PLK-inhibitor Onvansertib to prevent the inhibition of tumor-suppressor PLKs in- and outside of mitosis. The remarkable feature of PLKs to encompass a unique druggable domain, the polo-box-domain (PBD) that can be found only in PLKs offers the opportunity for the development of inhibitors that target PLKs exclusively. Beyond the development of mono-specific ATP-competitive PLK inhibitors, the PBD as drug target will support the design of new drugs that eradicate cancer cells based on the mitotic and non-mitotic function of PLK1 and PLK4.
Collapse
Affiliation(s)
| | - Monika Raab
- Department of Gynecology, Goethe-University, Frankfurt, Germany
| | - Sven Becker
- Department of Gynecology, Goethe-University, Frankfurt, Germany
| | - Klaus Strebhardt
- Department of Gynecology, Goethe-University, Frankfurt, Germany; German Cancer Consortium (DKTK), German Cancer Research Center, Partner Site Frankfurt am Main, Frankfurt, Germany.
| |
Collapse
|
20
|
RBBP6 interactome: RBBP6 isoform 3/DWNN and Nek6 interaction is critical for cell cycle regulation and may play a role in carcinogenesis. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
21
|
Jebelli A, Baradaran B, Mosafer J, Baghbanzadeh A, Mokhtarzadeh A, Tayebi L. Recent developments in targeting genes and pathways by RNAi-based approaches in colorectal cancer. Med Res Rev 2020; 41:395-434. [PMID: 32990372 DOI: 10.1002/med.21735] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/16/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022]
Abstract
A wide spectrum of genetic and epigenetic variations together with environmental factors has made colorectal cancer (CRC), which involves the colon and rectum, a challenging and heterogeneous cancer. CRC cannot be effectively overcomed by common conventional therapies including surgery, chemotherapy, targeted therapy, and hormone replacement which highlights the need for a rational design of novel anticancer therapy. Accumulating evidence indicates that RNA interference (RNAi) could be an important avenue to generate great therapeutic efficacy for CRC by targeting genes that are responsible for the viability, cell cycle, proliferation, apoptosis, differentiation, metastasis, and invasion of CRC cells. In this review, we underline the documented benefits of small interfering RNAs and short hairpin RNAs to target genes and signaling pathways related to CRC tumorigenesis. We address the synergistic effects of RNAi-mediated gene knockdown and inhibitors/chemotherapy agents to increase the sensitivity of CRC cells to common therapies. Finally, this review points new delivery systems/materials for improving the cellular uptake efficiency and reducing off-target effects of RNAi.
Collapse
Affiliation(s)
- Asiyeh Jebelli
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Mosafer
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, Wisconsin, USA
| |
Collapse
|
22
|
Ko YU, Song HY, Kim WK, Yune TY, Yun N, Oh YJ. Calpain-mediated cleavage of Fbxw7 during excitotoxicity. Neurosci Lett 2020; 736:135265. [PMID: 32707070 DOI: 10.1016/j.neulet.2020.135265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/14/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
Abstract
Neuronal cell death induced by ischemic injury has been attributed to glutamate receptor-mediated excitotoxicity, which is known to be accompanied by Ca2+ overload in the cytoplasm with concomitant activation of calcium-dependent mechanisms. More specifically, the overactivation of calpains, calcium-dependent cysteine proteases, have been associated with neuronal cell death following glutamate treatment. Previously, we observed decreased expression levels of F-box/WD repeat domain-containing protein 7 (Fbxw7) after the hyperactivation of cyclin-dependent kinase 5 (Cdk5) in cortical neurons challenged with glutamate. As determined using in vitro calpain cleavage assays, we demonstrated that the cleavage of Fbxw7 was mediated by activated calpain and attenuated in the presence of the calpain inhibitor, calpeptin. Using the rat middle cerebral artery occlusion model, we confirmed that Fbxw7 was indeed cleaved by activated calpain in the ipsilateral cortex. Based on our data, we hypothesize that the negative regulation of Fbxw7 by calpain may contribute to neuronal cell death and that the preservation of Fbxw7 by the inhibition of calpain, Cdk5, or both composes a novel protective mechanism following excitotoxicity.
Collapse
Affiliation(s)
- Yeon Uk Ko
- Department of Systems Biology, Yonsei University College of Life Science and Biotechnology, Seoul 03722, South Korea
| | - Hwa Young Song
- Department of Neuroscience, College of Medicine, Korea University, Seoul 02841, South Korea
| | - Won-Ki Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul 02841, South Korea
| | - Tae Young Yune
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul 02447, South Korea
| | - Nuri Yun
- Department of Systems Biology, Yonsei University College of Life Science and Biotechnology, Seoul 03722, South Korea.
| | - Young J Oh
- Department of Systems Biology, Yonsei University College of Life Science and Biotechnology, Seoul 03722, South Korea.
| |
Collapse
|
23
|
Zhang Z, Hu Q, Xu W, Liu W, Liu M, Sun Q, Ye Z, Fan G, Qin Y, Xu X, Yu X, Ji S. Function and regulation of F-box/WD repeat-containing protein 7. Oncol Lett 2020; 20:1526-1534. [PMID: 32724394 PMCID: PMC7377190 DOI: 10.3892/ol.2020.11728] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/16/2020] [Indexed: 01/03/2023] Open
Abstract
The ubiquitin-proteasome system is an important post-translational modification system involved in numerous biological processes, such as cell cycle regulation, gene transcription, signal transduction, apoptosis, differentiation and development. F-box/WD repeat-containing protein 7 (FBXW7) is one of the most studied F-box (FBX) proteins, serving as substrate recognition component of S phase kinase-associated protein 1-Cullin 1-FBX protein complexes. As a tumor suppressor, FBXW7 recognizes numerous proto-oncoproteins and promotes their ubiquitination and subsequent proteasomal degradation. FBXW7 is regulated at different levels, leading to tunable and specific control of the activity and abundance of its substrates. Therefore, genetic mutations or decreases in its expression serve an important biological role in tumor development. In-depth studies and identification of additional substrates targeted by FBXW7 have suggested a signaling network regulated by FBXW7, including its tumor-inhibitory role. The present review focused on the role of FBXW7 in tumor suppression and its application in cancer therapy.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Mengqi Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Qiqing Sun
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Guixiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Shanghai Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
24
|
Yumimoto K, Nakayama KI. Recent insight into the role of FBXW7 as a tumor suppressor. Semin Cancer Biol 2020; 67:1-15. [PMID: 32113998 DOI: 10.1016/j.semcancer.2020.02.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/15/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022]
Abstract
FBXW7 (also known as Fbw7, Sel10, hCDC4, or hAgo) is a tumor suppressor and the most frequently mutated member of the F-box protein family in human cancers. FBXW7 functions as the substrate recognition component of an SCF-type E3 ubiquitin ligase. It specifically controls the proteasome-mediated degradation of many oncoproteins such as c-MYC, NOTCH, KLF5, cyclin E, c-JUN, and MCL1. In this review, we summarize the molecular and biological features of FBXW7 and its substrates as well as the impact of mutations of FBXW7 on cancer development. We also address the clinical potential of anticancer therapy targeting FBXW7.
