51
|
D'Alesio C, Punzi S, Cicalese A, Fornasari L, Furia L, Riva L, Carugo A, Curigliano G, Criscitiello C, Pruneri G, Pelicci PG, Faretta M, Bossi D, Lanfrancone L. RNAi screens identify CHD4 as an essential gene in breast cancer growth. Oncotarget 2018; 7:80901-80915. [PMID: 27779108 PMCID: PMC5348363 DOI: 10.18632/oncotarget.12646] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/29/2016] [Indexed: 12/15/2022] Open
Abstract
Epigenetic regulation plays an essential role in tumor development and epigenetic modifiers are considered optimal potential druggable candidates. In order to identify new breast cancer vulnerabilities and improve therapeutic chances for patients, we performed in vivo and in vitro shRNA screens in a human breast cancer cell model (MCF10DCIS.com cell line) using epigenetic libraries. Among the genes identified in our screening, we deeply investigated the role of Chromodomain Helicase DNA binding Protein 4 (CHD4) in breast cancer tumorigenesis. CHD4 silencing significantly reduced tumor growth in vivo and proliferation in vitro of MCF10DCIS.com cells. Similarly, in vivo breast cancer growth was decreased in a spontaneous mouse model of breast carcinoma (MMTV-NeuT system) and in metastatic patient-derived xenograft models. Conversely, no reduction in proliferative ability of non-transformed mammary epithelial cells (MCF10A) was detected. Moreover, we showed that CHD4 depletion arrests proliferation by inducing a G0/G1 block of cell cycle associated with up-regulation of CDKN1A (p21). These results highlight the relevance of genetic screens in the identification of tumor frailties and the role of CHD4 as a potential pharmacological target to inhibit breast cancer growth.
Collapse
Affiliation(s)
- Carolina D'Alesio
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Simona Punzi
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Angelo Cicalese
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Lorenzo Fornasari
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Laura Furia
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Laura Riva
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milan 20139, Italy
| | - Alessandro Carugo
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy.,Department of Molecular and Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Giuseppe Curigliano
- Division of Experimental Therapeutics, European Institute of Oncology, Milan 20141, Italy
| | - Carmen Criscitiello
- Division of Experimental Therapeutics, European Institute of Oncology, Milan 20141, Italy
| | - Giancarlo Pruneri
- School of Medicine, University of Milan, Milan 20122, Italy.,Biobank for Translational Medicine Unit, Department of Pathology, European Institute of Oncology, Milan 20141, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy.,Department of Oncology, University of Milan, Milan 20139, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Daniela Bossi
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| |
Collapse
|
52
|
Pan R, Ruvolo V, Mu H, Leverson JD, Nichols G, Reed JC, Konopleva M, Andreeff M. Synthetic Lethality of Combined Bcl-2 Inhibition and p53 Activation in AML: Mechanisms and Superior Antileukemic Efficacy. Cancer Cell 2017; 32:748-760.e6. [PMID: 29232553 PMCID: PMC5730338 DOI: 10.1016/j.ccell.2017.11.003] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 09/16/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Evasion of apoptosis is a hallmark of cancer. Bcl-2 and p53 represent two important nodes in apoptosis signaling pathways. We find that concomitant p53 activation and Bcl-2 inhibition overcome apoptosis resistance and markedly prolong survival in three mouse models of resistant acute myeloid leukemia (AML). Mechanistically, p53 activation negatively regulates the Ras/Raf/MEK/ERK pathway and activates GSK3 to modulate Mcl-1 phosphorylation and promote its degradation, thus overcoming AML resistance to Bcl-2 inhibition. Moreover, Bcl-2 inhibition reciprocally overcomes apoptosis resistance to p53 activation by switching cellular response from G1 arrest to apoptosis. The efficacy, together with the mechanistic findings, reveals the potential of simultaneously targeting these two apoptosis regulators and provides a rational basis for clinical testing of this therapeutic approach.
