201
|
Wang WX, Xu CW, Chen YP, Liu W, Zhong LH, Chen FF, Zhuang W, Huang YJ, Huang ZZ, Chen RR, Guan YF, Yi X, Lv TF, Zhu WF, Lu JP, Wang XJ, Shi Y, Lin XD, Chen G, Song Y. TP53 mutations predict for poor survival in ALK rearrangement lung adenocarcinoma patients treated with crizotinib. J Thorac Dis 2018; 10:2991-2998. [PMID: 29997966 DOI: 10.21037/jtd.2018.04.98] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Advanced non-small cell lung cancer (NSCLC) patients who harbor anaplastic lymphoma kinase (ALK) rearrangement are sensitive to an ALK inhibitor (crizotinib), but not all ALK-positive patients benefit equally from crizotinib treatment. We analyze the impact of TP53 mutations on response to crizotinib in patients with ALK rearrangement NSCLC. Methods Sixty-six ALK rearrangement NSCLC patients receiving crizotinib were analyzed. 21 cases were detected successfully by the next generation sequencing validation FFPE before crizotinib. TP53 mutations were evaluated in 8 patients in relation to disease control rate (DCR), objective response rate (ORR), progression-free survival (PFS) and overall survival (OS). Results TP53 mutations were observed in 2 (25.00%), 1 (12.50%), 1 (12.50%) and 4 (50.00%) patients in exons 5, 6, 7 and 8, respectively. The majority of patients were male (75.00%, 6/8), less than 65 years old (62.50%, 5/8) and never smokers (75.00%, 6/8). ORR and DCR for crizotinib in the entire case series were 61.90% and 71.43%, respectively. Statistically significant difference was observed in terms of PFS and OS between TP53 gene wild group and mutation group patients (P=0.038, P=0.021, respectively). Conclusions TP53 mutations reduce responsiveness to crizotinib and worsen prognosis in ALK rearrangement NSCLC patients.
Collapse
Affiliation(s)
- Wen-Xian Wang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Chun-Wei Xu
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Yan-Ping Chen
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Wei Liu
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Li-Hua Zhong
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Fang-Fang Chen
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Wu Zhuang
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Yun-Jian Huang
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Zhang-Zhou Huang
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | | | | | - Xin Yi
- Geneplus-Beijing, Beijing 102200, China
| | - Tang-Feng Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing 210002, China
| | - Wei-Feng Zhu
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Jian-Ping Lu
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Xiao-Jiang Wang
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Yi Shi
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Xian-Dong Lin
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Gang Chen
- Department of Pathology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou 350014, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing 210002, China
| |
Collapse
|
202
|
GADD45A and CDKN1A are involved in apoptosis and cell cycle modulatory effects of viscumTT with further inactivation of the STAT3 pathway. Sci Rep 2018; 8:5750. [PMID: 29636527 PMCID: PMC5893628 DOI: 10.1038/s41598-018-24075-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/21/2018] [Indexed: 12/28/2022] Open
Abstract
ViscumTT, a whole mistletoe preparation, has shown synergistic induction of apoptosis in several pediatric tumor entities. High therapeutic potential has previously been observed in Ewing's sarcoma, rhabdomyosarcoma, ALL and AML. In this study, we analyzed modulatory effects on the cell cycle by viscumTT in three osteosarcoma cell lines with various TP53 statuses. ViscumTT treatment induced G1 arrest in TP53 wild-type and null-mutant cells, but S arrest in TP53 mutant cells. Blockage of G1/S transition was accompanied by down-regulation of the key regulators CDK4, CCND1, CDK2, CCNE, CCNA. However, investigations on the transcriptional level revealed secondary TP53 participation. Cell cycle arrest was predominantly mediated by transcriptionally increased expression of GADD45A and CDKN1A and decreased SKP2 levels. Enhanced CDKN1A and GADD45A expression further played a role in viscumTT-induced apoptosis with involvement of stress-induced MAPK8 and inactivation of MAPK1/3. Furthermore, viscumTT inhibited the pro-survival pathway STAT3 by dephosphorylation of the two sites, Tyr705 and Ser727, by down-regulation of total STAT3 and its direct downstream targets BIRC5 and C-MYC. Moreover, tests of the efficacy of viscumTT in vivo showing reduction of tumor volume confirmed the high therapeutic potential as an anti-tumoral agent for osteosarcoma.