Collapse
Affiliation(s)
- Kanae Yumimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan.
| |
Collapse
|
25
|
Badarudeen B, Gupta R, Nair SV, Chandrasekharan A, Manna TK. The ubiquitin ligase FBXW7 targets the centriolar assembly protein HsSAS-6 for degradation and thereby regulates centriole duplication. J Biol Chem 2020; 295:4428-4437. [PMID: 32086376 DOI: 10.1074/jbc.ac119.012178] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/19/2020] [Indexed: 11/06/2022] Open
Abstract
Formation of a single new centriole from a pre-existing centriole is strictly controlled to maintain correct centrosome number and spindle polarity in cells. However, the mechanisms that govern this process are incompletely understood. Here, using several human cell lines, immunofluorescence and structured illumination microscopy methods, and ubiquitination assays, we show that the E3 ubiquitin ligase F-box and WD repeat domain-containing 7 (FBXW7), a subunit of the SCF ubiquitin ligase, down-regulates spindle assembly 6 homolog (HsSAS-6), a key protein required for procentriole cartwheel assembly, and thereby regulates centriole duplication. We found that FBXW7 abrogation stabilizes HsSAS-6 and increases its recruitment to the mother centriole at multiple sites, leading to supernumerary centrioles. Ultrastructural analyses revealed that FBXW7 is broadly localized on the mother centriole and that its presence is reduced at the site where the HsSAS-6-containing procentriole is formed. This observation suggested that FBXW7 restricts procentriole assembly to a specific site to generate a single new centriole. In contrast, during HsSAS-6 overexpression, FBXW7 strongly associated with HsSAS-6 at the centriole. We also found that SCFFBXW7 interacts with HsSAS-6 and targets it for ubiquitin-mediated degradation. Further, we identified putative phosphodegron sites in HsSAS-6, whose substitutions rendered it insensitive to FBXW7-mediated degradation and control of centriole number. In summary, SCFFBXW7 targets HsSAS-6 for degradation and thereby controls centriole biogenesis by restraining HsSAS-6 recruitment to the mother centriole, a molecular mechanism that controls supernumerary centrioles/centrosomes and the maintenance of bipolar spindles.
Collapse
Affiliation(s)
- Binshad Badarudeen
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Ria Gupta
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Sreeja V Nair
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, Kerala, India
| | | | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, Thiruvananthapuram 695551, Kerala, India
| |
Collapse
|
26
|
Computational basis for the design of PLK-2 inhibitors. Struct Chem 2020. [DOI: 10.1007/s11224-019-01394-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
27
|
Linked-read Sequencing Analysis Reveals Tumor-specific Genome Variation Landscapes in Neurofibromatosis Type 2 (NF2) Patients. Otol Neurotol 2020; 40:e150-e159. [PMID: 30624408 DOI: 10.1097/mao.0000000000002096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
HYPOTHESIS We hypothesize that genomic variants including deletions, insertions, inversions, and tandem duplications beyond the changes in tumor suppressor NF2 gene affect gene expression of tumor-specific pathways in vestibular schwannomas (VS) patients with Neurofibromatosis type 2 (NF2), thus contributing to their clinical behavior. BACKGROUND Genomic variation could reconfigure transcription in NF2 transformation process. Therefore, genome-wide high-resolution characterization of structural variants (SV) landscapes in NF2 tumors can expand our understanding of the genes regulating the clinical phenotypes in NF2-associated VS. METHODS We performed whole-genome haplotype-specific structural variation analysis using synthetic linked reads generated through microfluidics-based barcoding of high molecular weight DNA followed by high-coverage Illumina paired-end whole-genome sequencing from 10 patients' tumors of different growth rates and their matching blood samples. RESULTS NF2 tumor-specific deletions and large SVs were detected and can be classified based on their association with tumor growth rates. Through detailed annotation of these mutations, we uncover common alleles affected by these deletions and large SVs that can be associated with signaling pathways implicated in cell proliferation and tumorigenesis. CONCLUSION The genomic variation landscape of NF2-related VS was investigated through whole-genome linked-read sequencing. Large SVs, in addition to deletions, were identified and may serve as modulators of clinical behavior.
Collapse
|
28
|
Ling H, Peng L, Wang J, Rahhal R, Seto E. Histone Deacetylase SIRT1 Targets Plk2 to Regulate Centriole Duplication. Cell Rep 2019; 25:2851-2865.e3. [PMID: 30517871 DOI: 10.1016/j.celrep.2018.11.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/04/2018] [Accepted: 11/02/2018] [Indexed: 11/17/2022] Open
Abstract
The protein deacetylase SIRT1 (Sirtuin 1) regulates many cellular processes, including cell-cycle progression, DNA damage response, and metabolism. Although the centrosome is a key regulator of cell-cycle progression and genome stability, little is known concerning SIRT1 controlled centrosome-associated events. Here we report that the centrosome protein Plk2 is acetylated and undergoes deacetylation by SIRT1. Acetylation protects Plk2 from ubiquitination, and SIRT1-mediated deacetylation promotes ubiquitin-dependent degradation of Plk2. SIRT1 controls centriole duplication by temporally modulating centrosomal Plk2 levels. AURKA phosphorylates SIRT1 and promotes the SIRT1-Plk2 interaction in mitosis. In early-mid G1, phosphorylated SIRT1 deacetylates and promotes Plk2 degradation. In late G1, SIRT1 is hypophosphorylated and its affinity to Plk2 is decreased, resulting in a rapid accumulation of centrosomal Plk2, which contributes to the timely initiation of centriole duplication. Collectively, our findings uncover a critical role of SIRT1 in centriole duplication and provide a mechanistic insight into SIRT1-mediated centrosome-associated functions.
