1
|
Marto CM, Laranjo M, Gonçalves AC, Paula A, Jorge J, Caetano-Oliveira R, Sousa MI, Oliveiros B, Ramalho-Santos J, Sarmento-Ribeiro AB, Marques-Ferreira M, Cabrita A, Botelho MF, Carrilho E. In Vitro Characterization of Reversine-Treated Gingival Fibroblasts and Their Safety Evaluation after In Vivo Transplantation. Pharmaceutics 2024; 16:207. [PMID: 38399261 PMCID: PMC10892828 DOI: 10.3390/pharmaceutics16020207] [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: 12/20/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Reversine is a purine derivative that has been investigated with regard to its biological effects, such as its anticancer properties and, mostly, its ability to induce the dedifferentiation of adult cells, increasing their plasticity. The obtained dedifferentiated cells have a high potential for use in regenerative procedures, such as regenerative dentistry (RD). Instead of replacing the lost or damaged oral tissues with synthetic materials, RD uses stem cells combined with matrices and an appropriate microenvironment to achieve tissue regeneration. However, the currently available stem cell sources present limitations, thus restricting the potential of RD. Based on this problem, new sources of stem cells are fundamental. This work aims to characterize mouse gingival fibroblasts (GFs) after dedifferentiation with reversine. Different administration protocols were tested, and the cells obtained were evaluated regarding their cell metabolism, protein and DNA contents, cell cycle changes, morphology, cell death, genotoxicity, and acquisition of stem cell characteristics. Additionally, their teratoma potential was evaluated after in vivo transplantation. Reversine caused toxicity at higher concentrations, with decreased cell metabolic activity and protein content. The cells obtained displayed polyploidy, a cycle arrest in the G2/M phase, and showed an enlarged size. Additionally, apoptosis and genotoxicity were found at higher reversine concentrations. A subpopulation of the GFs possessed stem properties, as supported by the increased expression of CD90, CD105, and TERT, the existence of a CD106+ population, and their trilineage differentiation capacity. The dedifferentiated cells did not induce teratoma formation. The extensive characterization performed shows that significant functional, morphological, and genetic changes occur during the dedifferentiation process. The dedifferentiated cells have some stem-like characteristics, which are of interest for RD.
Collapse
Affiliation(s)
- Carlos Miguel Marto
- Institute of Experimental Pathology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute of Integrated Clinical Practice and Laboratory of Evidence-Based and Precision Dentistry, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal (E.C.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Mafalda Laranjo
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Anabela Paula
- Institute of Integrated Clinical Practice and Laboratory of Evidence-Based and Precision Dentistry, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal (E.C.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Joana Jorge
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rui Caetano-Oliveira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
- Germano de Sousa—Centro de Diagnóstico Histopatológico CEDAP, University of Coimbra, 3000-377 Coimbra, Portugal
| | - Maria Inês Sousa
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Bárbara Oliveiros
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - João Ramalho-Santos
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Manuel Marques-Ferreira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Institute of Endodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - António Cabrita
- Institute of Experimental Pathology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Filomena Botelho
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Eunice Carrilho
- Institute of Integrated Clinical Practice and Laboratory of Evidence-Based and Precision Dentistry, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal (E.C.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| |
Collapse
|
2
|
Soria-Bretones I, Thu KL, Silvester J, Cruickshank J, El Ghamrasni S, Ba-alawi W, Fletcher GC, Kiarash R, Elliott MJ, Chalmers JJ, Elia AC, Cheng A, Rose AAN, Bray MR, Haibe-Kains B, Mak TW, Cescon DW. The spindle assembly checkpoint is a therapeutic vulnerability of CDK4/6 inhibitor-resistant ER + breast cancer with mitotic aberrations. SCIENCE ADVANCES 2022; 8:eabq4293. [PMID: 36070391 PMCID: PMC9451148 DOI: 10.1126/sciadv.abq4293] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
Inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6i) are standard first-line treatments for metastatic ER+ breast cancer. However, acquired resistance to CDK4/6i invariably develops, and the molecular phenotypes and exploitable vulnerabilities associated with resistance are not yet fully characterized. We developed a panel of CDK4/6i-resistant breast cancer cell lines and patient-derived organoids and demonstrate that a subset of resistant models accumulates mitotic segregation errors and micronuclei, displaying increased sensitivity to inhibitors of mitotic checkpoint regulators TTK and Aurora kinase A/B. RB1 loss, a well-recognized mechanism of CDK4/6i resistance, causes such mitotic defects and confers enhanced sensitivity to TTK inhibition. In these models, inhibition of TTK with CFI-402257 induces premature chromosome segregation, leading to excessive mitotic segregation errors, DNA damage, and cell death. These findings nominate the TTK inhibitor CFI-402257 as a therapeutic strategy for a defined subset of ER+ breast cancer patients who develop resistance to CDK4/6i.
Collapse
Affiliation(s)
- Isabel Soria-Bretones
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Segal Cancer Centre and Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Kelsie L. Thu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Sciences, St. Michael’s Hospital , Toronto,, ON, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jennifer Silvester
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Samah El Ghamrasni
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Wail Ba-alawi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Graham C. Fletcher
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Reza Kiarash
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mitchell J. Elliott
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto , ON, Canada
| | - Jordan J. Chalmers
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Andrea C. Elia
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Albert Cheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - April A. N. Rose
- Segal Cancer Centre and Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Mark R. Bray
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Tak W. Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David W. Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto , ON, Canada
| |
Collapse
|
3
|
Non-Canonical Programmed Cell Death in Colon Cancer. Cancers (Basel) 2022; 14:cancers14143309. [PMID: 35884370 PMCID: PMC9320762 DOI: 10.3390/cancers14143309] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/24/2022] [Accepted: 07/05/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Non-canonical PCD is an important player in colon cancer cell suicide. It influences colon cancer in many ways, such as through tumorigenesis, treatment, and prognosis. In this review, we present the mechanism, application, and prospect of different types of non-canonical PCD in colon cancer. Abstract Programmed cell death (PCD) is an evolutionarily conserved process of cell suicide that is regulated by various genes and the interaction of multiple signal pathways. Non-canonical programmed cell death (PCD) represents different signaling excluding apoptosis. Colon cancer is the third most incident and the fourth most mortal worldwide. Multiple factors such as alcohol, obesity, and genetic and epigenetic alternations contribute to the carcinogenesis of colon cancer. In recent years, emerging evidence has suggested that diverse types of non-canonical programmed cell death are involved in the initiation and development of colon cancer, including mitotic catastrophe, ferroptosis, pyroptosis, necroptosis, parthanatos, oxeiptosis, NETosis, PANoptosis, and entosis. In this review, we summarized the association of different types of non-canonical PCD with tumorigenesis, progression, prevention, treatments, and prognosis of colon cancer. In addition, the prospect of drug-resistant colon cancer therapy related to non-canonical PCD, and the interaction between different types of non-canonical PCD, was systemically reviewed.
