1
|
Liu AB, Liu J, Wang S, Ma L, Zhang JF. Biological role and expression of translationally controlled tumor protein (TCTP) in tumorigenesis and development and its potential for targeted tumor therapy. Cancer Cell Int 2024; 24:198. [PMID: 38835077 DOI: 10.1186/s12935-024-03355-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
Translationally controlled tumor protein (TCTP), also known as histamine-releasing factor (HRF) or fortilin, is a highly conserved protein found in various species. To date, multiple studies have demonstrated the crucial role of TCTP in a wide range of cellular pathophysiological processes, including cell proliferation and survival, cell cycle regulation, cell death, as well as cell migration and movement, all of which are major pathogenic mechanisms of tumorigenesis and development. This review aims to provide an in-depth analysis of the functional role of TCTP in tumor initiation and progression, with a particular focus on cell proliferation, cell death, and cell migration. It will highlight the expression and pathological implications of TCTP in various tumor types, summarizing the current prevailing therapeutic strategies that target TCTP.
Collapse
Affiliation(s)
- An-Bu Liu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Jia Liu
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Sheng Wang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Lei Ma
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China.
| | - Jun-Fei Zhang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China.
| |
Collapse
|
2
|
Yu F, Li JL, Feng WR, Tang YK, Su SY, Xu P, Zhong H. Heat Shock Procedure Affects Cell Division-Associated Genes in Gynogenetic Manipulation. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:354-365. [PMID: 35305189 DOI: 10.1007/s10126-022-10112-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/02/2022] [Indexed: 06/14/2023]
Abstract
Heat shock procedure is crucial for gynogenetic manipulation leading to diploidization of the maternal genomes; however, the underlying molecular mechanism especially the transcriptomic changes during this procedure has still not been unveiled yet. Here, the artificial gynogenesis of zebrafish (Danio rerio) using inactivated sperm from rare minnow (Gobiocypris rarus) was conducted. We found that artificial gynogenetic manipulation, including pseudo-fertilization and heat shock, decreased hatching rates, whereas heat shock treatment alone had medium hatching rates. The first cleavage changed the expression of genes associated with RNA transcription and protein synthesis. A co-expression network regulated by hub genes GIT1, Sepsecs, and FLNB was significantly correlated with heat shock procedure. The cyclin family and cyclin-dependent kinase-related genes were lowly expressed in embryos from gynogenetic zebrafish, and genes involved in controlling the cell cycle and genomic stability were significantly altered by the gynogenetic treatment. Our results show the effects of artificial gynogenesis on embryos and describe changes in gene expression that suggest drastic changes take place in cell division by heat shock procedure. These findings will contribute to an understanding of the molecular basis for germplasm improving, including the purifying effect and allogynogenetic biological effect by gynogenesis.
Collapse
Affiliation(s)
- Fan Yu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Jian-Lin Li
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Wen-Rong Feng
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Yong-Kai Tang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Sheng-Yan Su
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Pao Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
| | - Huan Zhong
- Hunan Research Center of Engineering Technology for Utilization of Distinctive Aquatic Resource, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
3
|
Wahner Hendrickson AE, Visscher DW, Hou X, Goergen KM, Atkinson HJ, Beito TG, Negron V, Lingle WL, Bruzek AK, Hurley RM, Wagner JM, Flatten KS, Peterson KL, Schneider PA, Larson MC, Maurer MJ, Kalli KR, Oberg AL, Weroha SJ, Kaufmann SH. CHFR and Paclitaxel Sensitivity of Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13236043. [PMID: 34885153 PMCID: PMC8657201 DOI: 10.3390/cancers13236043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/17/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022] Open
Abstract
The poly(ADP-ribose) binding protein CHFR regulates cellular responses to mitotic stress. The deubiquitinase UBC13, which regulates CHFR levels, has been associated with better overall survival in paclitaxel-treated ovarian cancer. Despite the extensive use of taxanes in the treatment of ovarian cancer, little is known about expression of CHFR itself in this disease. In the present study, tissue microarrays containing ovarian carcinoma samples from 417 women who underwent initial surgical debulking were stained with anti-CHFR antibody and scored in a blinded fashion. CHFR levels, expressed as a modified H-score, were examined for association with histology, grade, time to progression (TTP) and overall survival (OS). In addition, patient-derived xenografts from 69 ovarian carcinoma patients were examined for CHFR expression and sensitivity to paclitaxel monotherapy. In clinical ovarian cancer specimens, CHFR expression was positively associated with serous histology (p = 0.0048), higher grade (p = 0.000014) and higher stage (p = 0.016). After correction for stage and debulking, there was no significant association between CHFR staining and overall survival (p = 0.62) or time to progression (p = 0.91) in patients with high grade serous cancers treated with platinum/taxane chemotherapy (N = 249). Likewise, no association between CHFR expression and paclitaxel sensitivity was observed in ovarian cancer PDXs treated with paclitaxel monotherapy. Accordingly, differences in CHFR expression are unlikely to play a major role in paclitaxel sensitivity of high grade serous ovarian cancer.
Collapse
Affiliation(s)
- Andrea E. Wahner Hendrickson
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA; (X.H.); (J.M.W.); (S.J.W.)
- Correspondence: (A.E.W.H.); (S.H.K.); Tel.: +1-507-284-3731 (A.E.W.H.); +1-507-284-8950 (S.H.K.); Fax: +1-507-293-0107 (A.E.W.H. & S.H.K.)
| | - Daniel W. Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Xiaonan Hou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA; (X.H.); (J.M.W.); (S.J.W.)
| | - Krista M. Goergen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (K.M.G.); (H.J.A.); (M.C.L.); (M.J.M.); (A.L.O.)
| | - Hunter J. Atkinson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (K.M.G.); (H.J.A.); (M.C.L.); (M.J.M.); (A.L.O.)
| | | | - Vivian Negron
- Pathology Research Core, Mayo Clinic, Rochester, MN 55905, USA; (V.N.); (W.L.L.); (A.K.B.)
| | - Wilma L. Lingle
- Pathology Research Core, Mayo Clinic, Rochester, MN 55905, USA; (V.N.); (W.L.L.); (A.K.B.)
| | - Amy K. Bruzek
- Pathology Research Core, Mayo Clinic, Rochester, MN 55905, USA; (V.N.); (W.L.L.); (A.K.B.)
| | - Rachel M. Hurley
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA;
| | - Jill M. Wagner
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA; (X.H.); (J.M.W.); (S.J.W.)
| | - Karen S. Flatten
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA; (K.S.F.); (K.L.P.); (P.A.S.)
| | - Kevin L. Peterson
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA; (K.S.F.); (K.L.P.); (P.A.S.)
| | - Paula A. Schneider
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA; (K.S.F.); (K.L.P.); (P.A.S.)
| | - Melissa C. Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (K.M.G.); (H.J.A.); (M.C.L.); (M.J.M.); (A.L.O.)
| | - Matthew J. Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (K.M.G.); (H.J.A.); (M.C.L.); (M.J.M.); (A.L.O.)
| | | | - Ann L. Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; (K.M.G.); (H.J.A.); (M.C.L.); (M.J.M.); (A.L.O.)
| | - S. John Weroha
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA; (X.H.); (J.M.W.); (S.J.W.)
| | - Scott H. Kaufmann
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA;
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA; (K.S.F.); (K.L.P.); (P.A.S.)
- Correspondence: (A.E.W.H.); (S.H.K.); Tel.: +1-507-284-3731 (A.E.W.H.); +1-507-284-8950 (S.H.K.); Fax: +1-507-293-0107 (A.E.W.H. & S.H.K.)
| |
Collapse
|
4
|
Zhang D, Xu XL, Li F, Sun HC, Cui YQ, Liu S, Xu PY. Upregulation of the checkpoint protein CHFR is associated with tumor suppression in pancreatic cancers. Oncol Lett 2018; 14:8042-8050. [PMID: 29344247 PMCID: PMC5755226 DOI: 10.3892/ol.2017.7239] [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: 03/28/2017] [Accepted: 09/27/2017] [Indexed: 12/24/2022] Open
Abstract
The checkpoint with forkhead-associated (FHA) domain and RING-finger (CHFR) protein was identified as a cell cycle checkpoint protein and E3 ubiquitin ligase. In the present study, the potential functions of CHFR in pancreatic cancer were investigated. CHFR expression was measured in five pancreatic cancer cell lines by reverse transcription- quantitative polymerase chain reaction and western blotting. Capan-1 cells stably expressing CHFR were established by lentiviral vector transfection. Cell proliferation was assessed using Cell Counting Kit-8, and cell migration/invasion assay was determined using Transwell assays. Cell cycle and apoptosis induced by gemcitabine or docetaxel were evaluated using flow cytometry. CHFR expression levels were also evaluated in pancreatic ductal adenocarcinoma (PDAC) tumor samples as well as adjacent non-tumor tissues by immunohistochemistry. The significance of CHFR expression was determined, with respect to clinicopathological features and overall survival. Overexpression of CHFR in Capan-1 cells led to a decreased proliferative rate and reduced cell migration and invasion abilities. Results also indicated an increase in G1 phase cells in Capan-1 cells overexpressing CHFR. Docetaxel-induced apoptosis was inhibited in Capan-1 cells with CHFR-overexpression. A reduction in CHFR expression was detected in 51.9% of patients with PDAC, which significantly correlated with later T-stage. The results show CHFR functions as a tumor suppressor in pancreatic cancer, suggests its potential role in controlling the cell cycle of pancreatic cancer cells; however, CHFR overexpression is not a favorable factor in apoptosis induced by docetaxel.
Collapse
Affiliation(s)
- Di Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Xiao-Lan Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Hai-Chen Sun
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Ye-Qing Cui
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Shuang Liu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Ping-Yong Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| |
Collapse
|
5
|
Castellano L, Dabrowska A, Pellegrino L, Ottaviani S, Cathcart P, Frampton AE, Krell J, Stebbing J. Sustained expression of miR-26a promotes chromosomal instability and tumorigenesis through regulation of CHFR. Nucleic Acids Res 2017; 45:4401-4412. [PMID: 28126920 PMCID: PMC5416844 DOI: 10.1093/nar/gkx022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 12/14/2016] [Accepted: 01/06/2017] [Indexed: 12/29/2022] Open
Abstract
MicroRNA 26a (miR-26a) reduces cell viability in several cancers, indicating that miR-26a could be used as a therapeutic option in patients. We demonstrate that miR-26a not only inhibits G1-S cell cycle transition and promotes apoptosis, as previously described, but also regulates multiple cell cycle checkpoints. We show that sustained miR-26a over-expression in both breast cancer (BC) cell lines and mouse embryonic fibroblasts (MEFs) induces oversized cells containing either a single-large nucleus or two nuclei, indicating defects in mitosis and cytokinesis. Additionally, we demonstrate that miR-26a induces aneuploidy and centrosome defects and enhances tumorigenesis. Mechanistically, it acts by targeting G1-S transition genes as well as genes involved in mitosis and cytokinesis such as CHFR, LARP1 and YWHAE. Importantly, we show that only the re-expression of CHFR in miR-26a over-expressing cells partially rescues normal mitosis and impairs the tumorigenesis exerted by miR-26a, indicating that CHFR represents an important miR-26a target in the regulation of such phenotypes. We propose that miR-26a delivery might not be a viable therapeutic strategy due to the potential deleterious oncogenic activity of this miRNA.