Collapse
Affiliation(s)
- Hongbo Ling
- George Washington University Cancer Center, Washington, DC 20052, USA; Department of Biochemistry & Molecular Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Lirong Peng
- George Washington University Cancer Center, Washington, DC 20052, USA; Department of Biochemistry & Molecular Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Jianbo Wang
- Department of Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Raneen Rahhal
- George Washington University Cancer Center, Washington, DC 20052, USA; Department of Biochemistry & Molecular Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Edward Seto
- George Washington University Cancer Center, Washington, DC 20052, USA; Department of Biochemistry & Molecular Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA.
| |
Collapse
|
29
|
Site-specific phosphorylation of Fbxw7 by Cdk5/p25 and its resulting decreased stability are linked to glutamate-induced excitotoxicity. Cell Death Dis 2019; 10:579. [PMID: 31371703 PMCID: PMC6675790 DOI: 10.1038/s41419-019-1818-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/08/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a serine/threonine protein kinase that regulates brain development and neurodegeneration. Cdk5 is activated by p25 that is generated from calpain-dependent cleavage of p35. The generation of p25 is responsible for the aberrant hyper-activation of Cdk5, which causes neurodegeneration. Using in vitro assays, we discovered that F-box/WD repeat-containing protein 7 (Fbxw7) is a new substrate of Cdk5. Additionally, Cdk5-dependent phosphorylation of Fbxw7 was detected in the presence of p25, and two amino acid residues (S349 and S372) were determined to be major phosphorylation sites. This phosphorylation was eventually linked to decreased stability of Fbxw7. Using a culture model of cortical neurons challenged with glutamate, we confirmed that decreased stability of Fbxw7 was indeed Cdk5-dependent. Furthermore, diminished levels of Fbxw7 led to increased levels of transcription factor AP-1 (c-Jun), a known substrate of Fbxw7. Given that previous reports demonstrate that c-Jun plays a role in accelerating neuronal apoptosis in these pathological models, our data support the concepts of a molecular cascade in which Cdk5-mediated phosphorylation of Fbxw7 negatively regulates Fbxw7 expression, thereby contributing to neuronal cell death following glutamate-mediated excitotoxicity.
Collapse
|
30
|
Lin SF, Lin JD, Yeh CN, Huang YT, Chou TC, Wong RJ. Targeting PLKs as a therapeutic approach to well-differentiated thyroid cancer. Endocr Relat Cancer 2019; 26:727-738. [PMID: 31189135 PMCID: PMC7475022 DOI: 10.1530/erc-18-0555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022]
Abstract
Polo-like kinases (PLKs) are pivotal regulators of cell proliferation and cell survival; therefore, PLKs may be potential targets in the treatment of malignancy. The therapeutic effects of volasertib, a PLKs inhibitor for papillary and follicular thyroid cancer (known as well-differentiated thyroid cancer (WDTC)), were evaluated in this study. Volasertib inhibited cell proliferation in two papillary and two follicular thyroid cancer cell lines in a dose-dependent manner. Volasertib treatment reduced cells in the S phase and increased cells in the G2/M phase. Volasertib activated caspase-3 activity and induced apoptosis. Drug combinations of volasertib and sorafenib showed mostly synergism in four well-differentiated thyroid carcinoma cell lines in vitro. Volasertib treatment in vivo retarded the growth of a papillary thyroid tumor model. Furthermore, the combination of volasertib with sorafenib was more effective than a single treatment of either in a follicular thyroid cancer xenograft model. Promising safety profiles appeared in animals treated with either volasertib alone or volasertib and sorafenib combination therapy. These findings support volasertib as a potential drug for the treatment of patients with WDTC.
Collapse
Affiliation(s)
- Shu-Fu Lin
- Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Chang Gung University, Taoyuan, Taiwan
| | - Jen-Der Lin
- Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Chang Gung University, Taoyuan, Taiwan
| | - Chun-Nan Yeh
- Chang Gung University, Taoyuan, Taiwan
- Department of Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Tung Huang
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ting-Chao Chou
- Laboratory of Preclinical Pharmacology Core, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Current address: PD Science, Inc., 599 Mill Run, Paramus, NJ, USA
| | - Richard J. Wong
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
31
|
Abdullah M, Guruprasad L. Structural insights into the inhibitor binding and new inhibitor design to Polo-like kinase-1 Polo-box domain using computational studies. J Biomol Struct Dyn 2019; 37:3410-3421. [PMID: 30146942 DOI: 10.1080/07391102.2018.1515663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 08/13/2018] [Indexed: 12/27/2022]
Abstract
Polo box domain (PBD) from Polo-Like Kinase-1 (PLK-1) a cell cycle regulator is one of the important non-kinase targets implicated in various cancers. The crystal structure of PLK-1 PBD bound to phosphopeptide inhibitor is available and acylthiourea derivatives have been reported as potent PBD inhibitors. In this work, structure and ligand-based pharmacophore methods have been used to identify new PBD inhibitors. The binding of acylthiourea analogs and new inhibitors to PBD were assessed using molecular docking and molecular dynamics simulations to understand their binding interactions, investigate the complex stability and reveal the molecular basis for inhibition. This study provides the binding free energies and residue-wise contributions to decipher the essential interactions in the protein-inhibitor complementarity for complex formation and the design of new PBD inhibitors with better binding. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Maaged Abdullah
- a School of Chemistry , University of Hyderabad , Hyderabad , India
| | | |
Collapse
|
32
|
Richter KT, Kschonsak YT, Vodicska B, Hoffmann I. FBXO45-MYCBP2 regulates mitotic cell fate by targeting FBXW7 for degradation. Cell Death Differ 2019; 27:758-772. [PMID: 31285543 DOI: 10.1038/s41418-019-0385-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/14/2019] [Accepted: 06/18/2019] [Indexed: 11/09/2022] Open
Abstract
Cell fate decision upon prolonged mitotic arrest induced by microtubule-targeting agents depends on the activity of the tumor suppressor and F-box protein FBXW7. FBXW7 promotes mitotic cell death and prevents premature escape from mitosis through mitotic slippage. Mitotic slippage is a process that can cause chemoresistance and tumor relapse. Therefore, understanding the mechanisms that regulate the balance between mitotic cell death and mitotic slippage is an important task. Here we report that FBXW7 protein levels markedly decline during extended mitotic arrest. FBXO45 binds to a conserved acidic N-terminal motif of FBXW7 specifically under a prolonged delay in mitosis, leading to ubiquitylation and subsequent proteasomal degradation of FBXW7 by the FBXO45-MYCBP2 E3 ubiquitin ligase. Moreover, we find that FBXO45-MYCBP2 counteracts FBXW7 in that it promotes mitotic slippage and prevents cell death in mitosis. Targeting this interaction represents a promising strategy to prevent chemotherapy resistance.