Collapse
|
4
|
Xia P, Liang J, Jin D, Jin Z. Reversine inhibits proliferation, invasion and migration and induces cell apoptosis in gastric cancer cells by downregulating TTK. Exp Ther Med 2021; 22:929. [PMID: 34306198 PMCID: PMC8281506 DOI: 10.3892/etm.2021.10361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/02/2021] [Indexed: 12/29/2022] Open
Abstract
Reversine (Rev) has been used for the treatment of a number of cancers. However, there have been no previous reports for the use of Rev for gastric cancer (GC). The aim of the present study was to investigate the effect of Rev on cell proliferation, migration, invasion and cell apoptosis in human GC cells and TTK expression. Cell Counting Kit-8 and colony formation were used to assess cell proliferation. Wound healing and Transwell assays were performed to examine cell migration and invasion, respectively. Cell apoptosis was measured using TUNEL staining and western blotting. Reverse transcription-quantitative PCR and western blotting were performed to determine TTK expression in AGS and NCI-N87 GC cells. Rev treatment inhibited the viability of the two GC cells lines in a dose-dependent manner and suppressed their capacities of clone formation, migration and invasion. Rev-treated cells exhibited reduced matrix metalloproteinase (MMP)2/9 expression and increased apoptosis compared with those in control cells. In addition, expression of the anti-apoptotic protein Bcl-2 was significantly decreased, whilst the expression levels of the pro-apoptotic factors Bax and cleaved-caspase-3/9 were increased by Rev treatment compared with that in the control group that were not treated with Rev. In addition, TTK protein expression was decreased in cells treated with Rev compared with that in untreated cells. However, overexpression of TTK significantly reversed the aforementioned effects of Rev in GC cells. These results suggest that Rev may inhibit the proliferation, invasion and migration of GC cells whilst inducing cell apoptosis by suppressing TTK expression. Therefore, Rev may confer potential properties as a therapeutic anti-cancer agent. Additionally, TTK may serve as a molecular target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Pengfei Xia
- Department of Liver-Gallbladder and Gastric Diseases, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| | - Jin Liang
- Department of Liver-Gallbladder and Gastric Diseases, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| | - Di Jin
- Department of Liver-Gallbladder and Gastric Diseases, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| | - Zhanyong Jin
- Department of Liver-Gallbladder and Gastric Diseases, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| |
Collapse
|
5
|
Yan F, Shi F, Li X, Chang H, Jin M, Li Y. Prognostic significance of CEA, Ki67 and p53 in pseudomyxoma peritonei of appendiceal origin. J Int Med Res 2021; 49:3000605211022297. [PMID: 34187207 PMCID: PMC8492452 DOI: 10.1177/03000605211022297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Objective To determine the levels of carcinoembryonic antigen (CEA), proliferating
nuclear antigen Ki67 and p53 in pseudomyxoma peritonei (PMP) of appendiceal
origin and to correlate the levels with clinicopathological characteristics
and overall survival. Methods This retrospective study collected data on clinicopathological features and
immunohistochemical staining of CEA, Ki67 and p53 in patients with PMP of
appendiceal origin. Overall survival was evaluated using Kaplan–Meier plots.
Median survival time was estimated by Log-rank tests. Potential prognostic
factors were evaluated by Cox proportional hazards regression models. Results A total of 141 patients with PMP of appendiceal origin were enrolled in the
study with a median age of 54 years. Of these, 93 (66.0%) were diagnosed
with low-grade mucinous carcinoma, 43 (30.5%) with high-grade mucinous
carcinoma and five (3.5%) with high-grade with signet ring cells. CEA
exhibited ubiquitous immunopositivity in most cases and was not associated
with overall survival. Ki67 labelling index (LI) and p53 status were related
to histological grade and overall survival. The main pathological indicators
affecting survival included histological grade, lymph node involvement,
angiolymphatic invasion, Ki67 LI and p53. Conclusion Combined analysis of high Ki67 LI and aberrant p53 may provide the basis for
evaluating the biological behaviour of PMP and predicting clinical
outcome.
Collapse
Affiliation(s)
- Fengcai Yan
- Department of Pathology, 117968Beijing Shijitan Hospital, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Pathology, 74639Beijing Chaoyang Hospital, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Feng Shi
- Department of Pathology, 117968Beijing Shijitan Hospital, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Pathology, 74639Beijing Chaoyang Hospital, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xinbao Li
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hong Chang
- Department of Pathology, 117968Beijing Shijitan Hospital, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Mulan Jin
- Department of Pathology, 74639Beijing Chaoyang Hospital, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yan Li
- Department of Pathology, 117968Beijing Shijitan Hospital, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Piccoli M, Ghiroldi A, Monasky MM, Cirillo F, Ciconte G, Pappone C, Anastasia L. Reversine: A Synthetic Purine with a Dual Activity as a Cell Dedifferentiating Agent and a Selective Anticancer Drug. Curr Med Chem 2020; 27:3448-3462. [PMID: 30605049 DOI: 10.2174/0929867326666190103120725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/28/2018] [Accepted: 12/28/2018] [Indexed: 12/27/2022]
Abstract
The development of new therapeutic applications for adult and embryonic stem cells has dominated regenerative medicine and tissue engineering for several decades. However, since 2006, induced Pluripotent Stem Cells (iPSCs) have taken center stage in the field, as they promised to overcome several limitations of the other stem cell types. Nonetheless, other promising approaches for adult cell reprogramming have been attempted over the years, even before the generation of iPSCs. In particular, two years before the discovery of iPSCs, the possibility of synthesizing libraries of large organic compounds, as well as the development of high-throughput screenings to quickly test their biological activity, enabled the identification of a 2,6-disubstituted purine, named reversine, which was shown to be able to reprogram adult cells to a progenitor-like state. Since its discovery, the effect of reversine has been confirmed on different cell types, and several studies on its mechanism of action have revealed its central role in inhibitory activity on several kinases implicated in cell cycle regulation and cytokinesis. These key features, together with its chemical nature, suggested a possible use of the molecule as an anti-cancer drug. Remarkably, reversine exhibited potent cytotoxic activity against several tumor cell lines in vitro and a significant effect in decreasing tumor progression and metastatization in vivo. Thus, 15 years since its discovery, this review aims at critically summarizing the current knowledge to clarify the dual role of reversine as a dedifferentiating agent and anti-cancer drug.
Collapse
Affiliation(s)
- Marco Piccoli
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Andrea Ghiroldi
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Michelle M Monasky
- Arrhythmology Department, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Federica Cirillo
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Giuseppe Ciconte
- Arrhythmology Department, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Luigi Anastasia
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, via Luigi Mangiagalli 31, 20133 Milan, Italy
| |
Collapse
|
7
|
Simon Serrano S, Sime W, Abassi Y, Daams R, Massoumi R, Jemaà M. Inhibition of mitotic kinase Mps1 promotes cell death in neuroblastoma. Sci Rep 2020; 10:11997. [PMID: 32686724 PMCID: PMC7371706 DOI: 10.1038/s41598-020-68829-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/29/2020] [Indexed: 12/27/2022] Open
Abstract
Neuroblastoma is the most common paediatric cancer type. Patients diagnosed with high-risk neuroblastoma have poor prognosis and occasionally tumours relapse. As a result, novel treatment strategies are needed for relapse and refractory neuroblastoma patients. Here, we found that high expression of Mps1 kinase (mitotic kinase Monopolar Spindle 1) was associated with relapse-free neuroblastoma patient outcomes and poor overall survival. Silencing and inhibition of Mps1 in neuroblastoma or PDX-derived cells promoted cell apoptosis via the caspase-dependent mitochondrial apoptotic pathway. The mechanism of cell death upon Mps1 inhibition was dependent on the polyploidization/aneuploidization of the cells before undergoing mitotic catastrophe. Furthermore, tumour growth retardation was confirmed in a xenograft mouse model after Mps1-inhibitor treatment. Altogether, these results suggest that Mps1 expression and inhibition can be considered as a novel prognostic marker as well as a therapeutic strategy for the treatment of high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Sonia Simon Serrano
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Wondossen Sime
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Yasmin Abassi
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden.
| | - Mohamed Jemaà
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden.