Collapse
Affiliation(s)
- Leandro Castellano
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| | - Aleksandra Dabrowska
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| | - Loredana Pellegrino
- Division of Cancer Therapeutic, The Institute of Cancer Research (ICR), Sutton, London SM2 5NG, UK
| | - Silvia Ottaviani
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| | - Paul Cathcart
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| | - Adam E. Frampton
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| | - Jonathan Krell
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
6
|
Feringa FM, Krenning L, Koch A, van den Berg J, van den Broek B, Jalink K, Medema RH. Hypersensitivity to DNA damage in antephase as a safeguard for genome stability. Nat Commun 2016; 7:12618. [PMID: 27561326 PMCID: PMC5007458 DOI: 10.1038/ncomms12618] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/18/2016] [Indexed: 12/25/2022] Open
Abstract
Activation of the DNA-damage response can lead to the induction of an arrest at various stages in the cell cycle. These arrests are reversible in nature, unless the damage is too excessive. Here we find that checkpoint reversibility is lost in cells that are in very late G2, but not yet fully committed to enter mitosis (antephase). We show that antephase cells exit the cell cycle and enter senescence at levels of DNA damage that induce a reversible arrest in early G2. We show that checkpoint reversibility critically depends on the presence of the APC/C inhibitor Emi1, which is degraded just before mitosis. Importantly, ablation of the cell cycle withdrawal mechanism in antephase promotes cell division in the presence of broken chromosomes. Thus, our data uncover a novel, but irreversible, DNA-damage response in antephase that is required to prevent the propagation of DNA damage during cell division.
Collapse
Affiliation(s)
- Femke M Feringa
- Division of Cell Biology I and Cancer Genomics Center, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Lenno Krenning
- Division of Cell Biology I and Cancer Genomics Center, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands.,Hubrecht Institute, The Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht 3584CT, The Netherlands
| | - André Koch
- Division of Cell Biology I and Cancer Genomics Center, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Jeroen van den Berg
- Division of Cell Biology I and Cancer Genomics Center, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Bram van den Broek
- Division of Cell Biology I and Cancer Genomics Center, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Kees Jalink
- Division of Cell Biology I and Cancer Genomics Center, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - René H Medema
- Division of Cell Biology I and Cancer Genomics Center, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| |
Collapse
|
7
|
Brodie SA, Li G, Harvey D, Khuri FR, Vertino PM, Brandes JC. Small molecule inhibition of the CHFR-PARP1 interaction as novel approach to overcome intrinsic taxane resistance in cancer. Oncotarget 2016; 6:30773-86. [PMID: 26356822 PMCID: PMC4741567 DOI: 10.18632/oncotarget.5040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 08/20/2015] [Indexed: 01/24/2023] Open
Abstract
The mitotic checkpoint protein CHFR has emerged as a major mediator of taxane resistance in cancer. Here we show that CHFR's PAR-binding zinc finger domain (PBZ) mediates a protein interaction with poly-ADP ribosylated PARP1 leading to stabilization of CHFR. Disruption of the CHFR-PARP1 interaction through either PARP1 shRNA-mediated knockdown or overexpression of a PBZ domain peptide induces loss of CHFR protein expression. In an attempt to exploit this observation therapeutically, and to develop compounds with synthetic lethality in combination with taxanes, we performed a high-throughput computational screen of 5,256,508 chemical structures against the published crystal structure of the CHFR PBZ domain to identify candidate small molecule CHFR protein-protein interaction inhibitors. The 10 compounds with the best docking scores (< −9.7) were used for further in vitro testing. One lead compound in particular, termed ‘A3’, completely disrupted the protein-protein interaction between CHFR and PARP1, resulting in the inhibition of mitotic checkpoint function, and led to therapeutic synergy with docetaxel in cell viability and colony formation assays. In mouse xenografts, i.p. administration of ‘A3’ led to a significant reduction in nuclear CHFR protein expression with a maximal effect 4 hours after administration, confirming relevant pharmacodynamics following the peak of ‘A3’ plasma concentration in vivo. Furthermore, combination of A3 and taxane led to significant reduction of implanted tumor size without increase in hematological, hepatic or renal toxicity. These findings provide a proof-of-principle that small molecule inhibition of CHFR PBZ domain interaction is a novel potential therapeutic approach to increase the efficacy of taxane-based chemotherapy in cancer.
Collapse
Affiliation(s)
- Seth A Brodie
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Ge Li
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Donald Harvey
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Paula M Vertino
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Johann C Brandes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
8
|
Ma K, Cao B, Guo M. The detective, prognostic, and predictive value of DNA methylation in human esophageal squamous cell carcinoma. Clin Epigenetics 2016; 8:43. [PMID: 27110300 PMCID: PMC4840959 DOI: 10.1186/s13148-016-0210-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/14/2016] [Indexed: 12/17/2022] Open
Abstract
Esophageal cancer is one of the most common malignancies in the world. Squamous cell carcinoma accounts for approximately 90 % of esophageal cancer cases. Genetic and epigenetic changes have been found to accumulate during the development of various cancers, including esophageal squamous carcinoma (ESCC). Tobacco smoking and alcohol consumption are two major risk factors for ESCC, and both tobacco and alcohol were found to induce methylation changes in ESCC. Growing evidence demonstrates that aberrant epigenetic changes play important roles in the multiple-step processes of carcinogenesis and tumor progression. DNA methylation may occur in the key components of cancer-related signaling pathways. Aberrant DNA methylation affects genes involved in cell cycle, DNA damage repair, Wnt, TGF-β, and NF-κB signaling pathways, including P16, MGMT, SFRP2, DACH1, and ZNF382. Certain genes methylated in precursor lesions of the esophagus demonstrate that DNA methylation may serve as esophageal cancer early detection marker, such as methylation of HIN1, TFPI-2, DACH1, and SOX17. CHFR methylation is a late stage event in ESCC and is a sensitive marker for taxanes in human ESCC. FHIT methylation is associated with poor prognosis in ESCC. Aberrant DNA methylation changes may serve as diagnostic, prognostic, and chemo-sensitive markers. Characterization of the DNA methylome in ESCC will help to better understand its mechanisms and develop improved therapies.
Collapse
Affiliation(s)
- Kai Ma
- />Department of Thoracic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Baoping Cao
- />Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853 China
| | - Mingzhou Guo
- />Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853 China
| |
Collapse
|
9
|
Derks S, Cleven AHG, Melotte V, Smits KM, Brandes JC, Azad N, van Criekinge W, de Bruïne AP, Herman JG, van Engeland M. Emerging evidence for CHFR as a cancer biomarker: from tumor biology to precision medicine. Cancer Metastasis Rev 2015; 33:161-71. [PMID: 24375389 PMCID: PMC3988518 DOI: 10.1007/s10555-013-9462-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Novel insights in the biology of cancer have switched the paradigm of a “one-size-fits-all” cancer treatment to an individualized biology-driven treatment approach. In recent years, a diversity of biomarkers and targeted therapies has been discovered. Although these examples accentuate the promise of personalized cancer treatment, for most cancers and cancer subgroups no biomarkers and effective targeted therapy are available. The great majority of patients still receive unselected standard therapies with no use of their individual molecular characteristics. Better knowledge about the underlying tumor biology will lead the way toward personalized cancer treatment. In this review, we summarize the evidence for a promising cancer biomarker: checkpoint with forkhead and ring finger domains (CHFR). CHFR is a mitotic checkpoint and tumor suppressor gene, which is inactivated in a diverse group of solid malignancies, mostly by promoter CpG island methylation. CHFR inactivation has shown to be an indicator of poor prognosis and sensitivity to taxane-based chemotherapy. Here we summarize the current knowledge of altered CHFR expression in cancer, the impact on tumor biology and implications for personalized cancer treatment.
Collapse
Affiliation(s)
- Sarah Derks
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Arjen H. G. Cleven
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| | - Veerle Melotte
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| | - Kim M. Smits
- Department of Radiation Oncology (MAASTRO Clinic), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Johann C. Brandes
- Department of Hematology and Oncology, Atlanta VA Medical Center Winship Cancer Institute, Emory University, Atlanta, GA USA
| | - Nilofer Azad
- Department of Gastrointestinal Oncology, The Sidney Kimmel Comprehensive Cancer Center at the Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Wim van Criekinge
- Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
- MDxHealth, Irvine, CA USA
| | - Adriaan P. de Bruïne
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| | - James G. Herman
- Department of Tumor Biology, The Sidney Kimmel Comprehensive Cancer Center at the Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Manon van Engeland
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
10
|
Song A, Ye J, Zhang K, Yu H, Gao Y, Wang H, Sun L, Xing X, Yang K, Zhao M. Aberrant expression of the CHFR prophase checkpoint gene in human B-cell non-Hodgkin lymphoma. Leuk Res 2015; 39:536-43. [PMID: 25798877 DOI: 10.1016/j.leukres.2015.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 01/22/2015] [Accepted: 02/15/2015] [Indexed: 10/23/2022]
Abstract
Checkpoint with FHA and Ring Finger (CHFR) is a checkpoint protein that reportedly initiates a cell cycle delay in response to microtubule stress during prophase in mitosis, which has become an interesting target for understanding cancer pathogenesis. Recently, aberrant methylation of the CHFR gene associated with gene silencing has been reported in several cancers. In the present study, we examined the expression of CHFR in B-cell non-Hodgkin lymphoma (B-NHL) in vitro and in vivo. Our results showed that the expression level of CHFR mRNA and protein was reduced in B-NHL tissue samples and B cell lines. Furthermore, CHFR methylation was detected in 39 of 122 B-NHL patients, which was not found in noncancerous reactive hyperplasia of lymph node (RH) tissues. CHFR methylation correlated with the reduced expression of CHFR, high International Prognostic Index (IPI) scores and later pathologic Ann Arbor stages of B-NHL. Treatment with demethylation reagent, 5-Aza-dC, could eliminate the hypermethylation of CHFR, enhance CHFR expression and cell apoptosis and inhibit the cell proliferation of Raji cells, which could be induced by high expression of CHFR in Raji cells. Our results indicated that aberrant methylation of CHFR may be associated with the pathogenesis, progression for B-NHL, which might be a novel molecular marker as prognosis and treatment for B-NHL.