Collapse
Affiliation(s)
- Kai T Richter
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Yvonne T Kschonsak
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Barbara Vodicska
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Ingrid Hoffmann
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| |
Collapse
|
33
|
Wang Y, An Y, Ma Y, Guo J. F-box/WD-40 repeat-containing protein 7: A potential target in the progression and treatment of gastrointestinal malignancy. Oncol Lett 2019; 17:3625-3634. [PMID: 30881487 PMCID: PMC6403509 DOI: 10.3892/ol.2019.10036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/21/2019] [Indexed: 12/21/2022] Open
Abstract
Cancer is a principal cause of human morbidity and mortality, with gastrointestinal malignancies, in particular, resulting in a marked number of tumor-associated mortalities. The progression of gastrointestinal malignancy is regulated by a variety of aberrantly expressed proteins, a number of which facilitate tumor progression, whereas, others function as tumor suppressors. The detection of such proteins not only contributes to the early diagnosis of cancer, they may additionally serve as potential therapeutic targets. In normal tissues, numerous proteins encoded by proto-oncoproteins are degraded by ubiquitylation enzymes, consisting of F-box/WD-40 repeat-containing protein 7 (Fbw7) and other proteins, thus avoiding tumorigenesis and maintaining homeostasis. In tumor tissues, the downregulation of Fbw7, caused by various factors, leads to disorders in ubiquitinase synthesis, which may induce tumor progression and chemoresistance, particularly in gastrointestinal malignancy. Therefore, an in-depth study of the regulatory mechanisms involved in disorders of Fbw7 expression and the role of Fbw7 in chemoresistance of gastrointestinal tumors may suggest improvements for early diagnostic screening and targeted therapy.
Collapse
Affiliation(s)
- Yizhi Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yang An
- Department of Anesthesia, First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yi Ma
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
34
|
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: 331] [Impact Index Per Article: 55.2] [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
|
35
|
Cheng X, Zheng J, Li G, Göbel V, Zhang H. Degradation for better survival? Role of ubiquitination in epithelial morphogenesis. Biol Rev Camb Philos Soc 2018; 93:1438-1460. [PMID: 29493067 DOI: 10.1111/brv.12404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/31/2018] [Accepted: 02/05/2018] [Indexed: 02/06/2023]
Abstract
As a prevalent post-translational modification, ubiquitination is essential for many developmental processes. Once covalently attached to the small and conserved polypeptide ubiquitin (Ub), a substrate protein can be directed to perform specific biological functions via its Ub-modified form. Three sequential catalytic reactions contribute to this process, among which E3 ligases serve to identify target substrates and promote the activated Ub to conjugate to substrate proteins. Ubiquitination has great plasticity, with diverse numbers, topologies and modifications of Ub chains conjugated at different substrate residues adding a layer of complexity that facilitates a huge range of cellular functions. Herein, we highlight key advances in the understanding of ubiquitination in epithelial morphogenesis, with an emphasis on the latest insights into its roles in cellular events involved in polarized epithelial tissue, including cell adhesion, asymmetric localization of polarity determinants and cytoskeletal organization. In addition, the physiological roles of ubiquitination are discussed for typical examples of epithelial morphogenesis, such as lung branching, vascular development and synaptic formation and plasticity. Our increased understanding of ubiquitination in epithelial morphogenesis may provide novel insights into the molecular mechanisms underlying epithelial regeneration and maintenance.
Collapse
Affiliation(s)
- Xiaoxiang Cheng
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Gang Li
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Verena Göbel
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114,, U.S.A
| | - Hongjie Zhang
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| |
Collapse
|
36
|
Physiological functions of FBW7 in cancer and metabolism. Cell Signal 2018; 46:15-22. [PMID: 29474981 DOI: 10.1016/j.cellsig.2018.02.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/16/2018] [Accepted: 02/18/2018] [Indexed: 12/11/2022]
Abstract
FBW7 is one of the most well characterized F-box proteins that serve as substrate recognition subunits of SCF (Skp1-Cullin 1-F-box proteins) E3 ubiquitin ligase complexes. SCFFBW7 plays key roles in regulating cell cycle progression, differentiation, and stem cell maintenance largely through targeting a broad range of oncogenic substrates for proteasome-dependent degradation. The identification of an increasing number of FBW7 substrates for ubiquitination, and intensive in vitro and in vivo studies have revealed a network of signaling components controlled by FBW7 that contributes to metabolic regulation as well as its tumor suppressor role. Here we mainly focus on recent findings that highlight a critical role for FBW7 in cancer and metabolism.
Collapse
|
37
|
Nikonova AS, Deneka AY, Kiseleva AA, Korobeynikov V, Gaponova A, Serebriiskii IG, Kopp MC, Hensley HH, Seeger-Nukpezah TN, Somlo S, Proia DA, Golemis EA. Ganetespib limits ciliation and cystogenesis in autosomal-dominant polycystic kidney disease (ADPKD). FASEB J 2018; 32:2735-2746. [PMID: 29401581 DOI: 10.1096/fj.201700909r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is associated with progressive formation of renal cysts, kidney enlargement, hypertension, and typically end-stage renal disease. In ADPKD, inherited mutations disrupt function of the polycystins (encoded by PKD1 and PKD2), thus causing loss of a cyst-repressive signal emanating from the renal cilium. Genetic studies have suggested ciliary maintenance is essential for ADPKD pathogenesis. Heat shock protein 90 (HSP90) clients include multiple proteins linked to ciliary maintenance. We determined that ganetespib, a clinical HSP90 inhibitor, inhibited proteasomal repression of NEK8 and the Aurora-A activator trichoplein, rapidly activating Aurora-A kinase and causing ciliary loss in vitro. Using conditional mouse models for ADPKD, we performed long-term (10 or 50 wk) dosing experiments that demonstrated HSP90 inhibition caused durable in vivo loss of cilia, controlled cystic growth, and ameliorated symptoms induced by loss of Pkd1 or Pkd2. Ganetespib efficacy was not increased by combination with 2-deoxy-d-glucose, a glycolysis inhibitor showing some promise for ADPKD. These studies identify a new biologic activity for HSP90 and support a cilia-based mechanism for cyst repression.-Nikonova, A. S., Deneka, A. Y., Kiseleva, A. A., Korobeynikov, V., Gaponova, A., Serebriiskii, I. G., Kopp, M. C., Hensley, H. H., Seeger-Nukpezah, T. N., Somlo, S., Proia, D. A., Golemis, E. A. Ganetespib limits ciliation and cystogenesis in autosomal-dominant polycystic kidney disease (ADPKD).