| |
Collapse
|
8
|
Pathological prognostic factors of pseudomyxoma peritonei: comprehensive clinicopathological analysis of 155 cases. Hum Pathol 2020; 97:9-18. [PMID: 31926211 DOI: 10.1016/j.humpath.2019.12.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 12/29/2019] [Accepted: 12/30/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Pseudomyxoma peritonei (PMP) is an extremely rare malignancy, characterized by extensive peritoneal implantation and colloidal ascites. This study was to explore the pathological prognostic factors of PMP. METHODS Specimens from 155 PMP patients were analyzed by H&E and immunohistochemistry. Parameters included primary tumor location, histological grade, lymph node metastasis, tumor emboli in the blood and lymph vessels, perineural invasion, Ki67 labeling index, p53, mismatch repair (MMR) gene mutations, MUC1, MUC2, MUC5AC, and MUC6. Clinicopathological and follow-up data were subjected to univariate and multivariate analyses. RESULTS The patients included 63.2% (n = 98) low-grade mucinous carcinoma peritonei, 31.6% (n = 49) high-grade mucinous carcinoma peritonei and 5.2% (n = 8) high-grade mucinous carcinoma peritonei with signet ring cells. There were 9.7% (n = 15) with lymph node metastasis; 11.6% (n = 18) with angiolymphatic invasion; 6.3% (n = 8) with defective MMR (dMMR); 35.5% (n = 55) with Ki67 labeling index ≥ 50%; 36.1% (n = 56) with p53 mutation. For PMP from appendiceal origin (n = 140), univariate analysis identified 10 potential prognostic factors. But Multivariate analysis identified only histologic grade was the independent prognostic factor for OS. Mortality risk of high-grade peritoneal mucinous carcinoma or high-grade peritoneal mucinous carcinoma with signet ring cells was 7.056 times (P < .0001, 95% CI: 2.701-18.435) or 27.224 times (P < .0001, 95% CI: 6.207-119.408), respectively, higher than low-grade. CONCLUSIONS For PMP from the appendiceal origin, histological grade could be the only independent prognostic factor.
Collapse
|
9
|
Henriques AC, Ribeiro D, Pedrosa J, Sarmento B, Silva PMA, Bousbaa H. Mitosis inhibitors in anticancer therapy: When blocking the exit becomes a solution. Cancer Lett 2018; 440-441:64-81. [PMID: 30312726 DOI: 10.1016/j.canlet.2018.10.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/12/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Abstract
Current microtubule-targeting agents (MTAs) remain amongst the most important antimitotic drugs used against a broad range of malignancies. By perturbing spindle assembly, MTAs activate the spindle assembly checkpoint (SAC), which induces mitotic arrest and subsequent apoptosis. However, besides toxic side effects and resistance, mitotic slippage and failure in triggering apoptosis in various cancer cells are limiting factors of MTAs efficacy. Alternative strategies to target mitosis without affecting microtubules have, thus, led to the identification of small molecules, such as those that target spindle Kinesins, Aurora and Polo-like kinases. Unfortunately, these so-called second-generation of antimitotics, encompassing mitotic blockers and mitotic drivers, have failed in clinical trials. Our recent understanding regarding the mechanisms of cell death during a mitotic arrest pointed out apoptosis as the main variable, providing an opportunity to control the cell fates and influence the effectiveness of antimitotics. Here, we provide an overview on the second-generation of antimitotics, and discuss possible strategies that exploit SAC activity, mitotic slippage/exit and apoptosis induction, in order to improve the efficacy of anticancer strategies that target mitosis.
Collapse
Affiliation(s)
- Ana C Henriques
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; INEB, Instituto Nacional de Engenharia Biomédica, Universidade Do Porto, Porto, Portugal
| | - Diana Ribeiro
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade Do Porto, Porto, Portugal
| | - Joel Pedrosa
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal
| | - Bruno Sarmento
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; INEB, Instituto Nacional de Engenharia Biomédica, Universidade Do Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação Em Saúde, Universidade Do Porto, Porto, Portugal
| | - Patrícia M A Silva
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal
| | - Hassan Bousbaa
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade Do Porto, Porto, Portugal.
| |
Collapse
|
10
|
Synthesis of 2,6-Diamino-Substituted Purine Derivatives and Evaluation of Cell Cycle Arrest in Breast and Colorectal Cancer Cells. Molecules 2018; 23:molecules23081996. [PMID: 30103421 PMCID: PMC6222518 DOI: 10.3390/molecules23081996] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022] Open
Abstract
Reversine is a potent antitumor 2,6-diamino-substituted purine acting as an Aurora kinases inhibitor and interfering with cancer cell cycle progression. In this study we describe three reversine-related molecules, designed by docking calculation, that present structural modifications in the diamino units at positions 2 and 6. We investigated the conformations of the most stable prototropic tautomers of one of these molecules, the N6-cyclohexyl-N6-methyl-N2-phenyl-7H-purine-2,6-diamine (3), by Density Functional Theory (DFT) calculation in the gas phase, water and chloroform, the last solvent considered to give insights into the detection of broad signals in NMR analysis. In all cases the HN(9) tautomer resulted more stable than the HN(7) form, but the most stable conformations changed in different solvents. Molecules 1–3 were evaluated on MCF-7 breast and HCT116 colorectal cancer cell lines showing that, while being less cytotoxic than reversine, they still caused cell cycle arrest in G2/M phase and polyploidy. Unlike reversine, which produced a pronounced cell cycle arrest in G2/M phase in all the cell lines used, similar concentrations of 1–3 were effective only in cells where p53 was deleted or down-regulated. Therefore, our findings support a potential selective role of these structurally simplified, reversine-related molecules in p53-defective cancer cells.
Collapse
|
11
|
Jemaà M, Abassi Y, Kifagi C, Fezai M, Daams R, Lang F, Massoumi R. Reversine inhibits Colon Carcinoma Cell Migration by Targeting JNK1. Sci Rep 2018; 8:11821. [PMID: 30087398 PMCID: PMC6081478 DOI: 10.1038/s41598-018-30251-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer is one of the most commonly diagnosed cancers and the third most common cause of cancer-related death. Metastasis is the leading reason for the resultant mortality of these patients. Accordingly, development and characterization of novel anti-cancer drugs limiting colorectal tumor cell dissemination and metastasis are needed. In this study, we found that the small molecule Reversine reduces the migration potential of human colon carcinoma cells in vitro. A coupled kinase assay with bio-informatics approach identified the c-Jun N-terminal kinase (JNK) cascade as the main pathway inhibited by Reversine. Knockdown experiments and pharmacological inhibition identified JNK1 but not JNK2, as a downstream effector target in cancer cell migration. Xenograft experiments confirm the effect of JNK inhibition in the metastatic potential of colon cancer cells. These results highlight the impact of individual JNK isoforms in cancer cell metastasis and propose Reversine as a novel anti-cancer molecule for treatment of colon cancer patients.
Collapse
Affiliation(s)
- Mohamed Jemaà
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, 22381, Sweden. .,Department of Physiology I, Tübingen University, Gmelinstr. 5, D-72076, Tübingen, Germany.
| | - Yasmin Abassi
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, 22381, Sweden
| | - Chamseddine Kifagi
- Division of Immunology and Vaccinology, Technical University of Denmark, Copenhagen, Denmark
| | - Myriam Fezai
- Department of Physiology I, Tübingen University, Gmelinstr. 5, D-72076, Tübingen, Germany
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, 22381, Sweden
| | - Florian Lang
- Department of Physiology I, Tübingen University, Gmelinstr. 5, D-72076, Tübingen, Germany. .,Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany.