Collapse
Affiliation(s)
- Rongqing Pan
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vivian Ruvolo
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Mu
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Gwen Nichols
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center New York, New York, NY 10016, USA
| | - John C Reed
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Marina Konopleva
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
53
|
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
|
54
|
Yue X, Zhang C, Zhao Y, Liu J, Lin AW, Tan VM, Drake JM, Liu L, Boateng MN, Li J, Feng Z, Hu W. Gain-of-function mutant p53 activates small GTPase Rac1 through SUMOylation to promote tumor progression. Genes Dev 2017; 31:1641-1654. [PMID: 28947497 PMCID: PMC5647935 DOI: 10.1101/gad.301564.117] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 08/21/2017] [Indexed: 11/24/2022]
Abstract
Here, Yue et al. investigated the mechanisms underlying p53 gain-of-function (GOF) mutations and found that mutant p53 activates small GTPase Rac1 as a critical mechanism for mutant p53 GOF to promote tumor progression. Their findings provide insight into a new mechanism for Rac1 activation in tumors and show that activation of Rac1 is an unidentified and critical mechanism for mutant p53 GOF in tumorigenesis. Tumor suppressor p53 is frequently mutated in human cancer. Mutant p53 often promotes tumor progression through gain-of-function (GOF) mechanisms. However, the mechanisms underlying mutant p53 GOF are not well understood. In this study, we found that mutant p53 activates small GTPase Rac1 as a critical mechanism for mutant p53 GOF to promote tumor progression. Mechanistically, mutant p53 interacts with Rac1 and inhibits its interaction with SUMO-specific protease 1 (SENP1), which in turn inhibits SENP1-mediated de-SUMOylation of Rac1 to activate Rac1. Targeting Rac1 signaling by RNAi, expression of the dominant-negative Rac1 (Rac1 DN), or the specific Rac1 inhibitor NSC23766 greatly inhibits mutant p53 GOF in promoting tumor growth and metastasis. Furthermore, mutant p53 expression is associated with enhanced Rac1 activity in clinical tumor samples. These results uncover a new mechanism for Rac1 activation in tumors and, most importantly, reveal that activation of Rac1 is an unidentified and critical mechanism for mutant p53 GOF in tumorigenesis, which could be targeted for therapy in tumors containing mutant p53.
Collapse
Affiliation(s)
- Xuetian Yue
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Radiation Oncology, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Cen Zhang
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Radiation Oncology, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Yuhan Zhao
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Radiation Oncology, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Juan Liu
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Radiation Oncology, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Alan W Lin
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Victor M Tan
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Justin M Drake
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Lianxin Liu
- Key Laboratory of Hepatosplenic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Michael N Boateng
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Radiation Oncology, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Jun Li
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Radiation Oncology, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Zhaohui Feng
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Radiation Oncology, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, the State University of New Jersey, Piscataway, New Jersey 08854, USA
| | - Wenwei Hu
- Rutgers Cancer Institute of New Jersey, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Radiation Oncology, the State University of New Jersey, New Brunswick, New Jersey 08903, USA.,Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, the State University of New Jersey, Piscataway, New Jersey 08854, USA
| |
Collapse
|
55
|
Up-regulation of CIT promotes the growth of colon cancer cells. Oncotarget 2017; 8:71954-71964. [PMID: 29069760 PMCID: PMC5641103 DOI: 10.18632/oncotarget.18615] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 05/06/2017] [Indexed: 12/18/2022] Open
Abstract
Colon cancer is one of the major causes of cancer mortality worldwide. However, the underlying mechanism and therapeutic targets of colon cancer have not yet been fully elucidated. In the present study, we demonstrate that citron rho-interacting, serine/threonine kinase 21 (CIT) promotes the growth of human colon cancer cells. CIT is overexpressed in human colon cancer tissues and cell lines. High expression of CIT predicts poor survival for patients with colon cancer. In colon cancer cells, CIT knockdown represses cellular proliferation and colony formation. Our in vivo xenograft experiments showed that CIT knockdown reduces the growth rate of colon cancer cells and the final tumor weight. We found that CIT knockdown induces cell cycle arrest and apoptosis in colon cancer cells. Further microarray and bioinformatics analyses indicated that CIT regulates the p53 signaling pathway, which may account for the effects of CIT on colon cancer cells. Taken together, our findings provide evidence that CIT may promote the development of colon cancer, at least in part, through the p53 signaling pathway. Therefore, CIT may be a potential therapeutic target for colon cancer treatment.
Collapse
|
56
|
Yue X, Zhao Y, Xu Y, Zheng M, Feng Z, Hu W. Mutant p53 in Cancer: Accumulation, Gain-of-Function, and Therapy. J Mol Biol 2017; 429:1595-1606. [PMID: 28390900 PMCID: PMC5663274 DOI: 10.1016/j.jmb.2017.03.030] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 12/19/2022]
Abstract
Tumor suppressor p53 plays a central role in tumor suppression. p53 is the most frequently mutated gene in human cancer, and over half of human cancers contain p53 mutations. Majority of p53 mutations in cancer are missense mutations, leading to the expression of full-length mutant p53 (mutp53) protein. While the critical role of wild-type p53 in tumor suppression has been firmly established, mounting evidence has demonstrated that many tumor-associated mutp53 proteins not only lose the tumor-suppressive function of wild-type p53 but also gain new activities to promote tumorigenesis independently of wild-type p53, termed gain-of-function. Mutant p53 protein often accumulates to very high levels in tumors, contributing to malignant progression. Recently, mutp53 has become an attractive target for cancer therapy. Further understanding of the mechanisms underlying mutp53 protein accumulation and gain-of-function will accelerate the development of targeted therapies for human cancer harboring mutp53. In this review, we summarize the recent advances in the studies on mutp53 protein accumulation and gain-of-function and targeted therapies for mutp53 in human cancer.