Collapse
Affiliation(s)
- Anna S Nikonova
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Alexander Y Deneka
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Kazan Federal University, Kazan, Russia
| | - Anna A Kiseleva
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Kazan Federal University, Kazan, Russia
| | - Vladislav Korobeynikov
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Anna Gaponova
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Laboratory of Genome Engineering, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,Immanuel Kant Baltic Federal University, Konigsberg, Russia
| | - Ilya G Serebriiskii
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Kazan Federal University, Kazan, Russia
| | - Meghan C Kopp
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Cancer Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Harvey H Hensley
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Tamina N Seeger-Nukpezah
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Department I of Internal Medicine and Center for Integrated Oncology, University of Cologne, Cologne, Germany
| | - Stefan Somlo
- Departments of Internal Medicine and Genetics, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - David A Proia
- Synta Pharmaceuticals Corporation, Lexington, Massachusetts, USA
| | - Erica A Golemis
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
38
|
Zhan MM, Yang Y, Luo J, Zhang XX, Xiao X, Li S, Cheng K, Xie Z, Tu Z, Liao C. Design, synthesis, and biological evaluation of novel highly selective polo-like kinase 2 inhibitors based on the tetrahydropteridin chemical scaffold. Eur J Med Chem 2017; 143:724-731. [PMID: 29220793 DOI: 10.1016/j.ejmech.2017.11.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 01/08/2023]
Abstract
Polo-like kinase 2 (Plk2) is a potential target for the treatment of cancer, which displays an important role in tumor cell proliferation and survival. In this report, according to the analysis of critical amino acid residue differences among Plk1, Plk2 and Plk3, and structure-based drug design strategies, two novel series of selective Plk2 inhibitors based on tetrahydropteridin chemical scaffold were designed and synthesized to target two specific residues, Lys86 and Tyr161 of Plk2. All compounds were evaluated for their inhibitory activity against Plk1-Plk3 and the cellular inhibition activity on six different human cancer cell lines. All efforts led to the identification of the most potent compounds C2 (3.40 nM against Plk2) and C21 (4.88 nM against Plk2) from the first and second series of selective Plk2 inhibitors respectively. Additionally, the selectivity of C21 over Plk1/3 was significantly increased with the selectivity indexes of 12.57 and 910.06. Moreover, most of our compounds exhibited antitumor activity in the nanomolar range in the MTT assay, indicating that our compounds, especially C2 and C21 could be promising Plk2 inhibitors for further anticancer research.
Collapse
Affiliation(s)
- Mei-Miao Zhan
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Yang Yang
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Jinfeng Luo
- High Throughput Drug Screening Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, PR China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, PR China
| | - Xing-Xing Zhang
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Xuan Xiao
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Shiyu Li
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Kai Cheng
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Zhouling Xie
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Zhengchao Tu
- High Throughput Drug Screening Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, PR China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, PR China.
| | - Chenzhong Liao
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China.
| |
Collapse
|
39
|
Zitouni S, Méchali F, Papin C, Choquet A, Roche D, Baldin V, Coux O, Bonne-Andrea C. The stability of Fbw7α in M-phase requires its phosphorylation by PKC. PLoS One 2017; 12:e0183500. [PMID: 28850619 PMCID: PMC5574586 DOI: 10.1371/journal.pone.0183500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/05/2017] [Indexed: 12/21/2022] Open
Abstract
Fbw7 is a tumor suppressor often deleted or mutated in human cancers. It serves as the substrate-recruiting subunit of a SCF ubiquitin ligase that targets numerous critical proteins for degradation, including oncoproteins and master transcription factors. Cyclin E was the first identified substrate of the SCFFbw7 ubiquitin ligase. In human cancers bearing FBXW7-gene mutations, deregulation of cyclin E turnover leads to its aberrant expression in mitosis. We investigated Fbw7 regulation in Xenopus eggs, which, although arrested in a mitotic-like phase, naturally express high levels of cyclin E. Here, we report that Fbw7α, the only Fbw7 isoform detected in eggs, is phosphorylated by PKC (protein kinase C) at a key residue (S18) in a manner coincident with Fbw7α inactivation. We show that this PKC-dependent phosphorylation and inactivation of Fbw7α also occurs in mitosis during human somatic cell cycles, and importantly is critical for Fbw7α stabilization itself upon nuclear envelope breakdown. Finally, we provide evidence that S18 phosphorylation, which lies within the intrinsically disordered N-terminal region specific to the α-isoform reduces the capacity of Fbw7α to dimerize and to bind cyclin E. Together, these findings implicate PKC in an evolutionarily-conserved pathway that aims to protect Fbw7α from degradation by keeping it transiently in a resting, inactive state.