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, 22381, Sweden.
| |
Collapse
|
12
|
Cheng L, Wang H, Guo K, Wang Z, Zhang Z, Shen C, Chen L, Lin J. Reversine, a substituted purine, exerts an inhibitive effect on human renal carcinoma cells via induction of cell apoptosis and polyploidy. Onco Targets Ther 2018. [PMID: 29520153 PMCID: PMC5833753 DOI: 10.2147/ott.s158198] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Human renal cell carcinoma (RCC) is the most common type of kidney cancer that arises from the renal epithelium. Up to 33.3% of RCC patients treated with local tumor resections will subsequently develop recurrence or metastases. Thus, optimized therapeutic regimes are urgently needed to improve the prognosis of RCC. Reversine was recently reported to exert critical roles in cancer therapy. Materials and methods This study evaluated the anti-tumor effects of reversine on cell viability, colony formation, apoptosis, and cell cycle in 786-O and ACHN cell lines. Results It was demonstrated that reversine significantly inhibited the proliferation of both cell lines in time- and dose-dependent manners. Polyploidy formation was observed under high-concentration reversine treatment. In addition, reversine induced cell death via caspase-dependent apoptotic pathways, which could be partially inhibited by Z-VAD-FMK, a pan-caspase inhibitor. Conclusion Reversine could effectively suppress the proliferation of human RCC cells, and may serve as a novel therapeutic regimen for RCC in clinical practice.
Collapse
Affiliation(s)
- Li Cheng
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Kecun Guo
- Department of Urology, The Second People's Hospital of Liaocheng, Shandong, China
| | - Zicheng Wang
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China
| | - Zhongyuan Zhang
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China
| | - Cheng Shen
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China
| | - Liang Chen
- Medical Center of Reproductive and Genetics, Peking University First Hospital, Beijing, China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China
| |
Collapse
|
13
|
Zhao D, Tahaney WM, Mazumdar A, Savage MI, Brown PH. Molecularly targeted therapies for p53-mutant cancers. Cell Mol Life Sci 2017; 74:4171-4187. [PMID: 28643165 PMCID: PMC5664959 DOI: 10.1007/s00018-017-2575-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/30/2017] [Accepted: 06/15/2017] [Indexed: 02/08/2023]
Abstract
The tumor suppressor p53 is lost or mutated in approximately half of human cancers. Mutant p53 not only loses its anti-tumor transcriptional activity, but also often acquires oncogenic functions to promote tumor proliferation, invasion, and drug resistance. Traditional strategies have been taken to directly target p53 mutants through identifying small molecular compounds to deplete mutant p53, or to restore its tumor suppressive function. Accumulating evidence suggest that cancer cells with mutated p53 often exhibit specific functional dependencies on secondary genes or pathways to survive, providing alternative targets to indirectly treat p53-mutant cancers. Targeting these genes or pathways, critical for survival in the presence of p53 mutations, holds great promise for cancer treatment. In addition, mutant p53 often exhibits novel gain-of-functions to promote tumor growth and metastasis. Here, we review and discuss strategies targeting mutant p53, with focus on targeting the mutant p53 protein directly, and on the progress of identifying genes and pathways required in p53-mutant cells.
Collapse
Affiliation(s)
- Dekuang Zhao
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - William M Tahaney
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - Michelle I Savage
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - Powel H Brown
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
14
|
Sica A, Massarotti M. Myeloid suppressor cells in cancer and autoimmunity. J Autoimmun 2017; 85:117-125. [PMID: 28728794 DOI: 10.1016/j.jaut.2017.07.010] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 12/18/2022]
Abstract
A bottleneck for immunotherapy of cancer is the immunosuppressive microenvironment in which the tumor cells proliferate. Cancers harness the immune regulatory mechanism that prevents autoimmunity from evading immunosurveillance and promoting immune destruction. Regulatory T cells, myeloid suppressor cells, inhibitory cytokines and immune checkpoint receptors are the major components of the immune system acting in concert with cancer cells and causing the subversion of anti-tumor immunity. This redundant immunosuppressive network poses an impediment to efficacious immunotherapy by facilitating tumor progression. Tumor-associated myeloid cells comprise heterogeneous populations acting systemically (myeloid-derived suppressor cells/MDSCs) and/or locally in the tumor microenvironment (MDSCs and tumor-associated macrophages/TAMs). Both populations promote cancer cell proliferation and survival, angiogenesis and lymphangiogenesis and elicit immunosuppression through different pathways, including the expression of immunosuppressive cytokines and checkpoint inhibitors. Several evidences have demonstrated that myeloid cells can express different functional programs in response to different microenvironmental signals, a property defined as functional plasticity. The opposed extremes of this functional flexibility are generally represented by the classical macrophage activation, which identifies inflammatory and cytotoxic M1 polarized macrophages, and the alternative state of macrophage activation, which identifies M2 polarized anti-inflammatory and immunosuppressive macrophages. Functional skewing of myeloid cells occurs in vivo under physiological and pathological conditions, including cancer and autoimmunity. Here we discuss how myeloid suppressor cells can on one hand support tumor growth and, on the other, limit autoimmune responses, indicating that their therapeutic reprogramming can generate opportunities in relieving immunosuppression in the tumor microenvironment or reinstating tolerance in autoimmune conditions.
Collapse
Affiliation(s)
- Antonio Sica
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, CAAD, Novara, Italy.
| | - Marco Massarotti
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Rheumatology, University Hospitals of Morecambe Bay NHS Foundation Trust, Royal Lancaster Infirmary, Ashton Road, LA1 4RP Lancaster, United Kingdom
| |
Collapse
|
15
|
Choi M, Min YH, Pyo J, Lee CW, Jang CY, Kim JE. TC Mps1 12, a novel Mps1 inhibitor, suppresses the growth of hepatocellular carcinoma cells via the accumulation of chromosomal instability. Br J Pharmacol 2017; 174:1810-1825. [PMID: 28299790 DOI: 10.1111/bph.13782] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 03/01/2017] [Accepted: 03/05/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Chromosomal instability is not only a hallmark of cancer but also an attractive therapeutic target. A diverse set of mitotic kinases maintains chromosomal stability. One of these is monopolar spindle 1 (Mps1, also known as TTK), which is essential for chromosome alignment and for the spindle assembly checkpoint (SAC). Pharmacological inhibition of Mps1 has been suggested as a cancer therapeutic; however, despite the existence of a novel Mps1 inhibitor, TC Mps1 12, no such studies have been performed. EXPERIMENTAL APPROACH The effects of TC Mps1 12 on cell viability, chromosome alignment, centrosome number, mitotic duration, apoptosis and SAC were determined in hepatocellular carcinoma (HCC) cells. In addition, the association of Mps1 expression with the overall survival of HCC patients was analysed. KEY RESULTS Treatment of human HCC cells with TC Mps1 12 led to chromosome misalignment and missegregation, and disorganization of centrosomes. Even in the presence of these errors, TC Mps1 12-treated cells overrode the SAC, resulting in a shortened mitotic duration and mitotic slippage. This mitotic catastrophe triggered apoptosis and, finally, inhibited the growth of HCC cells. In addition, the expression of the Mps1-encoding TTK gene was associated with poor overall survival of HCC patients. CONCLUSION AND IMPLICATIONS TC Mps1 12 results in the accumulation of chromosomal instabilities and mitotic catastrophe in HCC cells. Overall, these data demonstrate that the inhibition of Mps1 kinase using TC Mps1 12 is a promising therapeutic approach for liver cancer.