Collapse
Affiliation(s)
- Xuetian Yue
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Yuhan Zhao
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Yang Xu
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Min Zheng
- State Key Lab of Diagnostic and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhaohui Feng
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA; Department of Pharmacology, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA.
| | - Wenwei Hu
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA; Department of Pharmacology, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA.
| |
Collapse
|
57
|
Zhang X, Cheng Q, Yin H, Yang G. Regulation of autophagy and EMT by the interplay between p53 and RAS during cancer progression (Review). Int J Oncol 2017; 51:18-24. [PMID: 28560457 DOI: 10.3892/ijo.2017.4025] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/05/2017] [Indexed: 11/06/2022] Open
Abstract
Cellular autophagy and epithelial-mesenchymal transition (EMT) are key events mostly resulted from the interplay of tumor suppressors and oncogenes during cancer progression. The master tumor suppressor p53 may control tumor cell autophagy and EMT through the transcriptional induction of multiple target genes, while the activated oncogene RAS may also play a critical role in regulating mitogenic signaling to tumor cell autophagy and EMT. Although the fundamental functions of p53 and RAS are well understood, the interactive effects of p53 and RAS on autophagy and EMT are still unclear. In this review, we highlight the recent advances in the regulation of autophagy and EMT by p53 and RAS, aiming to explore novel therapeutic targets and biomarkers in cancer treatment and prevention.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Qian Cheng
- Department of Orthopedics, the Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Huijing Yin
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Gong Yang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| |
Collapse
|
58
|
Meitinger F, Anzola JV, Kaulich M, Richardson A, Stender JD, Benner C, Glass CK, Dowdy SF, Desai A, Shiau AK, Oegema K. 53BP1 and USP28 mediate p53 activation and G1 arrest after centrosome loss or extended mitotic duration. J Cell Biol 2017; 214:155-66. [PMID: 27432897 PMCID: PMC4949453 DOI: 10.1083/jcb.201604081] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/24/2016] [Indexed: 12/14/2022] Open
Abstract
In normal human cells, centrosome loss induced by centrinone-a specific centrosome duplication inhibitor-leads to irreversible, p53-dependent G1 arrest by an unknown mechanism. A genome-wide CRISPR/Cas9 screen for centrinone resistance identified genes encoding the p53-binding protein 53BP1, the deubiquitinase USP28, and the ubiquitin ligase TRIM37. Deletion of TP53BP1, USP28, or TRIM37 prevented p53 elevation in response to centrosome loss but did not affect cytokinesis failure-induced arrest or p53 elevation after doxorubicin-induced DNA damage. Deletion of TP53BP1 and USP28, but not TRIM37, prevented growth arrest in response to prolonged mitotic duration. TRIM37 knockout cells formed ectopic centrosomal-component foci that suppressed mitotic defects associated with centrosome loss. TP53BP1 and USP28 knockouts exhibited compromised proliferation after centrosome removal, suggesting that centrosome-independent proliferation is not conferred solely by the inability to sense centrosome loss. Thus, analysis of centrinone resistance identified a 53BP1-USP28 module as critical for communicating mitotic challenges to the p53 circuit and TRIM37 as an enforcer of the singularity of centrosome assembly.