Collapse
Affiliation(s)
- Aiqin Song
- Department of Pediatric Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China.
| | - Junli Ye
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, Shangdong 266021, China
| | - Kunpeng Zhang
- Department of Pediatric Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Hongsheng Yu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Yanhua Gao
- Department of Hematology, Qingdao Women and Children's Medical Care Center, Qingdao, 266011, China
| | - Hongfang Wang
- Department of Pediatric Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Lirong Sun
- Department of Pediatric Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Xiaoming Xing
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shangdong 266003, China
| | - Kun Yang
- Center Laboratory, Affiliated Hospital of Qingdao University, Qingdao, Shangdong 266003, China
| | - Min Zhao
- Department of Pediatric Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| |
Collapse
|
11
|
Porcelli L, Guida G, Quatrale AE, Cocco T, Sidella L, Maida I, Iacobazzi RM, Ferretta A, Stolfa DA, Strippoli S, Guida S, Tommasi S, Guida M, Azzariti A. Aurora kinase B inhibition reduces the proliferation of metastatic melanoma cells and enhances the response to chemotherapy. J Transl Med 2015; 13:26. [PMID: 25623468 PMCID: PMC4314759 DOI: 10.1186/s12967-015-0385-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/08/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The poor response to chemotherapy and the brief response to vemurafenib in metastatic melanoma patients, make the identification of new therapeutic approaches an urgent need. Interestingly the increased expression and activity of the Aurora kinase B during melanoma progression suggests it as a promising therapeutic target. METHODS The efficacy of the Aurora B kinase inhibitor barasertib-HQPA was evaluated in BRAF mutated cells, sensitive and made resistant to vemurafenib after chronic exposure to the drug, and in BRAF wild type cells. The drug effectiveness has been evaluated as cell growth inhibition, cell cycle progression and cell migration. In addition, cellular effectors of drug resistance and response were investigated. RESULTS The characterization of the effectors responsible for the resistance to vemurafenib evidenced the increased expression of MITF or the activation of Erk1/2 and p-38 kinases in the newly established cell lines with a phenotype resistant to vemurafenib. The sensitivity of cells to barasertib-HQPA was irrespective of BRAF mutational status. Barasertib-HQPA induced the mitotic catastrophe, ultimately causing apoptosis and necrosis of cells, inhibited cell migration and strongly affected the glycolytic metabolism of cells inducing the release of lactate. In association i) with vemurafenib the gain in effectiveness was found only in BRAF(V600K) cells while ii) with nab-paclitaxel, the combination was more effective than each drug alone in all cells. CONCLUSIONS These findings suggest barasertib as a new therapeutic agent and as enhancer of chemotherapy in metastatic melanoma treatment.
Collapse
Affiliation(s)
- Letizia Porcelli
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Gabriella Guida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Anna E Quatrale
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Tiziana Cocco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Letizia Sidella
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Immacolata Maida
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Rosa M Iacobazzi
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Anna Ferretta
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Diana A Stolfa
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Sabino Strippoli
- Medical Oncology Department, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Stefania Guida
- Unit of Dermatology and Venereology, University of Bari, P.zza Giulio Cesare, 70124, Bari, Italy.
| | - Stefania Tommasi
- Molecular Genetics Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Michele Guida
- Medical Oncology Department, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| | - Amalia Azzariti
- Clinical and Preclinical Pharmacology Laboratory, National Cancer Research Centre Istituto Tumori Giovanni Paolo II, Viale O. Flacco,65, 70124, Bari, Italy.
| |
Collapse
|
12
|
Involvement of telomerase reverse transcriptase in heterochromatin maintenance. Mol Cell Biol 2014; 34:1576-93. [PMID: 24550003 DOI: 10.1128/mcb.00093-14] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In the fission yeast Schizosaccharomyces pombe, centromeric heterochromatin is maintained by an RNA-directed RNA polymerase complex (RDRC) and the RNA-induced transcriptional silencing (RITS) complex in a manner that depends on the generation of short interfering RNA. In association with the telomerase RNA component (TERC), the telomerase reverse transcriptase (TERT) forms telomerase and counteracts telomere attrition, and without TERC, TERT has been implicated in the regulation of heterochromatin at locations distinct from telomeres. Here, we describe a complex composed of human TERT (hTERT), Brahma-related gene 1 (BRG1), and nucleostemin (NS) that contributes to heterochromatin maintenance at centromeres and transposons. This complex produced double-stranded RNAs homologous to centromeric alpha-satellite (alphoid) repeat elements and transposons that were processed into small interfering RNAs targeted to these heterochromatic regions. These small interfering RNAs promoted heterochromatin assembly and mitotic progression in a manner dependent on the RNA interference machinery. These observations implicate the hTERT/BRG1/NS (TBN) complex in heterochromatin assembly at particular sites in the mammalian genome.
Collapse
|
13
|
Chopra A, Anderson A, Giardina C. Novel piperazine-based compounds inhibit microtubule dynamics and sensitize colon cancer cells to tumor necrosis factor-induced apoptosis. J Biol Chem 2013; 289:2978-91. [PMID: 24338023 DOI: 10.1074/jbc.m113.499319] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We recently identified a series of mitotically acting piperazine-based compounds that potently increase the sensitivity of colon cancer cells to apoptotic ligands. Here we describe a structure-activity relationship study on this compound class and identify a highly active derivative ((4-(3-chlorophenyl)piperazin-1-yl)(2-ethoxyphenyl)methanone), referred to as AK301, the activity of which is governed by the positioning of functional groups on the phenyl and benzoyl rings. AK301 induced mitotic arrest in HT29 human colon cancer cells with an ED50 of ≈115 nm. Although AK301 inhibited growth of normal lung fibroblast cells, mitotic arrest was more pronounced in the colon cancer cells (50% versus 10%). Cells arrested by AK301 showed the formation of multiple microtubule organizing centers with Aurora kinase A and γ-tubulin. Employing in vitro and in vivo assays, tubulin polymerization was found to be slowed (but not abolished) by AK301. In silico molecular docking suggests that AK301 binds to the colchicine-binding domain on β-tubulin, but in a novel orientation. Cells arrested by AK301 expressed elevated levels of TNFR1 on their surface and more readily activated caspases-8, -9, and -3 in the presence of TNF. Relative to other microtubule destabilizers, AK301 was the most active TNF-sensitizing agent and also stimulated Fas- and TRAIL-induced apoptosis. In summary, we report a new class of mitosis-targeting agents that effectively sensitizes cancer cells to apoptotic ligands. These compounds should help illuminate the role of microtubules in regulating apoptotic ligand sensitivity and may ultimately be useful for developing agents that augment the anti-cancer activities of the immune response.
Collapse
|
14
|
Shinde SR, Gangula NR, Kavela S, Pandey V, Maddika S. TOPK and PTEN participate in CHFR mediated mitotic checkpoint. Cell Signal 2013; 25:2511-7. [PMID: 24012691 PMCID: PMC3819987 DOI: 10.1016/j.cellsig.2013.08.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 08/24/2013] [Indexed: 01/15/2023]
Abstract
Mitotic progression is regulated by co-ordinated action of several proteins and is crucial for the maintenance of genomic stability. CHFR (Check point protein with FHA and RING domains) is an E3 ubiquitin ligase and a checkpoint protein that regulates entry into mitosis. But the molecular players involved in CHFR mediated mitotic checkpoint are not completely understood. In this study, we identified TOPK/PBK, a serine/threonine kinase and PTEN, a lipid phosphatase to play an important role in CHFR mediated mitotic transitions. We demonstrated that CHFR ubiquitinates and regulates TOPK levels, which is essential for its checkpoint function. Moreover, TOPK phosphorylates and inactivates PTEN, which in turn activates Akt that leads to proper G2/M progression. Collectively, our results reveal TOPK and PTEN as new players in CHFR mediated mitotic checkpoint. TOPK is identified as a novel CHFR associated protein. TOPK is a substrate of CHFR. TOPK participates in CHFR mediated mitotic stress check point. PTEN is phosphorylated by TOPK and is required for mitotic entry.
Collapse
Affiliation(s)
- Swapnil R Shinde
- Laboratory of Cell Death & Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad 500001, India
| | | | | | | | | |
Collapse
|
15
|
Pillai RN, Brodie SA, Sica GL, Shaojin Y, Li G, Nickleach DC, Yuan L, Varma VA, Bonta D, Herman JG, Brock MV, Ribeiro MJA, Ramalingam SS, Owonikoko TK, Khuri FR, Brandes JC. CHFR protein expression predicts outcomes to taxane-based first line therapy in metastatic NSCLC. Clin Cancer Res 2013; 19:1603-11. [PMID: 23386692 DOI: 10.1158/1078-0432.ccr-12-2995] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Currently, there is no clinically validated test for the prediction of response to tubulin-targeting agents in non-small cell lung cancer (NSCLC). Here, we investigated the significance of nuclear expression of the mitotic checkpoint gene checkpoint with forkhead and ringfinger domains (CHFR) as predictor of response and overall survival with taxane-based first-line chemotherapy in advanced stage NSCLC. METHODS We studied a cohort of 41 patients (median age 63 years) with advanced NSCLC treated at the Atlanta VAMC between 1999 and 2010. CHFR expression by immunohistochemistry (score 0-4) was correlated with clinical outcome using chi-square test and Cox proportional models. A cutoff score of "3" was determined by receiver operator characteristics analysis for "low" CHFR expression. Results were validated in an additional 20 patients who received taxane-based chemotherapy at Emory University Hospital and the Atlanta VAMC. RESULTS High expression (score = 4) of CHFR is strongly associated with adverse outcomes: the risk for progressive disease after first-line chemotherapy with carboplatin-paclitaxel was 52% in patients with CHFR-high versus only 19% in those with CHFR-low tumors (P = 0.033). Median overall survival was strongly correlated with CHFR expression status (CHFR low: 9.9 months; CHFR high: 6.2 months; P = 0.002). After multivariate adjustment, reduced CHFR expression remained a powerful predictor of improved overall survival (HR = 0.24; 95% CI, 0.1-0.58%; P = 0.002). In the validation set, low CHFR expression was associated with higher likelihood of clinical benefit (P = 0.03) and improved overall survival (P = 0.038). CONCLUSIONS CHFR expression is a novel predictive marker of response and overall survival in NSCLC patients treated with taxane-containing chemotherapy.