Collapse
Affiliation(s)
- Minji Choi
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Yoo Hong Min
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Jaehyuk Pyo
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon, Republic of Korea
| | - Chang-Young Jang
- College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea.,East-West Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea.,Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
16
|
López-García C, Sansregret L, Domingo E, McGranahan N, Hobor S, Birkbak NJ, Horswell S, Grönroos E, Favero F, Rowan AJ, Matthews N, Begum S, Phillimore B, Burrell R, Oukrif D, Spencer-Dene B, Kovac M, Stamp G, Stewart A, Danielsen H, Novelli M, Tomlinson I, Swanton C. BCL9L Dysfunction Impairs Caspase-2 Expression Permitting Aneuploidy Tolerance in Colorectal Cancer. Cancer Cell 2017; 31:79-93. [PMID: 28073006 PMCID: PMC5225404 DOI: 10.1016/j.ccell.2016.11.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 08/05/2016] [Accepted: 10/28/2016] [Indexed: 01/03/2023]
Abstract
Chromosomal instability (CIN) contributes to cancer evolution, intratumor heterogeneity, and drug resistance. CIN is driven by chromosome segregation errors and a tolerance phenotype that permits the propagation of aneuploid genomes. Through genomic analysis of colorectal cancers and cell lines, we find frequent loss of heterozygosity and mutations in BCL9L in aneuploid tumors. BCL9L deficiency promoted tolerance of chromosome missegregation events, propagation of aneuploidy, and genetic heterogeneity in xenograft models likely through modulation of Wnt signaling. We find that BCL9L dysfunction contributes to aneuploidy tolerance in both TP53-WT and mutant cells by reducing basal caspase-2 levels and preventing cleavage of MDM2 and BID. Efforts to exploit aneuploidy tolerance mechanisms and the BCL9L/caspase-2/BID axis may limit cancer diversity and evolution.
Collapse
Affiliation(s)
- Carlos López-García
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Laurent Sansregret
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Enric Domingo
- Oxford Centre for Cancer Gene Research, The Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK; Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Nicholas McGranahan
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Translational Cancer Therapeutics Laboratory, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC2E 6DD, UK
| | - Sebastijan Hobor
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nicolai Juul Birkbak
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Translational Cancer Therapeutics Laboratory, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC2E 6DD, UK
| | - Stuart Horswell
- Bioinformatics Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Eva Grönroos
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Francesco Favero
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Cancer System Biology, Centre for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby 2800, Denmark
| | - Andrew J Rowan
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nicholas Matthews
- Advanced Sequencing Facility, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sharmin Begum
- Advanced Sequencing Facility, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Benjamin Phillimore
- Advanced Sequencing Facility, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Rebecca Burrell
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Dahmane Oukrif
- Research Department of Pathology, University College London Medical School, University Street, London WC1E 6JJ, UK
| | - Bradley Spencer-Dene
- Experimental Histopathology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michal Kovac
- Oxford Centre for Cancer Gene Research, The Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Gordon Stamp
- Experimental Histopathology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Aengus Stewart
- Bioinformatics Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Havard Danielsen
- Institute for Cancer Genetics and Informatics, Norwegian Radium Hospital, Oslo University Hospital, Ullernchausseen 70, 0379 Oslo, Norway
| | - Marco Novelli
- Research Department of Pathology, University College London Medical School, University Street, London WC1E 6JJ, UK
| | - Ian Tomlinson
- Oxford Centre for Cancer Gene Research, The Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Translational Cancer Therapeutics Laboratory, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC2E 6DD, UK.
| |
Collapse
|
17
|
Lu HY, Lin RT, Zhou GX, Yu TM, Liu ZJ. Critical Role of p53 and K-ras in the Diagnosis of Early Colorectal Cancer: a One-year, Single-center Analysis. Int J Med Sci 2017; 14:1154-1162. [PMID: 29104470 PMCID: PMC5666547 DOI: 10.7150/ijms.20538] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022] Open
Abstract
Background: Colorectal cancer (CRC) is strongly associated with colorectal polyps, which has become the third most common cancer in China. In the present study, we revealed the susceptible population and risk factors of colorectal polyps, and analyzed the expression of Ki-67, p53 and K-ras in the intestinal mucosa of patients with colorectal polyps in order to explore their significance in the detection and prognosis of CRC at an early stage. Materials and Methods: Total 801 cases of colorectal polyps were collected during endoscopic resection including endoscopic mucosal resection (EMR) and endoscopic submucosal dissection (ESD). Expression of Ki-67, p53 and K-ras in the intestinal mucosa was detected by immunohistochemistry and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. Histological analysis was performed by Hematoxylin and eosin (HE) staining. Categorical variables were compared by one-way ANOVA, Pearson test, Spearman test, Kruskal-Wallis test and analysis of regression. Results: Of all patients with colorectal polyps, 90.76% of patients (n = 727) were ≥ 50 years old. 530 cases (66.17%) were males compared with 271 females (33.83%) in all 801 cases. More importantly, 1.03% patients (n = 7) underwent polypectomy and histological examination was confirmed to be the early stage of CRC. The expression of p53 was found to be significantly decreased, while K-ras was increased in tumor tissues of CRC compared with that in hyperplastic polyps and healthy controls. Conclusions: 1.03% patients (n = 7) underwent polypectomy was confirmed to be the early stage of CRC. Histological analysis for expression of p53 and K-ras can guarantee to screen the early stage of CRC.
Collapse
Affiliation(s)
- Hui-Ying Lu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Ri-Tian Lin
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Guang-Xi Zhou
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Tian-Ming Yu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Zhan-Ju Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| |
Collapse
|
18
|
Jemaà M, Manic G, Lledo G, Lissa D, Reynes C, Morin N, Chibon F, Sistigu A, Castedo M, Vitale I, Kroemer G, Abrieu A. Whole-genome duplication increases tumor cell sensitivity to MPS1 inhibition. Oncotarget 2016; 7:885-901. [PMID: 26637805 PMCID: PMC4808040 DOI: 10.18632/oncotarget.6432] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/18/2015] [Indexed: 12/31/2022] Open
Abstract
Several lines of evidence indicate that whole-genome duplication resulting in tetraploidy facilitates carcinogenesis by providing an intermediate and metastable state more prone to generate oncogenic aneuploidy. Here, we report a novel strategy to preferentially kill tetraploid cells based on the abrogation of the spindle assembly checkpoint (SAC) via the targeting of TTK protein kinase (better known as monopolar spindle 1, MPS1). The pharmacological inhibition as well as the knockdown of MPS1 kills more efficiently tetraploid cells than their diploid counterparts. By using time-lapse videomicroscopy, we show that tetraploid cells do not survive the aborted mitosis due to SAC abrogation upon MPS1 depletion. On the contrary diploid cells are able to survive up to at least two more cell cycles upon the same treatment. This effect might reflect the enhanced difficulty of cells with whole-genome doubling to tolerate a further increase in ploidy and/or an elevated level of chromosome instability in the absence of SAC functions. We further show that MPS1-inhibited tetraploid cells promote mitotic catastrophe executed by the intrinsic pathway of apoptosis, as indicated by the loss of mitochondrial potential, the release of the pro-apoptotic cytochrome c from mitochondria, and the activation of caspases. Altogether, our results suggest that MPS1 inhibition could be used as a therapeutic strategy for targeting tetraploid cancer cells.