Collapse
Affiliation(s)
- Franz Meitinger
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - John V Anzola
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Manuel Kaulich
- Institute of Biochemistry II, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Amelia Richardson
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Joshua D Stender
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Christopher Benner
- Department of Medicine, University of California, San Diego, La Jolla, California 92093
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Department of Medicine, University of California, San Diego, La Jolla, California 92093
| | - Steven F Dowdy
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Arshad Desai
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Andrew K Shiau
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Karen Oegema
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Ludwig Institute for Cancer Research, La Jolla, CA 92093
| |
Collapse
|
59
|
Zhirnov OP. Paramyxoviruses activation by host proteases in cultures of normal and cancer cells. Vopr Virusol 2017; 62:65-72. [PMID: 36494930 DOI: 10.18821/0507-4088-2017-62-2-65-72] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Indexed: 12/13/2022]
Abstract
Multiplication of paramyxovirus Sendai and Newcastle disease virus (NDV) was studied in cultures of normal and tumor cells. Production of noninfectious virus with uncleaved F0 was observed in canine kidney cell line MDCK (line H) and its derivatives carrying tetracycline-regulated expression of transmembrane protease HAT or TMPRSS2 with trypsin-like cleavage specificity. Under tetracycline induction, a cleavage F0 (65 kD)>F1 (50 kD)+F2(15 kD) and production of infectious virus were observed in these cell cultures. Under tetracycline induction, the additional subunit 38K (m.w. 38 kDa) of the F protein was detected both in infected MDCK-HAT cells and in newly synthesized Sendai virus in addition to F0, F1 and F2, indicating thereby a second HAT-sensitive proteolytic site in the F0 molecule. Highly infectious virus containing cleaved F1+F2 was produced in cultures of cancer cells Caco-2 and H1299. Virus Sendai synthesized in H1299 cells contained 38 K subunit indicating a cleavage of the F0 at a second site by H1299 host cell proteases. Levels of cleaved F1+F2 and infectious virions were higher at the late stage of infection in cancer cells, suggesting thus the induction of virus-activating proteases in Caco-2 and H1299 cells under infection with paramyxoviruses. NDV virus was found to induce more rapid death of cancer cells Caco-2 than Sendai virus. Cooperatively, the obtained data show that cancer cells in distinction to nonmalignant cells can synthesize protease(s) activating infectivity of paramyxoviruses. Thus, they are more vulnerable to paramyxovirus infection than normal cells.
Collapse
Affiliation(s)
- O P Zhirnov
- Federal State Budgetary Institution «Federal Research Centre of Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya»
| |
Collapse
|
60
|
Molecularly targeted therapies in cancer: a guide for the nuclear medicine physician. Eur J Nucl Med Mol Imaging 2017; 44:41-54. [PMID: 28396911 PMCID: PMC5541087 DOI: 10.1007/s00259-017-3695-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 01/01/2023]
Abstract
Molecular imaging continues to influence every aspect of cancer care including detection, diagnosis, staging and therapy response assessment. Recent advances in the understanding of cancer biology have prompted the introduction of new targeted therapy approaches. Precision medicine in oncology has led to rapid advances and novel approaches optimizing the use of imaging modalities in cancer care, research and development. This article focuses on the concept of targeted therapy in cancer and the challenges that exist for molecular imaging in cancer care.
Collapse
|
61
|
Hatoum D, Yagoub D, Ahadi A, Nassif NT, McGowan EM. Annexin/S100A Protein Family Regulation through p14ARF-p53 Activation: A Role in Cell Survival and Predicting Treatment Outcomes in Breast Cancer. PLoS One 2017; 12:e0169925. [PMID: 28068434 PMCID: PMC5222396 DOI: 10.1371/journal.pone.0169925] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022] Open
Abstract
The annexin family and S100A associated proteins are important regulators of diverse calcium-dependent cellular processes including cell division, growth regulation and apoptosis. Dysfunction of individual annexin and S100A proteins is associated with cancer progression, metastasis and cancer drug resistance. This manuscript describes the novel finding of differential regulation of the annexin and S100A family of proteins by activation of p53 in breast cancer cells. Additionally, the observed differential regulation is found to be beneficial to the survival of breast cancer cells and to influence treatment efficacy. We have used unbiased, quantitative proteomics to determine the proteomic changes occurring post p14ARF-p53 activation in estrogen receptor (ER) breast cancer cells. In this report we identified differential regulation of the annexin/S100A family, through unique peptide recognition at the N-terminal regions, demonstrating p14ARF-p53 is a central orchestrator of the annexin/S100A family of calcium regulators in favor of pro-survival functions in the breast cancer cell. This regulation was found to be cell-type specific. Retrospective human breast cancer studies have demonstrated that tumors with functional wild type p53 (p53wt) respond poorly to some chemotherapy agents compared to tumors with a non-functional p53. Given that modulation of calcium signaling has been demonstrated to change sensitivity of chemotherapeutic agents to apoptotic signals, in principle, we explored the paradigm of how p53 modulation of calcium regulators in ER+ breast cancer patients impacts and influences therapeutic outcomes.