Collapse
Affiliation(s)
- Sihem Zitouni
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, UMR 5237, Université de Montpellier, Montpellier, France
| | - Francisca Méchali
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, UMR 5237, Université de Montpellier, Montpellier, France
| | - Catherine Papin
- Institut de Génétique Humaine, CNRS, UMR 9002, Université de Montpellier, Montpellier, France
| | - Armelle Choquet
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Université de Montpellier, Montpellier, France
| | - Daniel Roche
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, UMR 5237, Université de Montpellier, Montpellier, France
- Institut de Biologie Computationnelle, LIRMM, CNRS, Université de Montpellier, Montpellier, France
| | - Véronique Baldin
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, UMR 5237, Université de Montpellier, Montpellier, France
| | - Olivier Coux
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, UMR 5237, Université de Montpellier, Montpellier, France
| | - Catherine Bonne-Andrea
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, UMR 5237, Université de Montpellier, Montpellier, France
- * E-mail:
| |
Collapse
|
40
|
Merkel cell polyomavirus small T antigen induces genome instability by E3 ubiquitin ligase targeting. Oncogene 2017; 36:6784-6792. [PMID: 28846109 PMCID: PMC5720911 DOI: 10.1038/onc.2017.277] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/29/2017] [Accepted: 07/03/2017] [Indexed: 02/08/2023]
Abstract
The formation of a bipolar mitotic spindle is an essential process for the equal segregation of duplicated DNA into two daughter cells during mitosis. As a result of deregulated cellular signaling pathways, cancer cells often suffer a loss of genome integrity that might etiologically contribute to carcinogenesis. Merkel cell polyomavirus (MCV) small T (sT) oncoprotein induces centrosome overduplication, aneuploidy, chromosome breakage and the formation of micronuclei by targeting cellular ligases through a sT domain that also inhibits MCV large T oncoprotein turnover. These results provide important insight as to how centrosome number and chromosomal stability can be affected by the E3 ligase targeting capacity of viral oncoproteins such as MCV sT, which may contribute to Merkel cell carcinogenesis.
Collapse
|
41
|
Mahathre MM, Rida PC, Aneja R. The more the messier: centrosome amplification as a novel biomarker for personalized treatment of colorectal cancers. J Biomed Res 2016; 30:441-451. [PMID: 27924065 PMCID: PMC5138576 DOI: 10.7555/jbr.30.20150109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/12/2015] [Indexed: 01/10/2023] Open
Abstract
Colon cancer is currently the third most common cancer and second most fatal cancer in the United States, resulting in approximately 600,000 deaths annually. Though colorectal cancer death rates are decreasing by about 3% every year, disease outcomes could be substantially improved with more research into the drivers of colon carcinogenesis, the determinants of aggressiveness in colorectal cancer and the identification of biomarkers that could enable choice of more optimal treatments. Colon carcinogenesis is notably a slow process that can take decades. Known factors that contribute to the development of colon cancer are mutational, epigenetic and environmental, and risk factors include age, history of polyps and family history of colon cancer. Colorectal cancers exhibit heterogeneity in their features and are often characterized by the presence of chromosomal instability, microscopic satellite instability, or CpG island methylator phenotype. In this review, we propose that centrosome amplification may be a widespread occurrence in colorectal cancers and could potently influence tumor biology. Moreover, the quantitation of this cancer-specific anomaly could offer valuable prognostic information and pave the way for further customization of treatment based on the organellar profile of patients. Patient stratification models that take into account centrosomal status could thus potentially reduce adverse side effects and result in improved outcomes for colorectal cancer patients.
Collapse
Affiliation(s)
- Monica M Mahathre
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Padmashree Cg Rida
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA.,Novazoi Theranostics Inc., Plano, TX 75025, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA.,Institute of Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Center for Obesity Research, Georgia State University, Atlanta, GA 30303, USA;
| |
Collapse
|
42
|
Kratz AS, Richter KT, Schlosser YT, Schmitt M, Shumilov A, Delecluse HJ, Hoffmann I. Fbxo28 promotes mitotic progression and regulates topoisomerase IIα-dependent DNA decatenation. Cell Cycle 2016; 15:3419-3431. [PMID: 27754753 DOI: 10.1080/15384101.2016.1246093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Topoisomerase IIα is an essential enzyme that resolves topological constraints in genomic DNA. It functions in disentangling intertwined chromosomes during anaphase leading to chromosome segregation thus preserving genomic stability. Here we describe a previously unrecognized mechanism regulating topoisomerase IIα activity that is dependent on the F-box protein Fbxo28. We find that Fbxo28, an evolutionarily conserved protein, is required for proper mitotic progression. Interfering with Fbxo28 function leads to a delay in metaphase-to-anaphase progression resulting in mitotic defects as lagging chromosomes, multipolar spindles and multinucleation. Furthermore, we find that Fbxo28 interacts and colocalizes with topoisomerase IIα throughout the cell cycle. Depletion of Fbxo28 results in an increase in topoisomerase IIα-dependent DNA decatenation activity. Interestingly, blocking the interaction between Fbxo28 and topoisomerase IIα also results in multinucleated cells. Our findings suggest that Fbxo28 regulates topoisomerase IIα decatenation activity and plays an important role in maintaining genomic stability.
Collapse
Affiliation(s)
- Anne-Sophie Kratz
- a Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center , Heidelberg , Germany
| | - Kai T Richter
- a Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center , Heidelberg , Germany
| | - Yvonne T Schlosser
- a Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center , Heidelberg , Germany
| | - Miriam Schmitt
- a Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center , Heidelberg , Germany
| | - Anatoliy Shumilov
- b Pathogenesis of Virus Associated Tumors, F100, German Cancer Research Center , Heidelberg , Germany
| | - Henri-Jacques Delecluse
- b Pathogenesis of Virus Associated Tumors, F100, German Cancer Research Center , Heidelberg , Germany
| | - Ingrid Hoffmann
- a Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center , Heidelberg , Germany
| |
Collapse
|
43
|
Abstract
For over a century, the abnormal movement or number of centrosomes has been linked with errors of chromosomes distribution in mitosis. While not essential for the formation of the mitotic spindle, the presence and location of centrosomes has a major influence on the manner in which microtubules interact with the kinetochores of replicated sister chromatids and the accuracy with which they migrate to resulting daughter cells. A complex network has evolved to ensure that cells contain the proper number of centrosomes and that their location is optimal for effective attachment of emanating spindle fibers with the kinetochores. The components of this network are regulated through a series of post-translational modifications, including ubiquitin and ubiquitin-like modifiers, which coordinate the timing and strength of signaling events key to the centrosome cycle. In this review, we examine the role of the ubiquitin system in the events relating to centriole duplication and centrosome separation, and discuss how the disruption of these functions impacts chromosome segregation.