Collapse
Affiliation(s)
- Rathi N Pillai
- Department of Hematology and Oncology, Emory University, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Burrows AC, Prokop J, Summers MK. Skp1-Cul1-F-box ubiquitin ligase (SCF(βTrCP))-mediated destruction of the ubiquitin-specific protease USP37 during G2-phase promotes mitotic entry. J Biol Chem 2012; 287:39021-9. [PMID: 23027877 PMCID: PMC3493943 DOI: 10.1074/jbc.m112.390328] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 09/25/2012] [Indexed: 12/26/2022] Open
Abstract
Ubiquitin-mediated proteolysis is a key regulatory process in cell cycle progression. The Skp1-Cul1-F-box (SCF) and anaphase-promoting complex (APC) ubiquitin ligases target numerous components of the cell cycle machinery for destruction. Throughout the cell cycle, these ligases cooperate to maintain precise levels of key regulatory proteins, and indirectly, each other. Recently, we have identified the deubiquitinase USP37 as a regulator of the cell cycle. USP37 expression is cell cycle-regulated, being expressed in late G(1) and ubiquitinated by APC(Cdh1) in early G(1). Here we report that in addition to destruction at G(1), a major fraction of USP37 is degraded at the G(2)/M transition, prior to APC substrates and similar to SCF(βTrCP) substrates. Consistent with this hypothesis, USP37 interacts with components of the SCF in a βTrCP-dependent manner. Interaction with βTrCP and subsequent degradation is phosphorylation-dependent and is mediated by the Polo-like kinase (Plk1). USP37 is stabilized in G(2) by depletion of βTrCP as well as chemical or genetic manipulation of Plk1. Similarly, mutation of the phospho-sites abolishes βTrCP binding and renders USP37 resistant to Plk1 activity. Expression of this mutant hinders the G(2)/M transition. Our data demonstrate that tight regulation of USP37 levels is required for proper cell cycle progression.
Collapse
Affiliation(s)
- Amy C. Burrows
- From the Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - John Prokop
- From the Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Matthew K. Summers
- From the Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
17
|
Wang Y, Li WZ, Johnson AE, Luo ZQ, Sun XL, Feoktistova A, McDonald WH, McLeod I, Yates JR, Gould KL, McCollum D, Jin QW. Dnt1 acts as a mitotic inhibitor of the spindle checkpoint protein dma1 in fission yeast. Mol Biol Cell 2012; 23:3348-56. [PMID: 22809626 PMCID: PMC3431938 DOI: 10.1091/mbc.e11-12-1020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The interaction between Dma1 and Dnt1 in fission yeast is characterized. The results show that, similar to its homologue Chfr in higher eukaryotes, Dma1 in fission yeast can also affect factors required for microtubule nucleation and spindle formation at early mitosis. The Schizosaccharomyces pombe checkpoint protein Dma1 couples mitotic progression with cytokinesis and is important in delaying mitotic exit and cytokinesis when kinetochores are not properly attached to the mitotic spindle. Dma1 is a ubiquitin ligase and potential functional relative of the human tumor suppressor Chfr. Dma1 delays mitotic exit and cytokinesis by ubiquitinating a scaffold protein (Sid4) of the septation initiation network, which, in turn, antagonizes the ability of the Polo-like kinase Plo1 to promote cell division. Here we identify Dnt1 as a Dma1-binding protein. Several lines of evidence indicate that Dnt1 inhibits Dma1 function during metaphase. First, Dnt1 interacts preferentially with Dma1 during metaphase. Second, Dma1 ubiquitin ligase activity and Sid4 ubiquitination are elevated in dnt1∆ cells. Third, the enhanced mitotic defects in dnt1Δ plo1 double mutants are partially rescued by deletion of dma1+, suggesting that the defects in dnt1∆ plo1 double mutants are attributable to excess Dma1 activity. Taken together, these data show that Dnt1 acts to restrain Dma1 activity in early mitosis to allow normal mitotic progression.
Collapse
Affiliation(s)
- Yamei Wang
- School of Life Sciences, Xiamen University, Xiamen 361005, Fujian, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Sanbhnani S, Yeong FM. CHFR: a key checkpoint component implicated in a wide range of cancers. Cell Mol Life Sci 2012; 69:1669-87. [PMID: 22159584 PMCID: PMC11114665 DOI: 10.1007/s00018-011-0892-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Revised: 11/13/2011] [Accepted: 11/16/2011] [Indexed: 02/06/2023]
Abstract
CHFR (Checkpoint with Forkhead-associated and RING finger domains) has been implicated in a checkpoint regulating entry into mitosis. However, the details underlying its roles and regulation are unclear due to conflicting lines of evidence supporting different notions of its functions. We provide here an overview of how CHFR is thought to contribute towards regulating mitotic entry and present possible explanations for contradictory observations published on the functions and regulation of CHFR. Furthermore, we survey key data showing correlations between promoter hypermethylation or down-regulation of CHFR and cancers, with a view on the likely reasons why different extents of correlations have been reported. Lastly, we explore the possibilities of exploiting CHFR promoter hypermethylation status in diagnostics and therapeutics for cancer patients. With keen interest currently focused on the association between hypermethylation of CHFR and cancers, details of how CHFR functions require further study to reveal how its absence might possibly contribute to tumorigenesis.
Collapse
Affiliation(s)
- Sheru Sanbhnani
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore
| | | |
Collapse
|
19
|
Expression level of the mitotic checkpoint protein and G2-M cell cycle regulators and prognosis in gastrointestinal stromal tumors in the stomach. Virchows Arch 2011; 460:163-9. [PMID: 22190007 DOI: 10.1007/s00428-011-1181-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 10/26/2011] [Accepted: 11/29/2011] [Indexed: 02/01/2023]
Abstract
The biological behavior of gastrointestinal stromal tumors (GISTs) ranges from benign to malignant, and the risk of an adverse outcome is correlated with the location of the primary tumor, tumor size, and mitotic counts. Cell cycle regulators are potentially associated with the tumorigenesis and progression of GISTs. Checkpoint with forkhead and ring finger (CHFR) functions as an important checkpoint protein in the early to mid-prophase to regulate mitosis. In this study, we evaluated the expression of CHFR and several cell cycle regulators, including cyclin A, cyclin B1, cdc2, and cdk2, by immunohistochemical staining in 53 cases of primary gastric GISTs, and compared the immunohistochemical results with the clinicopathological factors or the GIST risk grades as modified by Miettinen et al. Of the 53 cases, 18 (34%) showed decreased nuclear CHFR expression. Decreased CHFR expression was correlated with higher mitotic counts [>5/50 high-power fields (HPFs)] (p = 0.039) and a high-risk grade (p = 0.0475), but not with expression of other cell cycle regulators. Higher cyclin A labeling index (LI, >1.5%), cyclin B1 LI (>0.25%), cdc2 LI (>1.16%), Ki-67 LI (>4.9%), mitotic counts (>5/50 HPF) and high-risk grade were each associated with shorter disease-free survival (p = 0.0017, p = 0.003, p = 0.0471, p = 0.002, p < 0.001, and p = 0.0017, respectively). Our results suggest that modified risk grade and increased expression of G2-M regulators such as cyclin A, cyclin B1, and cdc2 are useful for predicting the biological behavior of gastric GISTs. In addition, decreased CHFR expression may play a role in increased proliferative activity of higher grade GISTs.
Collapse
|
20
|
Kim JS, Park YY, Park SY, Cho H, Kang D, Cho H. The auto-ubiquitylation of E3 ubiquitin-protein ligase Chfr at G2 phase is required for accumulation of polo-like kinase 1 and mitotic entry in mammalian cells. J Biol Chem 2011; 286:30615-30623. [PMID: 21768102 PMCID: PMC3162422 DOI: 10.1074/jbc.m111.231803] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 06/29/2011] [Indexed: 01/03/2023] Open
Abstract
The E3 ubiquitin-protein ligase Chfr is a mitotic stress checkpoint protein that delays mitotic entry in response to microtubule damage; however, the molecular mechanism by which Chfr accomplishes this remains elusive. Here, we show that Chfr levels are elevated in response to microtubule-damaging stress. Moreover, G(2)/M transition is associated with cell cycle-dependent turnover of Chfr accompanied by high autoubiquitylation activity, suggesting that regulation of Chfr levels and auto-ubiquitylation activity are functionally significant. To test this, we generated Chfr mutants Chfr-K2A and Chfr-K5A in which putative lysine target sites of auto-ubiquitylation were replaced with alanine. Chfr-K2A did not undergo cell cycle-dependent degradation, and its levels remained high during G(2)/M phase. The elevated levels of Chfr-K2A caused a significant reduction in phosphohistone H3 levels and cyclinB1/Cdk1 kinase activities, leading to mitotic entry delay. Notably, polo-like kinase 1 levels at G(2) phase, but not at S phase, were ∼2-3-fold lower in cells expressing Chfr-K2A than in wild-type Chfr-expressing cells. Consistent with this, ubiquitylation of Plk1 at G(2) phase was accelerated in Chfr-K2A-expressing cells. In contrast, Aurora A levels remained constant, indicating that Plk1 is a major target of Chfr in controlling the timing of mitotic entry. Indeed, overexpression of Plk1 in Chfr-K2A-expressing cells restored cyclin B1/Cdk1 kinase activity and promoted mitotic entry. Collectively, these data indicate that Chfr auto-ubiquitylation is required to allow Plk1 to accumulate to levels necessary for activation of cyclin B1/Cdk1 kinase and mitotic entry. Our results provide the first evidence that Chfr auto-ubiquitylation and degradation are important for the G(2)/M transition.
Collapse
Affiliation(s)
- Jo-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Suwon 443-721 Korea; Graduate School of Molecular Science and Technology, Ajou University, Suwon 443-721 Korea
| | - Yong-Yea Park
- Department of Biochemistry, Ajou University School of Medicine, Suwon 443-721 Korea; Graduate School of Molecular Science and Technology, Ajou University, Suwon 443-721 Korea
| | - Sun-Yi Park
- Department of Biochemistry, Ajou University School of Medicine, Suwon 443-721 Korea; Graduate School of Molecular Science and Technology, Ajou University, Suwon 443-721 Korea
| | - Hyeseon Cho
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Health, Bethesda, Maryland 20892
| | - Dongmin Kang
- Department of Life Science, Division of Life and Pharmaceutical Sciences, and the Center for Cell Signaling and Drug Discovery Research, Ewha Womans University, Seoul 120-750, Korea
| | - Hyeseong Cho
- Department of Biochemistry, Ajou University School of Medicine, Suwon 443-721 Korea; Graduate School of Molecular Science and Technology, Ajou University, Suwon 443-721 Korea.
| |
Collapse
|
21
|
CHFR: A Novel Mitotic Checkpoint Protein and Regulator of Tumorigenesis. Transl Oncol 2011; 1:57-64. [PMID: 18633460 DOI: 10.1593/tlo.08109] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 04/02/2008] [Accepted: 05/12/2008] [Indexed: 12/30/2022] Open
Abstract
Checkpoint with FHA and RING finger domains (CHFR) was first recognized as an early mitotic checkpoint protein that delayed the cell cycle in response to microtubule-targeting drugs. It is an E3 ubiquitin ligase that ubiquitinates target proteins to direct them to the proteasome for degradation or to alter their activity. To date, however, the downstream target proteins critical to CHFR's normal cellular functions largely remain unidentified with the exception of the key mitosis regulators, and oncogenes, PLK1 and Aurora A kinases. Rapidly growing evidence in mice, primary human tumors, and mammalian cell culture models indicate that CHFR may also function as a potent tumor suppressor. Interestingly, studies reported to date suggest that CHFR both controls a novel prophase checkpoint early in mitosis and regulates chromosome segregation later in mitosis to maintain genomic stability. In addition, loss of CHFR sensitizes cancer cells to microtubule poisons, altering chemoresponsiveness to taxanes and making it a potential biomarker for chemotherapeutic response. Importantly, CHFR may be one of the few proteins that are required for regulating the cell cycle and maintaining genomic instability to inhibit tumorigenesis.