Collapse
Affiliation(s)
- Diana Hatoum
- School of Life Sciences, Faculty of Science, Faculty of Engineering and IT, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Daniel Yagoub
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Alireza Ahadi
- Faculty of Engineering and IT, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Najah T. Nassif
- School of Life Sciences, Faculty of Science, Faculty of Engineering and IT, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Eileen M. McGowan
- School of Life Sciences, Faculty of Science, Faculty of Engineering and IT, University of Technology Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
62
|
Nagel R, Semenova EA, Berns A. Drugging the addict: non-oncogene addiction as a target for cancer therapy. EMBO Rep 2016; 17:1516-1531. [PMID: 27702988 PMCID: PMC5090709 DOI: 10.15252/embr.201643030] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 08/24/2016] [Indexed: 12/13/2022] Open
Abstract
Historically, cancers have been treated with chemotherapeutics aimed to have profound effects on tumor cells with only limited effects on normal tissue. This approach was followed by the development of small‐molecule inhibitors that can target oncogenic pathways critical for the survival of tumor cells. The clinical targeting of these so‐called oncogene addictions, however, is in many instances hampered by the outgrowth of resistant clones. More recently, the proper functioning of non‐mutated genes has been shown to enhance the survival of many cancers, a phenomenon called non‐oncogene addiction. In the current review, we will focus on the distinct non‐oncogenic addictions found in cancer cells, including synthetic lethal interactions, the underlying stress phenotypes, and arising therapeutic opportunities.
Collapse
Affiliation(s)
- Remco Nagel
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ekaterina A Semenova
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anton Berns
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
63
|
Leung AWY, de Silva T, Bally MB, Lockwood WW. Synthetic lethality in lung cancer and translation to clinical therapies. Mol Cancer 2016; 15:61. [PMID: 27686855 PMCID: PMC5041331 DOI: 10.1186/s12943-016-0546-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/21/2016] [Indexed: 01/06/2023] Open
Abstract
Lung cancer is a heterogeneous disease consisting of multiple histological subtypes each driven by unique genetic alterations. Despite the development of targeted therapies that inhibit the oncogenic mutations driving a subset of lung cancer cases, there is a paucity of effective treatments for the majority of lung cancer patients and new strategies are urgently needed. In recent years, the concept of synthetic lethality has been established as an effective approach for discovering novel cancer-specific targets as well as a method to improve the efficacy of existing drugs which provide partial but insufficient benefits for patients. In this review, we discuss the concept of synthetic lethality, the various types of synthetic lethal interactions in the context of oncology and the approaches used to identify these interactions, including recent advances that have transformed the ability to discover novel synthetic lethal combinations on a global scale. Lastly, we describe the specific synthetic lethal interactions identified in lung cancer to date and explore the pharmacological challenges and considerations in translating these discoveries to the clinic.
Collapse
Affiliation(s)
- Ada W. Y. Leung
- Experimental Therapeutics, BC Cancer Research Centre, 675 West 10th Ave, Vancouver, BC V5Z 1L3 Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Rm. G227-2211 Wesbrook Mall, Vancouver, BC V6T 2B5 Canada
| | - Tanya de Silva
- Department of Pathology and Laboratory Medicine, University of British Columbia, Rm. G227-2211 Wesbrook Mall, Vancouver, BC V6T 2B5 Canada
- Integrative Oncology, BC Cancer Research Centre, 675 West 10th Ave, Vancouver, BC V5Z 1L3 Canada
| | - Marcel B. Bally
- Experimental Therapeutics, BC Cancer Research Centre, 675 West 10th Ave, Vancouver, BC V5Z 1L3 Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Rm. G227-2211 Wesbrook Mall, Vancouver, BC V6T 2B5 Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
- Centre for Drug Research and Development, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - William W. Lockwood
- Department of Pathology and Laboratory Medicine, University of British Columbia, Rm. G227-2211 Wesbrook Mall, Vancouver, BC V6T 2B5 Canada
- Integrative Oncology, BC Cancer Research Centre, 675 West 10th Ave, Vancouver, BC V5Z 1L3 Canada
| |
Collapse
|
64
|
Kojima K, Ishizawa J, Andreeff M. Pharmacological activation of wild-type p53 in the therapy of leukemia. Exp Hematol 2016; 44:791-798. [PMID: 27327543 DOI: 10.1016/j.exphem.2016.05.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
Abstract
The tumor suppressor p53 is inactivated by mutations in the majority of human solid tumors. Conversely, p53 mutations are rare in leukemias and are only observed in a small fraction of the patient population, predominately in patients with complex karyotype acute myeloid leukemia or hypodiploid acute lymphoblastic leukemia. However, the loss of p53 function in leukemic cells is often caused by abnormalities in p53-regulatory proteins, including overexpression of MDM2/MDMX, deletion of CDKN2A/ARF, and alterations in ATM. For example, MDM2 inhibits p53-mediated transcription, promotes its nuclear export, and induces proteasome-dependent degradation. The MDM2 homolog MDMX is another direct regulator of p53 that inhibits p53-mediated transcription. Several small-molecule inhibitors and stapled peptides targeting MDM2 and MDMX have been developed and have recently entered clinical trials. The clinical trial results of the first clinically used MDM2 inhibitor, RG7112, illustrated promising p53 activation and apoptosis induction in leukemia cells as proof of concept. Side effects of RG7112 were most prominent in suppression of thrombopoiesis and gastrointestinal symptoms in leukemia patients. Predictive biomarkers for response to MDM2 inhibitors have been proposed, but they require further validation both in vitro and in vivo so that the accumulated knowledge concerning pathological p53 dysregulation in leukemia and novel molecular-targeted strategies to overcome this dysregulation can be translated safely and efficiently into novel clinical therapeutics.