Collapse
|
44
|
Plk2 promotes tumor growth and inhibits apoptosis by targeting Fbxw7/Cyclin E in colorectal cancer. Cancer Lett 2016; 380:457-466. [DOI: 10.1016/j.canlet.2016.07.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/20/2016] [Accepted: 07/10/2016] [Indexed: 12/21/2022]
|
45
|
Fontebasso AM, Shirinian M, Khuong-Quang DA, Bechet D, Gayden T, Kool M, De Jay N, Jacob K, Gerges N, Hutter B, Şeker-Cin H, Witt H, Montpetit A, Brunet S, Lepage P, Bourret G, Klekner A, Bognár L, Hauser P, Garami M, Farmer JP, Montes JL, Atkinson J, Lambert S, Kwan T, Korshunov A, Tabori U, Collins VP, Albrecht S, Faury D, Pfister SM, Paulus W, Hasselblatt M, Jones DTW, Jabado N. Non-random aneuploidy specifies subgroups of pilocytic astrocytoma and correlates with older age. Oncotarget 2016; 6:31844-56. [PMID: 26378811 PMCID: PMC4741644 DOI: 10.18632/oncotarget.5571] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 08/15/2015] [Indexed: 11/25/2022] Open
Abstract
Pilocytic astrocytoma (PA) is the most common brain tumor in children but is rare in adults, and hence poorly studied in this age group. We investigated 222 PA and report increased aneuploidy in older patients. Aneuploid genomes were identified in 45% of adult compared with 17% of pediatric PA. Gains were non-random, favoring chromosomes 5, 7, 6 and 11 in order of frequency, and preferentially affecting non-cerebellar PA and tumors with BRAF V600E mutations and not with KIAA1549-BRAF fusions or FGFR1 mutations. Aneuploid PA differentially expressed genes involved in CNS development, the unfolded protein response, and regulators of genomic stability and the cell cycle (MDM2, PLK2),whose correlated programs were overexpressed specifically in aneuploid PA compared to other glial tumors. Thus, convergence of pathways affecting the cell cycle and genomic stability may favor aneuploidy in PA, possibly representing an additional molecular driver in older patients with this brain tumor.
Collapse
Affiliation(s)
- Adam M Fontebasso
- Division of Experimental Medicine, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Margret Shirinian
- Department of Experimental Pathology, Immunology and Microbiology, American University Of Beirut, Beirut, Lebanon
| | - Dong-Anh Khuong-Quang
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Denise Bechet
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Tenzin Gayden
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Marcel Kool
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Nicolas De Jay
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Karine Jacob
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Noha Gerges
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Barbara Hutter
- Division of Theoretical Bioinformatics, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Huriye Şeker-Cin
- Division of Pediatric Neurooncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Hendrik Witt
- Division of Pediatric Neurooncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Alexandre Montpetit
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Sébastien Brunet
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Pierre Lepage
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Geneviève Bourret
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Almos Klekner
- Department of Neurosurgery, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - László Bognár
- Department of Neurosurgery, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Peter Hauser
- 2nd Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - Miklós Garami
- 2nd Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - Jean-Pierre Farmer
- Department of Neurosurgery, Montreal Children's Hospital and McGill University Health Centre, Montreal, Canada
| | - Jose-Luis Montes
- Department of Neurosurgery, Montreal Children's Hospital and McGill University Health Centre, Montreal, Canada
| | - Jeffrey Atkinson
- Department of Neurosurgery, Montreal Children's Hospital and McGill University Health Centre, Montreal, Canada
| | - Sally Lambert
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Tony Kwan
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Andrey Korshunov
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Uri Tabori
- Division of Pediatric Hematology-Oncology and The Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - V Peter Collins
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Steffen Albrecht
- Department of Pathology, Montreal Children's Hospital and McGill University Health Centre, Montreal, Canada
| | - Damien Faury
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Werner Paulus
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - David T W Jones
- Division of Pediatric Neurooncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Nada Jabado
- Division of Experimental Medicine, McGill University and McGill University Health Centre, Montreal, Quebec, Canada.,Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
46
|
Reddy MVR, Akula B, Jatiani S, Vasquez-Del Carpio R, Billa VK, Mallireddigari MR, Cosenza SC, Venkata Subbaiah DRC, Bharathi EV, Pallela VR, Ramkumar P, Jain R, Aggarwal AK, Reddy EP. Discovery of 2-(1H-indol-5-ylamino)-6-(2,4-difluorophenylsulfonyl)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one (7ao) as a potent selective inhibitor of Polo like kinase 2 (PLK2). Bioorg Med Chem 2016; 24:521-44. [PMID: 26762835 PMCID: PMC5947326 DOI: 10.1016/j.bmc.2015.11.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/20/2015] [Accepted: 11/29/2015] [Indexed: 11/20/2022]
Abstract
Several families of protein kinases have been shown to play a critical role in the regulation of cell cycle progression, particularly progression through mitosis. These kinase families include the Aurora kinases, the Mps1 gene product and the Polo Like family of protein kinases (PLKs). The PLK family consists of five members and of these, the role of PLK1 in human cancer is well documented. PLK2 (SNK), which is highly homologous to PLK1, has been shown to play a critical role in centriole duplication and is also believed to play a regulatory role in the survival pathway by physically stabilizing the TSC1/2 complex in tumor cells under hypoxic conditions. As a part of our research program, we have developed a library of novel ATP mimetic chemotypes that are cytotoxic against a panel of cancer cell lines. We show that one of these chemotypes, the 6-arylsulfonyl pyridopyrimidinones, induces apoptosis of human tumor cell lines in nanomolar concentrations. The most potent of these compounds, 7ao, was found to be a highly specific inhibitor of PLK2 when profiled against a panel of 288 wild type, 55 mutant and 12 lipid kinases. Here, we describe the synthesis, structure activity relationship, in vitro kinase specificity and biological activity of the lead compound, 7ao.