Collapse
Affiliation(s)
- Mohamed Jemaà
- CRBM, CNRS UMR5237, Université de Montpellier, Montpellier, France
| | | | - Gwendaline Lledo
- CRBM, CNRS UMR5237, Université de Montpellier, Montpellier, France
| | - Delphine Lissa
- Université Paris-Sud/Paris XI, Le Kremlin-Bicêtre, France.,INSERM, UMRS1138, Paris, France.,Equipe 11 Labelisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Gustave Roussy Cancer Campus, Villejuif, France
| | - Christelle Reynes
- EA 2415, Laboratoire de Biostatistique, d'Epidémiologie et de Recherche Clinique, Université de Montpellier, Montpellier, France
| | - Nathalie Morin
- CRBM, CNRS UMR5237, Université de Montpellier, Montpellier, France
| | - Frédéric Chibon
- Department of Biopathology, Institut Bergonié, Comprehensive Cancer Centre, Bordeaux, France.,INSERM U916, Bordeaux, France
| | | | - Maria Castedo
- Université Paris-Sud/Paris XI, Le Kremlin-Bicêtre, France.,INSERM, UMRS1138, Paris, France.,Equipe 11 Labelisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Gustave Roussy Cancer Campus, Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Guido Kroemer
- INSERM, UMRS1138, Paris, France.,Equipe 11 Labelisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Ariane Abrieu
- CRBM, CNRS UMR5237, Université de Montpellier, Montpellier, France
| |
Collapse
|
19
|
Reversine Induced Multinucleated Cells, Cell Apoptosis and Autophagy in Human Non-Small Cell Lung Cancer Cells. PLoS One 2016; 11:e0158587. [PMID: 27385117 PMCID: PMC4934785 DOI: 10.1371/journal.pone.0158587] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 06/17/2016] [Indexed: 12/21/2022] Open
Abstract
Reversine, an A3 adenosine receptor antagonist, has been shown to induce differentiated myogenic-lineage committed cells to become multipotent mesenchymal progenitor cells. We and others have reported that reversine has an effect on human tumor suppression. This study revealed anti-tumor effects of reversine on proliferation, apoptosis and autophagy induction in human non-small cell lung cancer cells. Treatment of these cells with reversine suppressed cell growth in a time- and dosage-dependent manner. Moreover, polyploidy occurred after reversine treatment. In addition, caspase-dependent apoptosis and activation of autophagy by reversine in a dosage-dependent manner were also observed. We demonstrated in this study that reversine contributes to growth inhibition, apoptosis and autophagy induction in human lung cancer cells. Therefore, reversine used as a potential therapeutic agent for human lung cancer is worthy of further investigation.
Collapse
|
20
|
Reversine triggers mitotic catastrophe and apoptosis in K562 cells. Leuk Res 2016; 48:26-31. [PMID: 27447890 DOI: 10.1016/j.leukres.2016.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/30/2016] [Accepted: 06/30/2016] [Indexed: 02/06/2023]
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasm of the hematopoietic stem cell characterized by presence of the oncoprotein BCR-ABL1, which have constitutive tyrosine kinase activity. BCR-ABL1 activation induces aurora kinase A (AURKA) and aurora kinase B (AURKB) expression, which are serine-threonine kinases that play an important function in chromosome alignment, segregation and cytokinesis during mitosis. Acquisition of resistance to tyrosine kinase inhibitors has emerged as a problem for CML patients and the identification of novel targets with an important contribution for CML phenotype is of interest. In the present study, we explored the cellular effects of reversine, an AURKA and AURKB inhibitor, in the BCR-ABL1+ K562 cells. Our results indicate that reversine reduces AURKA and AURKB expression, leads to reduction of cell viability and increased apoptosis in a dose- and time-dependent manner, as well as, induces mitotic catastrophe in K562 cells. Our preclinical study establishes that reversine presents an effective antileukemia activity against K562 cells and provide new insights on anticancer opportunities for CML.
Collapse
|
21
|
Spindle Assembly Checkpoint as a Potential Target in Colorectal Cancer: Current Status and Future Perspectives. Clin Colorectal Cancer 2016; 16:1-8. [PMID: 27435760 DOI: 10.1016/j.clcc.2016.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/03/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
Abstract
Colorectal cancer (CRC), one of the most common malignancies worldwide, is often diagnosed at an advanced stage, and resistance to chemotherapeutic and existing targeted therapy is a major obstacle to its successful treatment. New targets that offer alternative clinical options are therefore urgently needed. Recently, perturbation of the spindle assembly checkpoint (SAC), the surveillance mechanism that maintains anaphase inhibition until all chromosomes reach the metaphase plate, has been regarded as a promising target to fight cancer cells, either alone or in combination regimens. Consistent with this strategy, many cancers, including CRC, exhibit altered expression of SAC genes. In this article, we review our current knowledge on SAC activity status in CRC, and on current anti-CRC strategies and future therapeutic perspectives on the basis of SAC targeting experiments in vitro and in animal models.
Collapse
|
22
|
Kim WH, Shen H, Jung DW, Williams DR. Some leopards can change their spots: potential repositioning of stem cell reprogramming compounds as anti-cancer agents. Cell Biol Toxicol 2016; 32:157-68. [DOI: 10.1007/s10565-016-9333-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/28/2016] [Indexed: 01/14/2023]
|
23
|
Resolving cancer-stroma interfacial signalling and interventions with micropatterned tumour-stromal assays. Nat Commun 2014; 5:5662. [PMID: 25489927 PMCID: PMC4261930 DOI: 10.1038/ncomms6662] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 10/24/2014] [Indexed: 01/09/2023] Open
Abstract
Tumor-stromal interactions are a determining factor in cancer progression. In vivo, the interaction interface is associated with spatially-resolved distributions of cancer and stromal phenotypes. Here, we establish a micropatterned tumor-stromal assay (μTSA) with laser capture microdissection to control the location of co-cultured cells and analyze bulk and interfacial tumor-stromal signaling in driving cancer progression. μTSA reveals a spatial distribution of phenotypes in concordance with human estrogen receptor-positive (ER+) breast cancer samples, and heterogeneous drug activity relative to the tumor-stroma interface. Specifically, an unknown mechanism of reversine is shown in targeting tumor-stromal interfacial interactions using ER+ MCF-7 breast cancer and bone marrow-derived stromal cells. Reversine suppresses MCF-7 tumor growth and bone metastasis in vivo by reducing tumor stromalization including collagen deposition and recruitment of activated stromal cells. This study advocates μTSA as a platform for studying tumor microenvironmental interactions and cancer field effects with applications in drug discovery and development.
Collapse
|
24
|
Co-expression of POU4F2/Brn-3b with p53 may be important for controlling expression of pro-apoptotic genes in cardiomyocytes following ischaemic/hypoxic insults. Cell Death Dis 2014; 5:e1503. [PMID: 25356872 PMCID: PMC4649532 DOI: 10.1038/cddis.2014.452] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/15/2014] [Accepted: 08/19/2014] [Indexed: 02/06/2023]
Abstract
Cardiomyocyte death following ischaemic/hypoxic injury causes irreversible damage to cardiac function and contributes to chronic diseases such as heart failure. Understanding the mechanisms associated with myocyte loss under these conditions can help to identify strategies to minimise/abrogate such detrimental effects. The p53 protein can induce apoptosis or cell cycle arrest, but effects on cell fate depend on interactions with other regulators such as POU4F2/Brn-3b (Brn-3b), which co-operates with p53 to increase the expression of pro-apoptotic genes. In contrast, the related POU4F1/Brn-3a (Brn-3a) blocks p53-mediated apoptosis but co-operates with p53 to enhance cell cycle arrest. In this study, we showed that permanent coronary artery ligation in mouse hearts, which induced apoptotic markers, activated caspase-3 and -8 and necroptosis markers; RIP-1 and -3 also increased Brn-3b and Brn-3a expression. However, Brn-3a was only detected in uninjured myocardium but not at the site of injury, whereas Brn-3b showed generalised increase, including within the infarct zone. Conversely, p53 was detected in the infarct zone and in some cells adjacent to the site of injury but not in uninjured myocardium. Co-localisation studies showed Brn-3a co-expression with p53 in cardiomyocytes adjacent to the infarct zone, whereas Brn-3b was co-localised with p53 in the infarct zone only. Increased Brn-3b and p53 correlated with elevated expression of pro-apoptotic target genes, Bax, Noxa and PUMA, whereas cleaved caspase-3 confirmed the presence of apoptotic cells within this region of the injured heart. Similarly, simulated ischaemia/reoxygenation (sI/R) injury in neonatal rat ventricular cardiomyocytes (NRVM) and heart derived H9c2 myoblasts increased Brn-3b, p53 as well as apoptotic genes, and this was associated with enhanced apoptosis. Furthermore, targeted reduction of Brn-3b using shRNA caused reduction in pro-apoptotic Bax and Noxa proteins, even though p53 expression remained intact, suggesting that Brn-3b is important for controlling the fate of the myocardium in the injured heart.