Collapse
Affiliation(s)
- Kensuke Kojima
- Department of Hematology, Respiratory Medicine and Oncology, Division of Medicine, Saga University, Saga, Japan; Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Jo Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
65
|
Saito M, Shiraishi K, Kunitoh H, Takenoshita S, Yokota J, Kohno T. Gene aberrations for precision medicine against lung adenocarcinoma. Cancer Sci 2016; 107:713-20. [PMID: 27027665 PMCID: PMC4968599 DOI: 10.1111/cas.12941] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/28/2016] [Accepted: 03/29/2016] [Indexed: 12/17/2022] Open
Abstract
Lung adenocarcinoma (LADC), the most frequent histological type of lung cancer, is often triggered by an aberration in a driver oncogene in tumor cells. Examples of such aberrations are EGFR mutation and ALK fusion. Lung adenocarcinoma harboring such mutations can be treated with anticancer drugs that target the aberrant gene products. Additional oncogene aberrations, including RET,ROS1, and NRG1 fusions, skipping of exon 14 of MET, and mutations in BRAF,HER2,NF1, and MEK1, were recently added to the list of such “druggable” driver oncogene aberrations, and their responses to targeted therapies are currently being evaluated in clinical trials. However, approximately 30% and 50% of LADCs in patients in Japan and Europe/USA, respectively, lack the driver oncogene aberrations listed above. Therefore, novel therapeutic strategies, such as those that exploit the vulnerabilities of cancer cells with non‐oncogene aberrations, are urgently required. This review summarizes the current status of research on precision medicine against LADC and enumerates the research priorities for the near future.
Collapse
Affiliation(s)
- Motonobu Saito
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan.,Department of Organ Regulatory Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hideo Kunitoh
- Department of Medical Oncology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Seiichi Takenoshita
- Department of Organ Regulatory Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Jun Yokota
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan.,Cancer Genome Biology Group, Institute of Predictive and Personalized Medicine of Cancer, Barcelona, Spain
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
66
|
Christy B, Demaria M, Campisi J, Huang J, Jones D, Dodds SG, Williams C, Hubbard G, Livi CB, Gao X, Weintraub S, Curiel T, Sharp ZD, Hasty P. p53 and rapamycin are additive. Oncotarget 2016; 6:15802-13. [PMID: 26158292 PMCID: PMC4599238 DOI: 10.18632/oncotarget.4602] [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: 05/18/2015] [Accepted: 06/14/2015] [Indexed: 12/13/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a kinase found in a complex (mTORC1) that enables macromolecular synthesis and cell growth and is implicated in cancer etiology. The rapamycin-FK506 binding protein 12 (FKBP12) complex allosterically inhibits mTORC1. In response to stress, p53 inhibits mTORC1 through a separate pathway involving cell signaling and amino acid sensing. Thus, these different mechanisms could be additive. Here we show that p53 improved the ability of rapamycin to: 1) extend mouse life span, 2) suppress ionizing radiation (IR)-induced senescence-associated secretory phenotype (SASP) and 3) increase the levels of amino acids and citric acid in mouse embryonic stem (ES) cells. This additive effect could have implications for cancer treatment since rapamycin and p53 are anti-oncogenic.