Collapse
Affiliation(s)
- M V Ramana Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States.
| | - Balireddy Akula
- Department of Medicinal Chemistry, Onconova Therapeutics Inc., 375 Pheasant Run, Newtown, PA 18940-3423, United States
| | - Shashidhar Jatiani
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - Rodrigo Vasquez-Del Carpio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - Vinay K Billa
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - Muralidhar R Mallireddigari
- Department of Medicinal Chemistry, Onconova Therapeutics Inc., 375 Pheasant Run, Newtown, PA 18940-3423, United States
| | - Stephen C Cosenza
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - D R C Venkata Subbaiah
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - E Vijaya Bharathi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - Venkat R Pallela
- Department of Medicinal Chemistry, Onconova Therapeutics Inc., 375 Pheasant Run, Newtown, PA 18940-3423, United States
| | - Poornima Ramkumar
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - Rinku Jain
- Department of Structural & Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - Aneel K Aggarwal
- Department of Structural & Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States
| | - E Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., New York, NY 10029-6514, United States.
| |
Collapse
|
47
|
Abstract
Rapidly accumulating data indicate that F-box/WD repeat-containing protein 7 (Fbxw7) is one of the most frequently mutated genes in human cancers and regulates a network of crucial oncoproteins. These studies have generated important new insights into tumorigenesis and may soon enable therapies targeting the Fbxw7 pathway. We searched PubMed, Embase, and ISI Web of Science databases (1973-2015, especially recent 5 years) for articles published in the English language using the key words "Fbxw7," "Fbw7," "hCDC4," and "Sel-10," and we reviewed recent developments in the search for Fbxw7. Fbxw7 coordinates the ubiquitin-dependent proteolysis of several critical cellular regulators, thereby controlling essential processes, such as cell cycle, differentiation, and apoptosis. Fbxw7 contains 3 isoforms (Fbxw7α, Fbxw7β, and Fbxw7γ), and they are differently regulated in subtract recognition. Besides those, Fbxw7 activity is controlled at different levels, resulting in specific and tunable regulation of the abundance and activity of its substrates in a variety of human solid tumor types, including glioma malignancy, nasopharyngeal carcinoma, osteosarcoma, melanoma as well as colorectal, lung, breast, gastric, liver, pancreatic, renal, prostate, endometrial, and esophageal cancers. Fbxw7 is strongly associated with tumorigenesis, and the mechanisms and consequences of Fbxw7 deregulation in cancers may soon enable the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Jun Cao
- From the Zhejiang Cancer Research Institute (JC, Z-QL); and Department of Surgical Oncology, Zhejiang Province Cancer Hospital, Zhejiang Cancer Center, Hangzhou, China (JC, M-HG)
| | | | | |
Collapse
|
48
|
Hu ZB, Liao XH, Xu ZY, Yang X, Dong C, Jin AM, Lu H. PLK2 phosphorylates and inhibits enriched TAp73 in human osteosarcoma cells. Cancer Med 2015; 5:74-87. [PMID: 26625870 PMCID: PMC4708894 DOI: 10.1002/cam4.558] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 01/01/2023] Open
Abstract
TAp73, a member of the p53 tumor suppressor family, can substitute for p53 function, especially in p53‐null and p53‐mutant cells. However, TAp73 enrichment and phosphorylation change its transcriptional activity. Previously, we found that the antitumor function of TAp73 was reactivated by dephosphorylation. Polo‐like kinase 2 (PLK2) plays an important role in bone development. Using a biological information database and phosphorylation prediction software, we hypothesized that PLK2 phosphorylates TAp73 and inhibits TAp73 function in osteosarcomas. Actually,we determined that PLK2 physically binds to and phosphorylates TAp73 when TAp73 protein abundance is up‐regulated by cisplatin. PLK2‐phosphorylated TAp73 at residue Ser48 within the TA domain; phosphorylation of TAp73 was abolished by mutating this residue. Moreover, PLK2 inhibition combined with cisplatin treatment in osteosarcoma Saos2 cells up‐regulated p21 and puma mRNA expression to a greater extent than cisplatin treatment alone. Inhibiting PLK2 in TAp73‐enriched Saos2 cells resulted in inhibited cell proliferation, increased apoptosis, G1 phase arrest, and decreased cell invasion. However, these changes did not occur in TAp73 knockdown Saos2 cells. In conclusion, these findings reveal a novel PLK2 function in the phosphorylation of TAp73, which prevents TAp73 activity in osteosarcoma cells. Thereby, this research provides an insight into the clinical treatment of malignant tumors overexpressing TAp73.
Collapse
Affiliation(s)
- Zheng Bo Hu
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Xiao Hong Liao
- The State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510280, China
| | - Zun Ying Xu
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Xiao Yang
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Chao Dong
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - An Min Jin
- Department of Orthopedics, Zhu Jiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Hai Lu
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Academy of Orthopedics, Guangzhou, Guangdong, 510665, China
| |
Collapse
|
49
|
Xu W, Taranets L, Popov N. Regulating Fbw7 on the road to cancer. Semin Cancer Biol 2015; 36:62-70. [PMID: 26459133 DOI: 10.1016/j.semcancer.2015.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/13/2015] [Indexed: 12/22/2022]
Abstract
The F-box protein Fbw7 targets for degradation critical cellular regulators, thereby controlling essential processes in cellular homeostasis, including cell cycle, differentiation and apoptosis. Most Fbw7 substrates are strongly associated with tumorigenesis and Fbw7 can either suppress or promote tumor development in mouse models. Fbw7 activity is controlled at different levels, resulting in specific and tunable regulation of the abundance and activity of its substrates. Here we highlight recent studies on the role of Fbw7 in controlling tumorigenesis and on the mechanisms that modulate Fbw7 function.
Collapse
Affiliation(s)
- Wenshan Xu
- Department of Radiation Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Versbacher Str. 5, 97078 Würzburg, Germany
| | - Lyudmyla Taranets
- Department of Radiation Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Versbacher Str. 5, 97078 Würzburg, Germany
| | - Nikita Popov
- Department of Radiation Oncology and Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Versbacher Str. 5, 97078 Würzburg, Germany.
| |
Collapse
|
50
|
Zhou Z, He C, Wang J. Regulation mechanism of Fbxw7-related signaling pathways (Review). Oncol Rep 2015; 34:2215-24. [PMID: 26324296 DOI: 10.3892/or.2015.4227] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 05/29/2015] [Indexed: 11/05/2022] Open
Abstract
F-box and WD repeat domain-containing 7 (Fbxw7), the substrate-recognition component of SCFFbxw7 complex, is thought to be a tumor suppressor involved in cell growth, proliferation, differentiation and survival. Although an increasing number of ubiquitin substrates of Fbxw7 have been identified, the best characterized substrates are cyclin E and c-Myc. Fbxw7/cyclin E and Fbxw7/c-Myc pathways are tightly regulated by multiple regulators. Fbxw7 has been identified as a tumor suppressor in hepatocellular carcinoma. This review focused on the regulation of Fbxw7/cyclin E and Fbxw7/c-Myc pathways and discussed findings to gain a better understanding of the role of Fbxw7 in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhenyu Zhou
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Chuanchao He
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jie Wang
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|