Collapse
|
22
|
Protected from the inside: Endogenous histone deacetylase inhibitors and the road to cancer. Biochim Biophys Acta Rev Cancer 2011; 1815:241-52. [DOI: 10.1016/j.bbcan.2011.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 01/16/2011] [Accepted: 01/19/2011] [Indexed: 02/06/2023]
|
23
|
Aurora B kinase inhibitor AZD1152: determinants of action and ability to enhance chemotherapeutics effectiveness in pancreatic and colon cancer. Br J Cancer 2011; 104:769-80. [PMID: 21304529 PMCID: PMC3048212 DOI: 10.1038/bjc.2011.21] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background: AZD1152, the prodrug for AZD1152-hydroxyquinazoline pyrazol anilide (HQPA), is a selective inhibitor of Aurora B kinase activity. Preclinical evaluation of AZD1152 has been reported in several human cancer models. The potentiality of this compound in combination therapy warrants further investigation in solid tumours. Experimental design: This study explored the effects of AZD1152-HQPA in colon and pancreatic tumour cells. The antitumour properties of AZD1152, either as single agent or in combination with chemotherapeutics, were evaluated in each study model. The efficacy and the toxicity of AZD1152 alone and in combination with gemcitabine were validated in pancreatic tumour xenograft model. Results: AZD1152-HQPA treatment resulted in a dramatic increase of chromosome number, modification of cell cycle and induction of apoptosis. The most effective combination was that with chemotherapeutics given soon after AZD1152 in both tumour cell types. The effectiveness of the sequential schedule of AZD1152 with gemcitabine was confirmed in nude mice bearing MiaPaCa-2 tumours, showing inhibition of tumour volumes and delaying of tumour growth after the interruption of the treatments. Conclusion: Here we show that AZD1152-HQPA enhances oxaliplatin and gemcitabine effectiveness in colon and pancreatic cancer, respectively. First, we provide advances into administration schedules and dosing regimens for the combination treatment in in vivo pancreatic tumour.
Collapse
|
24
|
Castiel A, Danieli MM, David A, Moshkovitz S, Aplan PD, Kirsch IR, Brandeis M, Krämer A, Izraeli S. The Stil protein regulates centrosome integrity and mitosis through suppression of Chfr. J Cell Sci 2011; 124:532-9. [PMID: 21245198 DOI: 10.1242/jcs.079731] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Stil (Sil, SCL/TAL1 interrupting locus) is a cytosolic and centrosomal protein expressed in proliferating cells that is required for mouse and zebrafish neural development and is mutated in familial microcephaly. Recently the Drosophila melanogaster ortholog of Stil was found to be important for centriole duplication. Consistent with this finding, we report here that mouse embryonic fibroblasts lacking Stil are characterized by slow growth, low mitotic index and absence of clear centrosomes. We hypothesized that Stil regulates mitosis through the tumor suppressor Chfr, an E3 ligase that blocks mitotic entry in response to mitotic stress. Mouse fibroblasts lacking Stil by genomic or RNA interference approaches, as well as E9.5 Stil(-/-) embryos, express high levels of the Chfr protein and reduced levels of the Chfr substrate Plk1. Exogenous expression of Stil, knockdown of Chfr or overexpression of Plk1 reverse the abnormal mitotic phenotypes of fibroblasts lacking Stil. We further demonstrate that Stil increases Chfr auto-ubiquitination and reduces its protein stability. Thus, Stil is required for centrosome organization, entry into mitosis and cell proliferation, and these functions are at least partially mediated by Chfr and its targets. This is the first identification of a negative regulator of the Chfr mitotic checkpoint.
Collapse
Affiliation(s)
- Asher Castiel
- Sheba Cancer Research Center and Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Ramat Gan, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gavet O, Pines J. Activation of cyclin B1-Cdk1 synchronizes events in the nucleus and the cytoplasm at mitosis. ACTA ACUST UNITED AC 2010; 189:247-59. [PMID: 20404109 PMCID: PMC2856909 DOI: 10.1083/jcb.200909144] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The cyclin B-Cdk1 kinase triggers mitosis in most eukaryotes. In animal cells, cyclin B shuttles between the nucleus and cytoplasm in interphase before rapidly accumulating in the nucleus at prophase, which promotes disassembly of the nuclear lamina and nuclear envelope breakdown (NEBD). What triggers the nuclear accumulation of cyclin B1 is presently unclear, although the prevailing view is that the Plk1 kinase inhibits its nuclear export. In this study, we use a biosensor specific for cyclin B1-Cdk1 activity to show that activating cyclin B1-Cdk1 immediately triggers its rapid accumulation in the nucleus through a 40-fold increase in nuclear import that remains dependent on Cdk1 activity until NEBD. Nevertheless, a substantial proportion of cyclin B1-Cdk1 remains in the cytoplasm. The increase in nuclear import is driven by changes in the nuclear import machinery that require neither Plk1 nor inhibition of nuclear export. Thus, the intrinsic link between cyclin B1-Cdk1 activation and its rapid nuclear import inherently coordinates the reorganization of the nucleus and the cytoplasm at mitotic entry.
Collapse
Affiliation(s)
- Olivier Gavet
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, England, UK.
| | | |
Collapse
|
26
|
Abstract
Maintenance of genomic stability is needed for cells to survive many rounds of division throughout their lifetime. Key to the proper inheritance of intact genome is the tight temporal and spatial coordination of cell cycle events. Moreover, checkpoints are present that function to monitor the proper execution of cell cycle processes. For instance, the DNA damage and spindle assembly checkpoints ensure genomic integrity by delaying cell cycle progression in the presence of DNA or spindle damage, respectively. A checkpoint that has recently been gaining attention is the antephase checkpoint that acts to prevent cells from entering mitosis in response to a range of stress agents. We review here what is known about the pathway that monitors the status of the cells at the brink of entry into mitosis when cells are exposed to insults that threaten the proper inheritance of chromosomes. We highlight issues which are unresolved in terms of our understanding of the antephase checkpoint and provide some perspectives on what lies ahead in the understanding of how the checkpoint functions.
Collapse
|
27
|
SAEKI T, OUCHI M, OUCHI T. Physiological and oncogenic Aurora-A pathway. Int J Biol Sci 2009; 5:758-62. [PMID: 20011137 PMCID: PMC2793309 DOI: 10.7150/ijbs.5.758] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 11/24/2009] [Indexed: 01/03/2023] Open
Abstract
Aurora family of protein kinases have emerged as crucial factors of, not only mitosis and cytokinesis, but also human carcinogenesis. Among these family members is Aurora-A that is frequently overexpressed in varieties of human cancer. Both in vitro and in vivo studies demonstrated that Aurora-A induces tumorigenesis through genome instability. These studies have further shown that cell signaling cross-talk between Aurora-A and other cellular proteins are essential for fully-transformed phenotypes. This review summarizes recent progress of Aurora-A-associated carcinogenesis.
Collapse
Affiliation(s)
- Toshiaki SAEKI
- 1. Department of Breast Oncology, Saitama Medical School, Saitama, JAPAN
| | - Mutsuko OUCHI
- 2. NUHS, Systems Biology Program, Pritzker School of Medicine, University of Chicago, Evanston, IL 60201, USA
| | - Toru OUCHI
- 2. NUHS, Systems Biology Program, Pritzker School of Medicine, University of Chicago, Evanston, IL 60201, USA
| |
Collapse
|
28
|
Fu Z, Regan K, Zhang L, Muders MH, Thibodeau SN, French A, Wu Y, Kaufmann SH, Lingle WL, Chen J, Tindall DJ. Deficiencies in Chfr and Mlh1 synergistically enhance tumor susceptibility in mice. J Clin Invest 2009; 119:2714-24. [PMID: 19690386 DOI: 10.1172/jci37405] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 06/24/2009] [Indexed: 01/17/2023] Open
Abstract
Genetic instability, which leads to an accumulation of various genetic abnormalities, has been considered an essential component of the human neoplasic transformation process. However, the molecular basis of genomic instability during tumorigenesis remains incompletely understood. Growing evidence indicates that checkpoint with forkhead and ring finger domains (CHFR), a recently identified mitotic checkpoint protein, plays an important role in maintaining chromosome integrity and functions as a tumor suppressor. In this study, we used high-throughput technology to conduct gene expression profiling of human colon cancers and found that loss of CHFR expression frequently occurred in colon cancers with high microsatellite instability (MSI-H). Downregulation of CHFR expression was closely associated with overexpression of Aurora A, an important mitotic kinase. Mice with deficiencies in both Chfr and Mlh1 (the gene that encodes the DNA mismatch-repair protein Mlh1) displayed dramatically higher incidence of spontaneous tumors relative to mice deficient for only one of these genes. These results suggest that defects in both Chfr and Mlh1 synergistically increase predisposition to tumorigenesis.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Urology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kitzen JJEM, de Jonge MJA, Verweij J. Aurora kinase inhibitors. Crit Rev Oncol Hematol 2009; 73:99-110. [PMID: 19369091 DOI: 10.1016/j.critrevonc.2009.03.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2008] [Revised: 03/15/2009] [Accepted: 03/18/2009] [Indexed: 02/02/2023] Open
Abstract
Most human cancer cells are characterized by changes in the amount or organization of DNA resulting in chromosome instability and aneuploidy. Several mitotic kinases, Aurora kinases amongst others, regulate the progression of the cell through mitosis. So far three Aurora kinases have been identified in man: Aurora-A, Aurora-B and Aurora-C. Aurora kinases were recently identified as a potential target in anticancer therapy, and various Aurora-A and Aurora-B kinase inhibitors are in development. In this review we provide a brief insight into the mechanism of action as far as currently available. We review the available pre-clinical data, discuss the clinical phase I data and try to give a direction for future headings.