Collapse
Affiliation(s)
- Barbara Christy
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Marco Demaria
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Jing Huang
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Diane Jones
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sherry G Dodds
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Charnae Williams
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gene Hubbard
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Carolina B Livi
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Current address: Agilent Technologies, Inc., Santa Clara, CA, USA
| | - Xiaoli Gao
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Susan Weintraub
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tyler Curiel
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Z Dave Sharp
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Paul Hasty
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
67
|
Shin MH, He Y, Marrogi E, Piperdi S, Ren L, Khanna C, Gorlick R, Liu C, Huang J. A RUNX2-Mediated Epigenetic Regulation of the Survival of p53 Defective Cancer Cells. PLoS Genet 2016; 12:e1005884. [PMID: 26925584 PMCID: PMC4771715 DOI: 10.1371/journal.pgen.1005884] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 01/29/2016] [Indexed: 01/15/2023] Open
Abstract
The inactivation of p53 creates a major challenge for inducing apoptosis in cancer cells. An attractive strategy is to identify and subsequently target the survival signals in p53 defective cancer cells. Here we uncover a RUNX2-mediated survival signal in p53 defective cancer cells. The inhibition of this signal induces apoptosis in cancer cells but not non-transformed cells. Using the CRISPR technology, we demonstrate that p53 loss enhances the apoptosis caused by RUNX2 knockdown. Mechanistically, RUNX2 provides the survival signal partially through inducing MYC transcription. Cancer cells have high levels of activating histone marks on the MYC locus and concomitant high MYC expression. RUNX2 knockdown decreases the levels of these histone modifications and the recruitment of the Menin/MLL1 (mixed lineage leukemia 1) complex to the MYC locus. Two inhibitors of the Menin/MLL1 complex induce apoptosis in p53 defective cancer cells. Together, we identify a RUNX2-mediated epigenetic mechanism of the survival of p53 defective cancer cells and provide a proof-of-principle that the inhibition of this epigenetic axis is a promising strategy to kill p53 defective cancer cells. Because activated p53 is a potent inducer of apoptosis, several approaches centering on p53 activation are designed for killing cancer cells. However, more than half of human tumors have p53 inactivation, which renders these p53-activating approaches less effective in killing cancer cells. Targeting the survival signals specific to p53 defective cancer cells offers an opportunity to circumvent the challenge of p53 inactivation. In this study, we showed that one such survival signal is the RUNX2 signaling pathway. To investigate the mechanism underlying this survival signal, we used biochemical, genetic, and genomic approaches. The MYC gene was identified as a novel mediator of the pro-survival function of RUNX2. We further studied the regulatory mechanism of MYC by RUNX2 and found that RUNX2 recruits the Menin/MLL1 epigenetic complex to induce the expression of MYC. Using small molecule inhibitors of the Menin/MLL1 complex, we showed that targeting RUNX2/Menin/MLL1/MYC axis is a feasible strategy for killing p53 defective cancer cells. Our study paves the road for the future development of targeted therapies for OS.
Collapse
Affiliation(s)
- Min Hwa Shin
- Cancer and Stem Cell Epigenetics Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Yunlong He
- Cancer and Stem Cell Epigenetics Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Eryney Marrogi
- Cancer and Stem Cell Epigenetics Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Sajida Piperdi
- Division of Pediatric Hematology-Oncology, The Children’s Hospital at Montefiore, Bronx, New York, United States of America
| | - Ling Ren
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Chand Khanna
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Richard Gorlick
- Division of Pediatric Hematology-Oncology, The Children’s Hospital at Montefiore, Bronx, New York, United States of America
| | - Chengyu Liu
- Transgenic Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, United States of America
| | - Jing Huang
- Cancer and Stem Cell Epigenetics Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
68
|
Jiang W, Zhao S, Jiang X, Zhang E, Hu G, Hu B, Zheng P, Xiao J, Lu Z, Lu Y, Ni J, Chen C, Wang X, Yang L, Wan R. The circadian clock gene Bmal1 acts as a potential anti-oncogene in pancreatic cancer by activating the p53 tumor suppressor pathway. Cancer Lett 2016; 371:314-25. [PMID: 26683776 DOI: 10.1016/j.canlet.2015.12.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/02/2015] [Accepted: 12/02/2015] [Indexed: 12/29/2022]
Abstract
Disruption of the circadian clock has been shown to be associated with tumor development. This study aimed to investigate the role of the core circadian gene Bmal1 in pancreatic cancer (PC). We first found that the levels of Bmal1 were downregulated in PC samples and were closely correlated with the clinicopathological features of patients. To dissect the underlying mechanism, we performed a RNA-seq assay followed by systematic gene function and pathway enrichment analyses. We detected an anti-apoptotic and pro-proliferative transcriptome profile after Bmal1 knockdown in PC cells. Further in vitro and in vivo studies confirmed that Bmal1 overexpression significantly inhibited cell proliferation and invasion and induced G2/M cell cycle arrest, whereas Bmal1 knockdown promoted PC growth, as demonstrated in Bmal1-manipulated AsPC-1 and BxPC-3 cell lines. Our mechanistic studies indicated that Bmal1 could directly bind to the p53 gene promoter and thereby transcriptionally activate the downstream tumor suppressor pathway in a p53-dependent manner. In sum, our findings suggest that Bmal1 acts as an anti-oncogene in PC and represents a potential biomarker for its diagnosis.