Collapse
|
203
|
Neven KY, Saenen ND, Tarantini L, Janssen BG, Lefebvre W, Vanpoucke C, Bollati V, Nawrot TS. Placental promoter methylation of DNA repair genes and prenatal exposure to particulate air pollution: an ENVIRONAGE cohort study. Lancet Planet Health 2018; 2:e174-e183. [PMID: 29615218 DOI: 10.1016/s2542-5196(18)30049-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Exposure to particulate air pollution has been linked with risk of carcinogenesis. Damage to repair pathways might have long-term adverse health effects. We aimed to investigate the association of prenatal exposure to air pollution with placental mutation rate and the DNA methylation of key placental DNA repair genes. METHODS This cohort study used data from the ongoing ENVironmental Influence ON early AGEing (ENVIRONAGE) birth cohort, which enrols pairs of mothers and neonates (singleton births only) at the East-Limburg Hospital (Genk, Belgium). Placental DNA samples were collected after birth. We used bisulfite-PCR-pyrosequencing to investigate the mutation rate of Alu (a marker for overall DNA mutation) and DNA methylation in the promoter genes of key DNA repair and tumour suppressor genes (APEX1, OGG1, PARP1, ERCC1, ERCC4, p53, and DAPK1). We used a high-resolution air pollution model to estimate exposure to particulate matter with a diameter less than 2·5 μm (PM2·5), black carbon, and NO2 over the entire pregnancy on the basis of maternal address. Alu mutation was analysed with a linear regression model, and methylation values of the selected genes were analysed in mixed-effects models. Effect estimates are presented as the relative percentage change in methylation for an ambient air pollution increment of one IQR (ie, the difference between the first and third quartiles of exposure in the entire cohort). FINDINGS 500 biobanked placental DNA samples were randomly selected from 814 pairs of mothers and neonates who were recruited to the cohort between Feb 1, 2010, and Dec 31, 2014, of which 463 samples met the pyrosequencing quality control criteria. IQR exposure increments were 3·84 μg/m3 for PM2·5, 0·36 μg/m3 for black carbon, and 5·34 μg/m3 for NO2. Among these samples, increased Alu mutation rate was associated with greater exposure to PM2·5 (r=0·26, p<0·0001) and black carbon (r=0·33, p<0·0001), but not NO2. Promoter methylation was positively associated with PM2·5 in APEX1 (7·34%, 95% CI 0·52 to 14·16, p=0·009), OGG1 (13·06, 3·88 to 22·24, p=0·005), ERCC4 (16·31%, 5·43 to 27·18, p=0·01), and p53 (10·60%, 4·46 to 16·74, p=0·01), whereas promoter methylation of DAPK1 (-12·92%, -22·35 to -3·49, p=0·007) was inversely associated with PM2·5 exposure. Black carbon exposure was associated with elevated promoter methylation in APEX1 (9·16%, 4·06 to 14·25, p=0·01) and ERCC4 (27·56%, 17·58 to 37·55, p<0·0001). Promoter methylation was not associated with pollutant exposure in PARP1 and ERCC1, and NO2 exposure was not associated with methylation in any of the genes studied. INTERPRETATION Transplacental in-utero exposure to particulate matter is associated with an increased overall placental mutation rate (as measured with Alu), which occurred in concert with epigenetic alterations in key DNA repair and tumour suppressor genes. Our results suggest that exposure to air pollution can induce changes to fetal and neonatal DNA repair capacity. Future studies will be essential to elucidate whether these changes persist and have a role in carcinogenic insults later in life. FUNDING European Research Council and the Flemish Scientific Fund.