Collapse
Affiliation(s)
- J J E M Kitzen
- Department of Medical Oncology, Erasmus University Medical Center, EA Rotterdam, The Netherlands
| | | | | |
Collapse
|
30
|
Maddika S, Sy SMH, Chen J. Functional interaction between Chfr and Kif22 controls genomic stability. J Biol Chem 2009; 284:12998-3003. [PMID: 19321445 DOI: 10.1074/jbc.m900333200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Proper activation of checkpoint during mitotic stress is an important mechanism to prevent genomic instability. Chfr (Check point protein with FHA (Forkhead-associated domain) and RING domains) is a ubiquitin-protein isopeptide ligase (E3) that is important for the control of an early mitotic checkpoint, which delays entry into metaphase in response to mitotic stress. Because several lines of evidence indicate that Chfr is a potential tumor suppressor, it is critically important for us to identify Chfr substrates and understand how Chfr may regulate these substrates, control mitotic transitions, and thus, act as a tumor suppressor in vivo. Here, we report the discovery of a new Chfr-associated protein Kif22, a chromokinesin that binds to both DNA and microtubules. We demonstrated that Kif22 is a novel substrate of Chfr. We showed that Chfr-mediated Kif22 down-regulation is critical for the maintenance of chromosome stability. Collectively, our results reveal a new substrate of Chfr that plays a role in the maintenance of genome integrity.
Collapse
Affiliation(s)
- Subbareddy Maddika
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | |
Collapse
|
31
|
Brooks L, Heimsath EG, Loring GL, Brenner C. FHA-RING ubiquitin ligases in cell division cycle control. Cell Mol Life Sci 2008; 65:3458-66. [PMID: 18597043 PMCID: PMC2588411 DOI: 10.1007/s00018-008-8220-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Despite the common occurrence of forkhead associated (FHA) phosphopeptide-binding domains and really interesting new gene (RING) E3 ubiquitin ligase domains, gene products containing both an N-terminal FHA domain and C-terminal RING domain constitute a highly distinctive intersection. Characterized FHA-RING ligases include the two vertebrate proteins, Checkpoint with FHA and RING (Chfr) and RING finger 8 (Rnf8), as well as three fungal proteins, Defective in mitosis (Dma1), Chf1 and Chf2. These FHA-RING ligases play roles in negative regulation of the cell division cycle, apparently by coupling protein phosphorylation events to specific ubiquitylation of target proteins. Here, the available data on upstream and downstream regulation of and by FHA-RING ligases are reviewed.
Collapse
Affiliation(s)
- L. Brooks
- Departments of Genetics and Biochemistry and Norris Cotton Cancer Center, Dartmouth Medical School, Rubin 733–HB7937, Lebanon, NH 03756 USA
| | - E. G. Heimsath
- Departments of Genetics and Biochemistry and Norris Cotton Cancer Center, Dartmouth Medical School, Rubin 733–HB7937, Lebanon, NH 03756 USA
| | | | - C. Brenner
- Departments of Genetics and Biochemistry and Norris Cotton Cancer Center, Dartmouth Medical School, Rubin 733–HB7937, Lebanon, NH 03756 USA
| |
Collapse
|
32
|
Loss of CHFR in human mammary epithelial cells causes genomic instability by disrupting the mitotic spindle assembly checkpoint. Neoplasia 2008; 10:643-52. [PMID: 18592005 DOI: 10.1593/neo.08176] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Revised: 04/23/2008] [Accepted: 04/24/2008] [Indexed: 01/01/2023] Open
Abstract
CHFR is an E3 ubiquitin ligase and an early mitotic checkpoint protein implicated in many cancers and in the maintenance of genomic stability. To analyze the role of CHFR in genomic stability, by siRNA, we decreased its expression in genomically stable MCF10A cells. Lowered CHFR expression quickly led to increased aneuploidy due to many mitotic defects. First, we confirmed that CHFR interacts with the mitotic kinase Aurora A to regulate its expression. Furthermore, we found that decreased CHFR led to disorganized multipolar mitotic spindles. This was supported by the finding that CHFR interacts with alpha-tubulin and can regulate its ubiquitination in response to nocodazole and the amount of acetylated alpha-tubulin, a component of the mitotic spindle. Finally, we found a novel CHFR interacting protein, the spindle checkpoint protein MAD2. Decreased CHFR expression resulted in the mislocalization of both MAD2 and BUBR1 during mitosis and impaired MAD2/CDC20 complex formation. Further evidence of a compromised spindle checkpoint was the presence of misaligned metaphase chromosomes, lagging anaphase chromosomes, and defective cytokinesis in CHFR knockdown cells. Importantly, our results suggest a novel role for CHFR regulating chromosome segregation where decreased expression, as seen in cancer cells, contributes to genomic instability by impairing the spindle assembly checkpoint.
Collapse
|
33
|
Takeshita M, Koga T, Takayama K, Kouso H, Nishimura-Ikeda Y, Yoshino I, Maehara Y, Nakanishi Y, Sueishi K. CHFR expression is preferentially impaired in smoking-related squamous cell carcinoma of the lung, and the diminished expression significantly harms outcomes. Int J Cancer 2008; 123:1623-30. [PMID: 18623126 DOI: 10.1002/ijc.23673] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Loss of tumor suppressors and activation of oncogenes lead to carcinogenesis. Abnormal expression of CHFR, a novel checkpoint gene, or of Aurora kinases, key regulators of mitosis, has been detected in a variety of solid tumors. Recently, CHFR has been revealed to ensure chromosomal stability by controlling the expression level of Aurora-A in vitro. However, the frequency of aberrant expression of these proteins and the association with clinicopathologic parameters remain poorly defined in nonsmall-cell lung cancer (NSCLC). In this study, we investigated the immunohistochemical protein expression of CHFR and Aurora-A in 157 NSCLC cases and evaluated the association between clinicopathologic parameters statistically. The relationship between CHFR protein and mRNA levels and the association between this relationship and promoter methylation of the CHFR gene were also examined in 20 frozen sections of NSCLC. Overexpression of Aurora-A and reduced expression of CHFR were found in 94 cases (59.8%) and 62 cases (39%) of NSCLC, respectively, and those were significantly correlated with tumor differentiation and size. Moreover, diminished CHFR expression was significantly associated with smoking-related squamous cell carcinoma cases and poor prognosis. Multivariate analysis revealed that CHFR expression was an independent prognostic factor. A statistical correlation was evident between CHFR protein and mRNA expression. In conclusion, our results suggest the aberrant expression of Aurora-A and/or of CHFR contributed to the increase in the malignant potential of NSCLC. We also revealed that CHFR expression was predominantly impaired in smoking-related squamous cell carcinoma and might be a useful prognostic marker in NSCLC.
Collapse
Affiliation(s)
- Masafumi Takeshita
- Division of Pathophysiological and Experimental Pathology, Department of Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bae JB, Mukhopadhyay SS, Liu L, Zhang N, Tan J, Akhter S, Liu X, Shen X, Li L, Legerski RJ. Snm1B/Apollo mediates replication fork collapse and S Phase checkpoint activation in response to DNA interstrand cross-links. Oncogene 2008; 27:5045-56. [PMID: 18469862 PMCID: PMC2805112 DOI: 10.1038/onc.2008.139] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 03/10/2008] [Accepted: 03/31/2008] [Indexed: 12/13/2022]
Abstract
The removal of DNA interstrand cross-links (ICLs) has proven to be notoriously complicated due to the involvement of multiple pathways of DNA repair, which include the Fanconi anemia/BRCA pathway, homologous recombination and components of the nucleotide excision and mismatch repair pathways. Members of the SNM1 gene family have also been shown to have a role in mediating cellular resistance to ICLs, although their precise function has remained elusive. Here, we show that knockdown of Snm1B/Apollo in human cells results in hypersensitivity to mitomycin C (MMC), but not to IR. We also show that Snm1B-deficient cells exhibit a defective S phase checkpoint in response to MMC, but not to IR, and this finding may account for the specific sensitivity to the cross-linking drug. Interestingly, although previous studies have largely implicated ATR as the major kinase activated in response to ICLs, we show that it is activation of the ATM-mediated checkpoint that is defective in Snm1B-deficient cells. The requirement for Snm1B in ATM checkpoint activation specifically after ICL damage is correlated with its role in promoting double-strand break formation, and thus replication fork collapse. Consistent with this result Snm1B was found to interact directly with Mus81-Eme1, an endonuclease previously implicated in fork collapse. In addition, we also show that Snm1B interacts with the Mre11-Rad50-Nbs1 (MRN) complex and with FancD2 further substantiating its role as a checkpoint/DNA repair protein.
Collapse
Affiliation(s)
- Jae-Bum Bae
- Department of Cancer Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Sudit S. Mukhopadhyay
- Department of Cancer Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Lingling Liu
- Department of Cancer Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Nianxiang Zhang
- Department of Cancer Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Jeff Tan
- Department of Cancer Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Shamimi Akhter
- Department of Cancer Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Xiaojun Liu
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Xi Shen
- Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Lei Li
- Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Randy J. Legerski
- Department of Cancer Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
35
|
Fukuda T, Kondo Y, Nakagama H. The anti-proliferative effects of the CHFR depend on the forkhead associated domain, but not E3 ligase activity mediated by ring finger domain. PLoS One 2008; 3:e1776. [PMID: 18335050 PMCID: PMC2258000 DOI: 10.1371/journal.pone.0001776] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 02/12/2008] [Indexed: 12/11/2022] Open
Abstract
The CHFR protein comprises fork head associated- (FHA) and RING-finger (RF) domain and is frequently downregulated in human colon and gastric cancers up to 50%. The loss of CHFR mRNA expression is a consequence of promoter methylation, suggesting a tumor suppressor role for this gene in gastrointestinal carcinogenesis. In terms of the biological functions of CHFR, it has been shown to activate cell cycle checkpoint when cells are treated with microtubule depolymerizing agents. Furthermore, CHFR was reported to have E3 ligase activity and promote ubiquitination and degradation of oncogenic proteins such as Aurora A and polo-like kinase 1. However, molecular pathways involved in the tumor suppressive function of CHFR are not yet clear since the two established roles of this protein are likely to inhibit cell growth. In this study, we have identified that the FHA domain of CHFR protein is critical for growth suppressive properties, whereas the RF and cysteine rich domains (Cys) are not required for this function. In contrast, the RF and Cys domains are essential for E3 ligase activity of CHFR. By the use of a cell cycle checkpoint assay, we also confirmed that the FHA domain of CHFR plays an important role in initiating a cell cycle arrest at G2/M, indicating a functional link exists between the anti-proliferative effects and checkpoint function of this tumor suppressor protein via this domain. Collectively, our data show that the checkpoint function of the FHA domain of CHFR is a core component of anti-proliferative properties against the gastrointestinal carcinogenesis.