Collapse
Affiliation(s)
- Weiliang Jiang
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Senlin Zhao
- Department of General Surgery, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohua Jiang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Erquan Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Guoyong Hu
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Hu
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zheng
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junhua Xiao
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhanjun Lu
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Lu
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianbo Ni
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Congying Chen
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xingpeng Wang
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lijuan Yang
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Rong Wan
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
69
|
Teng YC, Shen ZQ, Kao CH, Tsai TF. Hepatocellular carcinoma mouse models: Hepatitis B virus-associated hepatocarcinogenesis and haploinsufficient tumor suppressor genes. World J Gastroenterol 2016; 22:300-325. [PMID: 26755878 PMCID: PMC4698494 DOI: 10.3748/wjg.v22.i1.300] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/14/2015] [Accepted: 11/24/2015] [Indexed: 02/06/2023] Open
Abstract
The multifactorial and multistage pathogenesis of hepatocellular carcinoma (HCC) has fascinated a wide spectrum of scientists for decades. While a number of major risk factors have been identified, their mechanistic roles in hepatocarcinogenesis still need to be elucidated. Many tumor suppressor genes (TSGs) have been identified as being involved in HCC. These TSGs can be classified into two groups depending on the situation with respect to allelic mutation/loss in the tumors: the recessive TSGs with two required mutated alleles and the haploinsufficient TSGs with one required mutated allele. Hepatitis B virus (HBV) is one of the most important risk factors associated with HCC. Although mice cannot be infected with HBV due to the narrow host range of HBV and the lack of a proper receptor, one advantage of mouse models for HBV/HCC research is the numerous and powerful genetic tools that help investigate the phenotypic effects of viral proteins and allow the dissection of the dose-dependent action of TSGs. Here, we mainly focus on the application of mouse models in relation to HBV-associated HCC and on TSGs that act either in a recessive or in a haploinsufficient manner. Discoveries obtained using mouse models will have a great impact on HCC translational medicine.
Collapse
|
70
|
Ma H, Takahashi A, Sejimo Y, Adachi A, Kubo N, Isono M, Yoshida Y, Kanai T, Ohno T, Nakano T. Targeting of Carbon Ion-Induced G2 Checkpoint Activation in Lung Cancer Cells Using Wee-1 Inhibitor MK-1775. Radiat Res 2015; 184:660-9. [DOI: 10.1667/rr14171.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Hongyu Ma
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Japan; and
| | | | | | - Akiko Adachi
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Japan; and
| | - Nobuteru Kubo
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Japan; and
| | - Mayu Isono
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Yukari Yoshida
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Tatsuaki Kanai
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Tatsuya Ohno
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Takashi Nakano
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Japan; and
| |
Collapse
|
71
|
GUO HUA, LI YANG, GU JUNLIAN, WANG YUE, LIU LIANQIN, ZHANG PING, LIU YANAN. Effect of vascular endothelial growth factor siRNA and wild-type p53 co-expressing plasmid in MDA-MB-231 cells. Mol Med Rep 2015; 13:461-8. [PMID: 26573068 DOI: 10.3892/mmr.2015.4571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 09/22/2015] [Indexed: 11/05/2022] Open
|
72
|
Monteverde T, Muthalagu N, Port J, Murphy DJ. Evidence of cancer-promoting roles for AMPK and related kinases. FEBS J 2015; 282:4658-71. [PMID: 26426570 DOI: 10.1111/febs.13534] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/16/2015] [Accepted: 09/25/2015] [Indexed: 12/14/2022]
Abstract
The discovery that the 5'AMP-activated protein kinase (AMPK) serves to link the tumour suppressors LKB1 and the tuberous sclerosis complex and functions to slow macromolecular synthesis through attenuation of the mechanistic target of rapamycin complex 1 revealed a role for AMPK in tumour suppression. On the other hand, the well-recognized role of AMPK in maintaining ATP homeostasis, through suppression of anabolism and promotion of catabolism, as well as the role of AMPK in neutralizing reactive oxygen species, via maintenance of NADPH-dependent reductive capacity, point to tumour-protective roles in the context of metabolic stress, which is a key feature of many solid tumours. A growing number of studies thus suggest a duality of functions for AMPK that are either pro- or anti-cancer, depending upon context. Importantly, AMPK is composed of three subunits, and multiple isoforms exist for all three, allowing for different permutations to assemble and the potential for specific AMPK complexes to regulate distinct cellular processes. Moreover, certain subunits of the AMPK complex are frequently overexpressed in a spectrum of human cancer types, suggesting an outright oncogenic function for specific AMPK complexes. Adding complexity to this picture, the catalytic AMPK alpha subunits belong to a family of 14 kinases that can all be activated by LKB1 and studies are beginning to reveal a similar duality of roles in cancer for other members of the AMPK-related kinase family.
Collapse
Affiliation(s)
| | | | - Jennifer Port
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Daniel J Murphy
- Institute of Cancer Sciences, University of Glasgow, UK.,CRUK Beatson Institute, Glasgow, UK
| |
Collapse
|