Collapse
Affiliation(s)
- Kristof Y Neven
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Nelly D Saenen
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Letitzia Tarantini
- EPIGET-Epidemiology, Epigenetics and Toxicology Lab, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Bram G Janssen
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | | | | | - Valentina Bollati
- EPIGET-Epidemiology, Epigenetics and Toxicology Lab, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium; Environment & Health unit, Leuven University, Leuven, Belgium.
| |
Collapse
|
204
|
Feng Q, Tian T, Liu J, Zhang L, Qi J, Lin X. Deregulation of microRNA‑31a‑5p is involved in the development of primary hypertension by suppressing apoptosis of pulmonary artery smooth muscle cells via targeting TP53. Int J Mol Med 2018; 42:290-298. [PMID: 29620173 PMCID: PMC5979825 DOI: 10.3892/ijmm.2018.3597] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/09/2018] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to identify the association between microRNA (miRNA/miR)-31a-5p and the development of hypertension, and its potential molecular mechanism. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analyses were performed to validate the candidate miRNA and genes involved in hypertension, following which an online miRNA database search, luciferase assay, and RT-qPCR and western blot analyses were performed to confirm the interaction between miR-31a-5p and TP53. A MTT assay and flow cytometric analysis were utilized to determine the effect of miR-31a-5p on cell growth and apoptosis. The results revealed that miR-31a-5p and TP53 were the candidate miRNA and gene regulating hypertension, and that TP53 was the virtual target gene of miR-31a-5p with a binding site located in the TP53 3′ untranslated region (3′UTR). It was confirmed by luciferase activity that miR-31a-5p markedly reduced the luciferase activity of the Luc-wild-type-TP53-3′UTR, whereas the mutated putative miR-31a-5p binding located on the TP53-3′UTR was found to eliminate such an inhibitory effect. miR-31a-5p had no effect on specificity protein 1, E2F transcription factor 2 or forkhead box P3 luciferase activity. Smooth muscle cells collected from spontaneously hypertensive rats treated with gold nano-particles containing anti-rno-miR-31a-5p exhibited a lower growth rate and a higher apoptotic rate. The results of the RT-qPCR and western blot analyses showed that miR-31a-5p negatively regulated the expression of TP53, and transfection with the hsa-miR-31a-5p mimic significantly promoted cell growth and inhibited cell apoptosis, whereas transfection with the anti-hsa-miR-31a-5p mimic significantly suppressed cell growth and induced cell apoptosis. Taken together, these findings indicated that miR-31a-5p is involved in hypertension via the accelerated proliferation of arterial smooth muscle cells and inhibition of apoptosis through targeting TP53.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Laboratory, The People's Hospital of Tongchuan, Tongchuan, Shaanxi 727000, P.R. China
| | - Tao Tian
- Department of Laboratory, Second Affiliated Hospital of Shaanxi Chinese Traditional Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Junfeng Liu
- Department of Infection, The People's Hospital of Tongchuan, Tongchuan, Shaanxi 727000, P.R. China
| | - Li Zhang
- Department of Gynecology and Obstetrics, The People's Hospital of Tongchuan, Tongchuan, Shaanxi 727000, P.R. China
| | - Jiangang Qi
- Department of Laboratory, Tongchuan Hospital of Chinese Traditional Medicine, Tongchuan, Shaanxi 727000, P.R. China
| | - Xiaojuan Lin
- Department of Cardiology, The People's Hospital of Tongchuan, Tongchuan, Shaanxi 727000, P.R. China
| |
Collapse
|
205
|
Abstract