Collapse
|
25
|
Park JG, Lee DH, Moon YS, Kim KH. Reversine increases the plasticity of lineage-committed preadipocytes to osteogenesis by inhibiting adipogenesis through induction of TGF-β pathway in vitro. Biochem Biophys Res Commun 2014; 446:30-6. [PMID: 24548409 DOI: 10.1016/j.bbrc.2014.02.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 02/07/2014] [Indexed: 10/25/2022]
Abstract
Reversine has been reported to reverse differentiation of lineage-committed cells to mesenchymal stem cells (MSCs), which then enables them to be differentiated into other various lineages. Both adipocytes and osteoblasts are known to originate from common MSCs, and the balance between adipogenesis and osteogenesis in MSCs is reported to modulate the progression of various human diseases, such as obesity and osteoporosis. However, the role of reversine in modulating the adipogenic potential of lineage-committed preadipocytes and their plasticity to osteogenesis is unclear. Here we report that reversine has an anti-adipogenic function in 3T3-L1 preadipocytes in vitro and alters cell morphology and viability. The transforming growth factor-β (TGF-β) pathway appears to be required for the anti-adipogenic effect of reversine, due to reversine-induced expression of genes involved in TGF-β pathway and reversal of reversine-inhibited adipogenesis by inhibition of TGF-β pathway. We show that treatment with reversine transformed 3T3-L1 preadipocytes into MSC-like cells, as evidenced by the expression of MSCs marker genes. This, in turn, allowed differentiation of lineage-committed 3T3-L1 preadipocytes to osteoblasts under the osteogenic condition in vitro. Collectively, these findings reveal a new function of reversine in reversing lineage-committed preadipocytes to osteogenesis in vitro, and provide new insights into adipose tissue-based regeneration of osteoblasts.
Collapse
Affiliation(s)
- Jeong Geun Park
- Department of Animal Science and Biotechnology, Gyeongnam National University of Science and Technology, 33 Dongjin-ro, Jinju 660-758, Republic of Korea
| | - Dae-Hee Lee
- Sempio Fermentation Research Center, Sempio Foods Company, 183 Osongsaengmyeong 4ro, Osongeup, Cheongwongun, Chungcheongbukdo 363-954, Republic of Korea
| | - Yang Soo Moon
- Department of Animal Science and Biotechnology, Gyeongnam National University of Science and Technology, 33 Dongjin-ro, Jinju 660-758, Republic of Korea.
| | - Kee-Hong Kim
- Department of Food Science, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
26
|
Autophagy, a novel target for chemotherapeutic intervention of thyroid cancer. Cancer Chemother Pharmacol 2013; 73:439-49. [DOI: 10.1007/s00280-013-2363-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/16/2013] [Indexed: 01/07/2023]
|
27
|
Characterization of novel MPS1 inhibitors with preclinical anticancer activity. Cell Death Differ 2013; 20:1532-45. [PMID: 23933817 DOI: 10.1038/cdd.2013.105] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 06/10/2013] [Accepted: 07/08/2013] [Indexed: 11/08/2022] Open
Abstract
Monopolar spindle 1 (MPS1), a mitotic kinase that is overexpressed in several human cancers, contributes to the alignment of chromosomes to the metaphase plate as well as to the execution of the spindle assembly checkpoint (SAC). Here, we report the identification and functional characterization of three novel inhibitors of MPS1 of two independent structural classes, N-(4-{2-[(2-cyanophenyl)amino][1,2,4]triazolo[1,5-a]pyridin-6-yl}phenyl)-2-phenylacetamide (Mps-BAY1) (a triazolopyridine), N-cyclopropyl-4-{8-[(2-methylpropyl)amino]-6-(quinolin-5-yl)imidazo[1,2-a]pyrazin-3-yl}benzamide (Mps-BAY2a) and N-cyclopropyl-4-{8-(isobutylamino)imidazo[1,2-a]pyrazin-3-yl}benzamide (Mps-BAY2b) (two imidazopyrazines). By selectively inactivating MPS1, these small inhibitors can arrest the proliferation of cancer cells, causing their polyploidization and/or their demise. Cancer cells treated with Mps-BAY1 or Mps-BAY2a manifested multiple signs of mitotic perturbation including inefficient chromosomal congression during metaphase, unscheduled SAC inactivation and severe anaphase defects. Videomicroscopic cell fate profiling of histone 2B-green fluorescent protein-expressing cells revealed the capacity of MPS1 inhibitors to subvert the correct timing of mitosis as they induce a premature anaphase entry in the context of misaligned metaphase plates. Hence, in the presence of MPS1 inhibitors, cells either divided in a bipolar (but often asymmetric) manner or entered one or more rounds of abortive mitoses, generating gross aneuploidy and polyploidy, respectively. In both cases, cells ultimately succumbed to the mitotic catastrophe-induced activation of the mitochondrial pathway of apoptosis. Of note, low doses of MPS1 inhibitors and paclitaxel (a microtubular poison) synergized at increasing the frequency of chromosome misalignments and missegregations in the context of SAC inactivation. This resulted in massive polyploidization followed by the activation of mitotic catastrophe. A synergistic interaction between paclitaxel and MPS1 inhibitors could also be demonstrated in vivo, as the combination of these agents efficiently reduced the growth of tumor xenografts and exerted superior antineoplastic effects compared with either compound employed alone. Altogether, these results suggest that MPS1 inhibitors may exert robust anticancer activity, either as standalone therapeutic interventions or combined with microtubule-targeting chemicals.
Collapse
|
28
|
Doménech E, Malumbres M. Mitosis-targeting therapies: a troubleshooting guide. Curr Opin Pharmacol 2013; 13:519-28. [PMID: 23583638 DOI: 10.1016/j.coph.2013.03.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 03/23/2013] [Accepted: 03/25/2013] [Indexed: 12/22/2022]
Abstract
Several mitotic kinases and kinesins are currently considered as cancer targets based on their critical role during the cell division cycle and their significant level of expression in human tumors. Yet, their use is limited by the lack of selectivity against tumor cells, the low percentage of mitotic cells in many human tumors, and dose-limiting side-effects. As a consequence, initial clinical trials have shown limited responses. Despite these drawbacks, inhibiting mitosis is a promising strategy that deserves further development. Future advances will benefit from more specific inhibitors with better pharmacodynamic properties, a clear physiological characterization and cell-type-specific requirements of old and new mitotic targets, and rational strategies based on synthetic lethal interactions to improve selectivity against tumor cells.