Collapse
Affiliation(s)
- Tomokazu Fukuda
- Biochemistry Division, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasuyuki Kondo
- Biochemistry Division, National Cancer Center Research Institute, Tokyo, Japan
| | - Hitoshi Nakagama
- Biochemistry Division, National Cancer Center Research Institute, Tokyo, Japan
- * E-mail:
| |
Collapse
|
36
|
Li JP, Yang JL. Cyclin B1 proteolysis via p38 MAPK signaling participates in G2 checkpoint elicited by arsenite. J Cell Physiol 2007; 212:481-8. [PMID: 17373649 DOI: 10.1002/jcp.21042] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Timely induction of cyclin B1 controls mitotic entry, whereas its proteolysis is essential for mitotic exit. By contrast, cyclin B1 transcription is repressed during G(2) arrest induced by DNA damage. The p38 mitogen-activated protein kinase is involved in the G(2) checkpoint; yet, its impact on cyclin B1 protein levels remains unclear. Here we show that untimely proteolysis of cyclin B1 following p38 activation contributes to G(2) checkpoint. Exposing early G(2) cells to arsenite impeded cyclin B1 protein accumulation, Cdk1 activation, and G(2)-to-M progression. Conversely, cyclin B1 was non-degradable in late G(2) and mitotic cells after arsenite. Cyclin B1 proteolysis was enhanced by arsenite in early G(2) and asynchronous cells. This rapid destruction of cyclin B1 was mediated via the ubiquitin-proteasome pathway probably in a Cdc20 and Cdh1 independent mechanism. Under arsenite, inhibition of p38 activation or depletion of p38alpha suppressed cyclin B1 ubiquitination and proteolysis, while forced expression of MKK6-p38 accelerated these events. Inactivation of p38 in arsenite-treated early G(2) cells allowed G(2)-to-M progression, blocked apoptosis, increased cell viability, and decreased micronucleus formation. Thus, p38 signaling pathway triggering cyclin B1 proteolysis after arsenite may play an important role in connecting G(2) arrest with apoptosis or genome instability.
Collapse
Affiliation(s)
- Ju-Pi Li
- Molecular Carcinogenesis Laboratory, Institute of Biotechnology and Department of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | | |
Collapse
|
37
|
Privette LM, González ME, Ding L, Kleer CG, Petty EM. Altered expression of the early mitotic checkpoint protein, CHFR, in breast cancers: implications for tumor suppression. Cancer Res 2007; 67:6064-74. [PMID: 17596595 DOI: 10.1158/0008-5472.can-06-4109] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Checkpoint with FHA and Ring Finger (CHFR) is hypothesized to mediate a delay in cell cycle progression early in mitosis in response to microtubule stress, independent of the spindle assembly checkpoint. As a potential regulator of cell cycle progression, CHFR naturally becomes an interesting target for understanding cancer cells. In recent years, there has been increasing evidence supporting the role of CHFR as a tumor suppressor, most of which report loss of expression, occasionally due to promoter hypermethylation, in cancers compared with patient-matched normal tissues. We studied both a panel of breast cancer cell lines as well as primary tissue samples from breast cancer patients to investigate CHFR as a relevant tumor suppressor in breast cancer and to determine whether CHFR expression was associated with clinical and pathologic variables. We report that 41% of cell lines and 36% of patient samples showed low or negative CHFR protein expression or staining. In addition, lack of CHFR detection was associated with increased tumor size and weakly correlated with estrogen receptor-negative tumors from patients. To study the effects of low CHFR expression in vitro, we stably expressed a short hairpin RNA construct targeting CHFR in two lines of immortalized human mammary epithelial cells. Notably, decreased CHFR expression resulted in the acquisition of many phenotypes associated with malignant progression, including accelerated growth rates, higher mitotic index, enhanced invasiveness, increased motility, greater aneuploidy, and amplified colony formation in soft agar, further supporting the role of CHFR as a tumor suppressor in breast cancer.
Collapse
Affiliation(s)
- Lisa M Privette
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109-0638, USA
| | | | | | | | | |
Collapse
|
38
|
Gao YJ, Xin Y. Advance in the relationship between checkpoint with fork head associated and ring finger gene and carcinomas of digestive tract. Shijie Huaren Xiaohua Zazhi 2007; 15:1745-1749. [DOI: 10.11569/wcjd.v15.i15.1745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
CHFR (checkpoint with fork head associated and ring finger), a novel checkpoint gene, was frequently inactivated in human cancers. In response to mitotic stress, it causes a delay in chromosome condensation during prophase. Studies have showed that the direct target of the CHFR pathway was Plk1. In vitro-translated Plk1 is ubiquitinated, in a CHFR-dependent manner, both in Xenopus interphase extracts as well as in a purified system reconstituted with recombinant proteins. In addition, by excluding Cyclin B1 from the nucleus, regulating Aurora-A level and acting with the P38 stress kinases, CHFR blocks entry to mitosis prophase in mammalian cells. Besides, USP7 can remove ubiquitin moiety from the autoubiquitinated CHFR both in vivo and in vitro, which results in the accumulation of CHFR in the cells. Thus, USP7-mediated deubiquitination of CHFR leads to its accumulation, which might be a key regulatory step for CHFR activation. CHFR expression is frequently silenced by aberrant methylation in the carcinomas of digestive tract. In this article, we reviewed the progress of research on the structure of CHFR gene and effect of CHFR protein as well as its relation to the carcinomas of digestive tract.
Collapse
|
39
|
Gao YJ, Xin Y, Zhang SN, Zhang JH, Wu DY. Protein expression of checkpoint with fork head associated and ring finger and mutant p53 and their clinicopathological significances in gastric cancer. Shijie Huaren Xiaohua Zazhi 2007; 15:1622-1627. [DOI: 10.11569/wcjd.v15.i14.1622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the relationship between the expression of checkpoint with fork head associated and ring finger (CHFR) and P53 protein in gastric cancer (GC) and the clinicopathologic characteristics, and to explore the correlated molecular mechanism of CHFR and p53 genes in gastric carcinogenesis.
METHODS: Five paraffin blocks of tissue microarray were constructed using a Tissue Array Machine (Steve Leighton Beecher Instruments, USA), including 151 cases of primary GC (101 cases with matched normal mucosa, intestinal metaplasia or dysplasia). Envision immunohistochemical method was employed to detect the protein expression of CHFR and mutant p53 in GC and precancerous tissues mentioned above.
RESULTS: The positive rate of CHFR protein expression in GC (49.67%, 75/151) was significantly lower than that in normal gastric mucosa (85.25%, 52/61)(P < 0.05). The down-regulation or absence of mitotic checkpoint CHFR protein expression was correlated with the sex of GC patients. The absent rate of CHFR protein expression in the female GC patients was significantly higher than that in the male GC ones (64% vs 43.56%, P < 0.05). The absent rates was also significantly different between GC patients of Borrmann Ⅲ + Ⅳ and Ⅰ + Ⅱ types (57.14% vs 34.78%, P < 0.05). In the present study, though CHFR protein expression showed no significant difference among various histological types of GC, the absent rate of CHFR protein expression was the highest (71.43%) in signet ring cell carcinoma. The absent expression of CHFR protein was not related to the depth of invasion and lymph node metastasis of GC. In addition, no correlation was found between the expression of CHFR and P53 protein expression in GC (P > 0.05).
CONCLUSION: Down-regulation or absence of mitotic checkpoint CHFR protein expression is frequent events in GC and may take a part in gastric carcinogenesis. Abnormal expression of CHFR may be of more importance in the development of female patients and diffuse-type GC.
Collapse
|
40
|
Ricaud L, Proux C, Renou JP, Pichon O, Fochesato S, Ortet P, Montané MH. ATM-mediated transcriptional and developmental responses to gamma-rays in Arabidopsis. PLoS One 2007; 2:e430. [PMID: 17487278 PMCID: PMC1855986 DOI: 10.1371/journal.pone.0000430] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2006] [Accepted: 04/19/2007] [Indexed: 11/19/2022] Open
Abstract
ATM (Ataxia Telangiectasia Mutated) is an essential checkpoint kinase that signals DNA double-strand breaks in eukaryotes. Its depletion causes meiotic and somatic defects in Arabidopsis and progressive motor impairment accompanied by several cell deficiencies in patients with ataxia telangiectasia (AT). To obtain a comprehensive view of the ATM pathway in plants, we performed a time-course analysis of seedling responses by combining confocal laser scanning microscopy studies of root development and genome-wide expression profiling of wild-type (WT) and homozygous ATM-deficient mutants challenged with a dose of γ-rays (IR) that is sublethal for WT plants. Early morphologic defects in meristematic stem cells indicated that AtATM, an Arabidopsis homolog of the human ATM gene, is essential for maintaining the quiescent center and controlling the differentiation of initial cells after exposure to IR. Results of several microarray experiments performed with whole seedlings and roots up to 5 h post-IR were compiled in a single table, which was used to import gene information and extract gene sets. Sequence and function homology searches; import of spatio-temporal, cell cycling, and mutant-constitutive expression characteristics; and a simplified functional classification system were used to identify novel genes in all functional classes. The hundreds of radiomodulated genes identified were not a random collection, but belonged to functional pathways such as those of the cell cycle; cell death and repair; DNA replication, repair, and recombination; and transcription; translation; and signaling, indicating the strong cell reprogramming and double-strand break abrogation functions of ATM checkpoints. Accordingly, genes in all functional classes were either down or up-regulated concomitantly with downregulation of chromatin deacetylases or upregulation of acetylases and methylases, respectively. Determining the early transcriptional indicators of prolonged S-G2 phases that coincided with cell proliferation delay, or an anticipated subsequent auxin increase, accelerated cell differentiation or death, was used to link IR-regulated hallmark functions and tissue phenotypes after IR. The transcription burst was almost exclusively AtATM-dependent or weakly AtATR-dependent, and followed two major trends of expression in atm: (i)-loss or severe attenuation and delay, and (ii)-inverse and/or stochastic, as well as specific, enabling one to distinguish IR/ATM pathway constituents. Our data provide a large resource for studies on the interaction between plant checkpoints of the cell cycle, development, hormone response, and DNA repair functions, because IR-induced transcriptional changes partially overlap with the response to environmental stress. Putative connections of ATM to stem cell maintenance pathways after IR are also discussed.