Mice that have homozygous deletion of the p53 tumor suppressor protein universally die of malignancy, generally before 6 months of age. We show that hemizygous deficiency of RALBP1 (RLIP76 or Rlip) confers a degree of protection from spontaneous malignancy that has never previously been observed. This discovery introduces a paradigm for p53 function, in which Rlip plays a central role as an effector that appears necessary for the cancer susceptibility of p53 null mice. Because p53 loss has a powerful effect on genomic instability that contributes to the initiation and promotion of cancers and to drug and radiation resistance in humans, our findings provide a method for prevention and therapy of p53-deficient cancer. TP53 (p53) is a tumor suppressor whose functions are lost or altered in most malignancies. p53 homozygous knockout (p53−/−) mice uniformly die of spontaneous malignancy, typically T-cell lymphoma. RALBP1 (RLIP76, Rlip) is a stress-protective, mercapturic acid pathway transporter protein that also functions as a Ral effector involved in clathrin-dependent endocytosis. In stark contrast to p53−/− mice, Rlip−/− mice are highly resistant to carcinogenesis. We report here that partial Rlip deficiency induced by weekly administration of an Rlip-specific phosphorothioate antisense oligonucleotide, R508, strongly inhibited spontaneous as well as benzo(a)pyrene-induced carcinogenesis in p53−/− mice. This treatment effectively prevented large-scale methylomic and transcriptomic abnormalities suggestive of inflammation found in cancer-bearing p53−/− mice. The remarkable efficiency with which Rlip deficiency suppresses spontaneous malignancy in p53−/− mice has not been observed with any previously reported pharmacologic or genetic intervention. These findings are supported by cross-breeding experiments demonstrating that hemizygous Rlip deficiency also reduces the spontaneous malignancy phenotype of p53+/− mice. Rlip is found on the cell surface, and antibodies directed against Rlip were found to inhibit growth and promote apoptosis of cell lines as effectively as Rlip siRNA. The work presented here investigates several features, including oxidative DNA damage of the Rlip–p53 association in malignant transformation, and offers a paradigm for the mechanisms of tumor suppression by p53 and the prospects of suppressing spontaneous malignancy in hereditary cancer syndromes such as Li-Fraumeni.
Collapse
|
206
|
Nikitakis NG, Rassidakis GZ, Tasoulas J, Gkouveris I, Kamperos G, Daskalopoulos A, Sklavounou A. Alterations in the expression of DNA damage response-related molecules in potentially preneoplastic oral epithelial lesions. Oral Surg Oral Med Oral Pathol Oral Radiol 2018; 125:637-649. [PMID: 29705090 DOI: 10.1016/j.oooo.2018.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/04/2018] [Accepted: 03/06/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the expression levels of DNA damage response (DDR) markers in potentially preneoplastic oral epithelial lesions (PPOELs). STUDY DESIGN Immunohistochemical expression of DDR markers (γΗ2 ΑΧ, pChk2, 53 BP1, p53, and phosphorylated at Ser 15 p53) was assessed in 41 oral leukoplakias, ranging from hyperplasia (H) to dysplasia (D) and in comparison with oral squamous cell carcinoma (OSCC) and normal mucosa (NM). Statistical and receiver operating characteristic curve analysis were performed. RESULTS γH2 AX immunoexpression demonstrated a gradual increase and upper layer extension from NM to H to higher D degrees to OSCC. pChk2 expression was minimal in NM, relatively low in PPOELs, with an increasing tendency from H to D, and higher in OSCC. 53 BP1 demonstrated higher levels in OSCC than in NM, whereas its expression in PPOELs was heterogeneous, gradually increasing according to D. p53 demonstrated progressively higher levels and upper layer extension from H to D to OSCC. Phosphorylated p53 was absent in NM and relatively low in PPOELs and OSCC. CONCLUSIONS DDR markers' expression is variable in PPOELs, showing a tendency to increase along with dysplasia. Activated DDR mechanisms may play an important protective role at early stages of oral carcinogenesis, but probably suffer progressive deregulation, eventually failing to suppress malignant transformation.