Collapse
Affiliation(s)
- Elena Doménech
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | |
Collapse
|
29
|
Mapping genetic alterations causing chemoresistance in cancer: identifying the roads by tracking the drivers. Oncogene 2013; 32:5315-30. [PMID: 23474753 DOI: 10.1038/onc.2013.48] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 01/16/2013] [Accepted: 01/16/2013] [Indexed: 12/12/2022]
Abstract
Although new agents are implemented to cancer therapy, we lack fundamental understandings of the mechanisms of chemoresistance, the main obstacle to cure in cancer. Here we review clinical evidence linking molecular defects to drug resistance across different tumour forms and discuss contemporary experimental evidence exploring these mechanisms. Although evidence, in general, is sparse and fragmentary, merging knowledge links drug resistance, and also sensitivity, to defects in functional pathways having a key role in cell growth arrest or death and DNA repair. As these pathways may act in concert, there is a need to explore multiple mechanisms in parallel. Taking advantage of massive parallel sequencing and other novel high-throughput technologies and base research on biological hypotheses, we now have the possibility to characterize functional defects related to these key pathways and to design a new generation of studies identifying the mechanisms controlling resistance to different treatment regimens in different tumour forms.
Collapse
|
30
|
Synergistic antitumor activity of reversine combined with aspirin in cervical carcinoma in vitro and in vivo. Cytotechnology 2013; 65:643-53. [PMID: 23475158 DOI: 10.1007/s10616-012-9520-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 11/18/2012] [Indexed: 02/07/2023] Open
Abstract
A recent report showed that reversine treatment could induce murine myoblasts dedifferentiation into multipotent progenitor cells and inhibit proliferation of some tumors, and other reports showed that apoptosis of lung adenocarcinoma cells could be induced by aspirin. The aim of the present study was to evaluate the synergistic antitumor effects of reversine and aspirin on cervical cancer. The inhibition rate of reversine and aspirin on cervical cancer cell lines' (HeLa and U14) was determined by MTT method, cell cycle of HeLa and U14 cells was analyzed by FACS, mitochondrial membrane potential of HeLa and U14 was detected using a JC-1 kit. HeLa and U14 colony formation was analyzed by soft agar colony formation assay. The expression of caspase-3, Bcl-2/Bax, cyclin D1 and p21 was detected by qRT-PCR and Western Blotting. Moreover, tumor weight and tumor volume was assessed using a murine model of cervical cancer with U14 cells subcutaneously (s.c.) administered into the neck, separately or combined with drug administration via the intraperitoneal (i.p.) route. The inhibition rate of cells in the combination group (10 μmol/L reversine, 10 mmol/L aspirin) increased significantly in comparison to that when the drugs were used alone (P < 0.05); moreover, this combination could synergistically inhibit the proliferation of five cervical cancer cell lines (HeLa, U14, Siha, Caski and C33A). In the therapeutic mouse model, tumor weight and tumor volume of cervical cancer bearing mice was more reduced when compared with the control agents (P < 0.05) in tumor-bearing mice. The combination of reversine and aspirin exerts synergistic growth inhibition and apoptosis induction on cervical cancers cells.
Collapse
|
31
|
Boilève A, Senovilla L, Vitale I, Lissa D, Martins I, Métivier D, van den Brink S, Clevers H, Galluzzi L, Castedo M, Kroemer G. Immunosurveillance against tetraploidization-induced colon tumorigenesis. Cell Cycle 2013; 12:473-9. [PMID: 23324343 DOI: 10.4161/cc.23369] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Circumstantial evidence suggests that colon carcinogenesis can ensue the transient tetraploidization of (pre-)malignant cells. In line with this notion, the tumor suppressors APC and TP53, both of which are frequently inactivated in colon cancer, inhibit tetraploidization in vitro and in vivo. Here, we show that-contrarily to their wild-type counterparts- Tp53 (-/-) colonocytes are susceptible to drug-induced or spontaneous tetraploidization in vitro. Colon organoids generated from tetraploid Tp53 (-/-) cells exhibit a close-to-normal morphology as compared to their diploid Tp53 (-/-) counterparts, yet the colonocytes constituting these organoids are characterized by an increased cell size and an elevated expression of the immunostimulatory protein calreticulin on the cell surface. The subcutaneous injection of tetraploid Tp53 (-/-) colon organoids led to the generation of proliferating tumors in immunodeficient, but not immunocompetent, mice. Thus, tetraploid Tp53 (-/-) colonocytes fail to survive in immunocompetent mice and develop neoplastic lesions in immunocompromised settings only. These results suggest that tetraploidy is particularly oncogenic in the context of deficient immunosurveillance.
Collapse
|
32
|
Vitale I, Jemaà M, Galluzzi L, Metivier D, Castedo M, Kroemer G. Cytofluorometric assessment of cell cycle progression. Methods Mol Biol 2013; 965:93-120. [PMID: 23296653 DOI: 10.1007/978-1-62703-239-1_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
One of the most prominent features of cellular senescence, a stress response that prevents the propagation of cells that have accumulated potentially oncogenic alterations, is a permanent loss of proliferative potential. Thus, at odds with quiescent cells, which resume proliferation when stimulated to do so, senescent cells cannot proceed through the cell cycle even in the presence of mitogenic factors. Here, we describe a set of cytofluorometric techniques for studying how chemical and/or physical stimuli alter the cell cycle in vitro, in both qualitative and quantitative terms. Taken together, these methods allow for the identification of bona fide cytostatic effects as well as for a refined characterization of cell cycle distributions, providing information on proliferation, DNA content as well as on the presence of cell cycle phase-specific markers. At the end of the chapter, a set of guidelines is offered to assist researchers that approach the study of the cell cycle with the interpretation of results.
Collapse
|
33
|
Lu CH, Liu YW, Hua SC, Yu HI, Chang YP, Lee YR. Autophagy induction of reversine on human follicular thyroid cancer cells. Biomed Pharmacother 2012; 66:642-7. [PMID: 23089471 DOI: 10.1016/j.biopha.2012.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 08/13/2012] [Indexed: 01/07/2023] Open
Abstract
The incurable differentiated thyroid cancer (DTC), poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC) are the most aggressive in all of the thyroid cancers. Unfortunately, there are almost no effective therapies. A novel and effective treatment is urgently needed to develop. Recently, reversine, a small synthetic purine analogue, has been reported to be effective in human thyroid cancer suppression through cell cycle arrest and apoptosis induction. In this study, we performed an in vitro evaluation of reversine on autophagy activation, one of the programmed cell death, and the related mechanisms in human follicular thyroid cancer cell line WRO. Incubation of WRO cells with reversine induced autophagosome formation in a short time treatment. LC3-II overexpression in a dosage-dependent manner with reversine treatment was demonstrated in the autophagy activation. Moreover, reversine suppressed Akt/mTOR related signaling pathway activation, a major pathway for autophagy activation, was also revealed in WRO cells. Our data demonstrated that reversine is effective to induce autophagy. Moreover, the LC3-II overexpression and the p62 protein were degraded in a time-dependent manner, indicating that the autophagic flux has happened in the reversine treated WRO cells. In addition, the activation of Akt/mTOR/p70S6K related pathways were shown to be reduced, suggesting these pathways may involve in the reversine mediated autophagy induction. Reversine is therefore worthy of further investigation in clinical therapeutics.
Collapse
Affiliation(s)
- Chieh-Hsiang Lu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chiayi Christian Hospital, Chiayi, Taiwan
| | | | | | | | | | | |
Collapse
|