Collapse
Affiliation(s)
- Lilian Ricaud
- CEA, DSV, Institut de Biologie Environnementale et de Biotechnologie (iBEB), Service de biologie végétale et de microbiologie environnementales (SBVME), Cadarache, Saint Paul-lez-Durance, France
| | - Caroline Proux
- Unité de Recherche en Génomique Végétale, UMR INRA 1165 - CNRS 8114 - UEVE, Evry, France
| | - Jean-Pierre Renou
- Unité de Recherche en Génomique Végétale, UMR INRA 1165 - CNRS 8114 - UEVE, Evry, France
| | - Olivier Pichon
- Unité de Recherche en Génomique Végétale, UMR INRA 1165 - CNRS 8114 - UEVE, Evry, France
| | - Sylvain Fochesato
- CEA, DSV, Institut de Biologie Environnementale et de Biotechnologie (iBEB), Service de biologie végétale et de microbiologie environnementales (SBVME), Cadarache, Saint Paul-lez-Durance, France
| | - Philippe Ortet
- CEA, DSV, Institut de Biologie Environnementale et de Biotechnologie (iBEB), Service de biologie végétale et de microbiologie environnementales (SBVME), Cadarache, Saint Paul-lez-Durance, France
| | - Marie-Hélène Montané
- CEA, DSV, Institut de Biologie Environnementale et de Biotechnologie (iBEB), Service de biologie végétale et de microbiologie environnementales (SBVME), Cadarache, Saint Paul-lez-Durance, France
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
41
|
Rosell R, Santarpia M, Moran T, Salazar MF, Kaen DL, Ramirez JL. Age-related genetic abnormalities: the Achilles' heel for customizing therapy in elderly lung cancer patients. Per Med 2007; 4:59-72. [PMID: 29793303 DOI: 10.2217/17410541.4.1.59] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aging and cancer are closely related, and DNA repair systems, mainly involving the nucleotide excision repair pathway, have an important caretaker function in both processes. More than half of non-small cell lung cancer patients are elderly, and the expression of some genes involved in the mitotic checkpoint, such as BubR1, declines with aging. Cisplatin-based chemotherapy is the standard treatment for advanced non-small cell lung cancer; if performance status is good, both elderly and younger patients can tolerate this treatment equally well. Customized cisplatin treatment, based on reduction of the nucleotide excision repair pathway function, could be an attractive approach, and the assessment of mitotic checkpoint genes can be used for selecting docetaxel treatment. Epidermal growth factor receptor (EGFR) mutations are particularly frequent in elderly lung cancer patients who are never-smokers and constitute an attractive target for treatment with EGFR tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Rafael Rosell
- Medical Oncology Service, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet, s/n, 08916 Badalona (Barcelona), Spain.
| | - Mariacarmela Santarpia
- Medical Oncology Service, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet, s/n, 08916 Badalona (Barcelona), Spain.
| | - Teresa Moran
- Medical Oncology Service, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet, s/n, 08916 Badalona (Barcelona), Spain.
| | | | - Diego Lucas Kaen
- Medical Oncology Service, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet, s/n, 08916 Badalona (Barcelona), Spain.
| | - Jose Luis Ramirez
- Medical Oncology Service, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet, s/n, 08916 Badalona (Barcelona), Spain.
| |
Collapse
|
42
|
Soncini C, Carpinelli P, Gianellini L, Fancelli D, Vianello P, Rusconi L, Storici P, Zugnoni P, Pesenti E, Croci V, Ceruti R, Giorgini ML, Cappella P, Ballinari D, Sola F, Varasi M, Bravo R, Moll J. PHA-680632, a Novel Aurora Kinase Inhibitor with Potent Antitumoral Activity. Clin Cancer Res 2006; 12:4080-9. [PMID: 16818708 DOI: 10.1158/1078-0432.ccr-05-1964] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Aurora kinases play critical roles during mitosis in chromosome segregation and cell division. The aim of this study was to determine the preclinical profile of a novel, highly selective Aurora kinase inhibitor, PHA-680632, as a candidate for anticancer therapy. EXPERIMENTAL DESIGN The activity of PHA-680632 was assayed in a biochemical ATP competitive kinase assay. A wide panel of cell lines was evaluated for antiproliferative activity. Cell cycle analysis. Immunohistochemistry, Western blotting, and Array Scan were used to follow mechanism of action and biomarker modulation. Specific knockdown of the targets by small interfering RNA was followed to validate the observed phenotypes. Efficacy was determined in different xenograft models and in a transgenic animal model of breast cancer. RESULTS PHA-680632 is active on a wide range of cancer cell lines and shows significant tumor growth inhibition in different animal tumor models at well-tolerated doses. The mechanism of action of PHA-680632 is in agreement with inhibition of Aurora kinases. Histone H3 phosphorylation in Ser10 is mediated by Aurora B kinase, and our kinetic studies on its inhibition by PHA-680632 in vitro and in vivo show that phosphorylation of histone H3 is a good biomarker to follow activity of PHA-680632. CONCLUSIONS PHA-680632 is the first representative of a new class of Aurora inhibitors with a high potential for further development as an anticancer therapeutic. On treatment, different cell lines respond differentially, suggesting the absence of critical cell cycle checkpoints that could be the basis for a favorable therapeutic window.
Collapse
|
43
|
Gutierrez GJ, Ronai Z. Ubiquitin and SUMO systems in the regulation of mitotic checkpoints. Trends Biochem Sci 2006; 31:324-32. [PMID: 16647857 PMCID: PMC3061571 DOI: 10.1016/j.tibs.2006.04.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 03/08/2006] [Accepted: 04/07/2006] [Indexed: 01/09/2023]
Abstract
Proteolysis mediated by the ubiquitin-proteasome system is a crucial regulatory mechanism in signal transduction cascades of temporal cellular processes such as cell division. Two principal subtypes of modular ubiquitin ligase, the anaphase-promoting complex or cyclosome (APC/C) and the Skp1/Cullin-1/F-box protein complex, have emerged as essential regulators of key events in the cell cycle. The importance of these ligases is best illustrated by their roles in the checkpoint and repair pathways or in response to multiple stresses, where they affect activation of the M-phase-promoting factor or proper formation and/or maintenance of the mitotic spindle. Recent studies have considerably improved our understanding of the function of the concerted action of the phosphorylation and ubiquitin or SUMO systems in the regulation of the stability and activity of key components of the mitotic checkpoint.
Collapse
Affiliation(s)
- Gustavo J Gutierrez
- Signal Transduction Program, Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | |
Collapse
|
44
|
Cooper S, Iyer G, Tarquini M, Bissett P. Nocodazole does not synchronize cells: implications for cell-cycle control and whole-culture synchronization. Cell Tissue Res 2006; 324:237-42. [PMID: 16432713 DOI: 10.1007/s00441-005-0118-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 11/03/2005] [Indexed: 10/25/2022]
Abstract
It has been predicted that nocodazole-inhibited cells are not synchronized because nocodazole-arrested cells with a G2-phase amount of DNA would not have a narrow cell-size range reflecting the cell size of some specific, presumably G2-phase, cell-cycle age. Size measurements of nocodazole-inhibited cells now fully confirm this prediction. Further, release from nocodazole inhibition does not produce cells that move through the cell cycle mimicking the passage of normal unperturbed cells through the cell cycle. Nocodazole, an archetypal whole-culture synchronization method, can inhibit growth to produce cells with a G2-phase amount of DNA, but such cells are not synchronized. Cells produced by a selective (i.e., non-whole-culture) method not only have a specific DNA content, but also have a narrow size distribution. The current view of cell-cycle control that is based on methods that are not suitable for cell-cycle analysis must therefore be reconsidered when results are based on whole-culture synchronization.
Collapse
Affiliation(s)
- Stephen Cooper
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109-0620, USA.
| | | | | | | |
Collapse
|
45
|
Baker DJ, Chen J, van Deursen JMA. The mitotic checkpoint in cancer and aging: what have mice taught us? Curr Opin Cell Biol 2005; 17:583-9. [PMID: 16226453 DOI: 10.1016/j.ceb.2005.09.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Accepted: 09/29/2005] [Indexed: 12/11/2022]
Abstract
The spindle assembly checkpoint is a cellular surveillance mechanism that functions to ensure faithful chromosome segregation during mitosis. Failure of this checkpoint can result in aneuploidy, a state of having abnormal numbers of chromosomes. Most human cancers consist of aneuploid cells, but it is unclear if the aneuploidy is a cause or a consequence of tumorigenesis. Over recent years, mouse models for spindle assembly checkpoint failure have been generated to investigate the biological relevance of the different spindle assembly checkpoint genes and the pathologies associated with chromosome number instability. Most of these models exhibit susceptibility to carcinogenesis. Moreover, one model has led to the identification of the spindle checkpoint protein BubR1 as a regulator of the normal aging process.
Collapse
Affiliation(s)
- Darren J Baker
- Department of Pediatric, Mayo Clinic, 200 1st ST SW, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
46
|
Skladanowski A, Côme MG, Sabisz M, Escargueil AE, Larsen AK. Down-Regulation of DNA Topoisomerase IIα Leads to Prolonged Cell Cycle Transit in G2and Early M Phases and Increased Survival to Microtubule-Interacting Agents. Mol Pharmacol 2005; 68:625-34. [PMID: 15942022 DOI: 10.1124/mol.105.013995] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Microtubule binders are cell cycle-specific agents with preferential cytotoxicity toward mitotic cells. We have characterized vincristine-selected human leukemia cells to establish whether development of vincristine resistance was accompanied by changes in cell cycle kinetics and distribution. Our results indicate that vincristine resistance is accompanied by delayed G2 transit and prolonged early mitosis in both the absence and the presence of the microtubule binder nocodazole. The altered G2/M regulation is accompanied by resistance to short-term (12 h) but not continuous nocodazole exposure in agreement with the transient nature of the observed cell cycle alterations. Western blot analysis indicates that vincristine-selection is accompanied by down-regulation of topoisomerase IIalpha without detectable alterations of the other mitotic regulators studied, including Cdk1, p21, 14-3-3sigma, and 14-3-3epsilon. This was associated with at least 7-fold less chromosome-associated topoisomerase IIalpha, decreased catalytic activity, and cross-resistance to topoisomerase II inhibitors. Characterization of isogenic cell lines expressing different levels of topoisomerase II proteins shows that cellular levels of topoisomerase IIalpha, but not the closely related topoisomerase IIbeta, directly influence the cell cycle kinetics in G2 and early mitosis as well as the resistance to nocodazole. These results underline the importance of topoisomerase IIalpha in late G2 and early M phases and provide evidence for an as-yet-unsuspected interaction between topoisomerase II and microtubule-directed agents.
Collapse
Affiliation(s)
- Andrzej Skladanowski
- Group of Molecular and Clinical Cancer Therapeutics, INSERM U673, Hôpital Saint-Antoine, 75571 Paris 12, France
| | | | | | | | | |
Collapse
|