Collapse
Affiliation(s)
- Nikolaos G Nikitakis
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Greece.
| | | | - Jason Tasoulas
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Ioannis Gkouveris
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Greece; Division of Diagnostics and Surgical Sciences, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Georgios Kamperos
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Greece
| | - Argyrios Daskalopoulos
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Greece
| | - Alexandra Sklavounou
- Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
207
|
Profiling of Germline Mutations in Major Hotspot Codons of TP53 Using PCR-RFLP. Pathol Oncol Res 2018; 25:1373-1377. [PMID: 29500733 DOI: 10.1007/s12253-018-0394-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/21/2018] [Indexed: 01/10/2023]
Abstract
Tumor suppressor protein, TP53 also known as the "guardian of the genome" plays a key role in preventing malignant transformation. Almost 50% of human tumors carry mutations in this gene; in the remaining tumors, the TP53 network is functionally inoperative. The majority of TP53 mutations are missense mutations and more than 90% of the missense mutations affect specific codons in the DNA-binding domain, called "hotspot codons." The present study was aimed at analyzing the germline mutation status of four hotspot codons in TP53 namely, codon 175, codon 245, codon 248 (within the DNA binding domain) and codon 72 (outside the DNA binding domain) in cancer cases encountered in a tertiary care hospital in South India by PCR-RFLP. The case-control study included 85-10 subjects respectively. The results of the study indicated that majority of the cancer cases did not harbor germline mutations in the four hot spot codons of TP53. The study further highlights the usefulness of PCR-RFLP as a simple and cost effective tool for checking gene mutations.
Collapse
|
208
|
Mihalyova J, Jelinek T, Growkova K, Hrdinka M, Simicek M, Hajek R. Venetoclax: A new wave in hematooncology. Exp Hematol 2018; 61:10-25. [PMID: 29477371 DOI: 10.1016/j.exphem.2018.02.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/31/2018] [Accepted: 02/05/2018] [Indexed: 12/22/2022]
Abstract
Inhibitors of antiapoptotic proteins of the BCL2 family can successfully restart the deregulated process of apoptosis in malignant cells. Whereas nonselective agents have been limited by their affinity to different BCL2 members, thus inducing excessive toxicity, the highly selective BCL2 inhibitor venetoclax (ABT-199, Venclexta™) has an acceptable safety profile. To date, it has been approved in monotherapy for the treatment of relapsed or refractory chronic lymphocytic leukemia (CLL) with 17p deletion. Extension of indications can be expected in monotherapy and in combination regimens. Sensitivity to venetoclax is not common in lymphomas, but promising outcomes have been achieved in the mantle cell lymphoma group. Venetoclax is also active in multiple myeloma patients, especially in those with translocation t(11;14), even if high-risk features such as del17p are also present. Surprisingly, positive results are being obtained in elderly acute myeloid leukemia patients, in whom inhibition of BCL2 is able to substantially increase the efficacy of low-dose cytarabine or hypomethylating agents. Here, we provide a summary of available results from clinical trials and describe a specific mechanism of action that stands behind the efficacy of venetoclax in hematological malignancies.
Collapse
Affiliation(s)
- Jana Mihalyova
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic; Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Tomas Jelinek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic; Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic; Faculty of Science, University of Ostrava, Ostrava, Czech Republic.
| | - Katerina Growkova
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic; Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic; Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Matous Hrdinka
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic; Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Michal Simicek
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic; Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Roman Hajek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic; Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
209
|
Chira S, Gulei D, Hajitou A, Berindan-Neagoe I. Restoring the p53 'Guardian' Phenotype in p53-Deficient Tumor Cells with CRISPR/Cas9. Trends Biotechnol 2018; 36:653-660. [PMID: 29478674 DOI: 10.1016/j.tibtech.2018.01.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 12/26/2022]
Abstract
With an increasing prevalence in the human population, cancer has become one of the most investigated fields of medicine. Among the potential targets for cancer therapy is the tumor suppressor gene TP53, which is found in a mutated state in approximately 50% of human cancers and is often associated with poor prognosis. We propose a novel, highly tumor-specific delivery system for TP53, based on the CRISPR/Cas9 genome editing technology. This system will restore the normal p53 phenotype in tumor cells by replacing the mutant TP53 gene with a functional copy, leading to sustained expression of p53 protein and tumor regression.
Collapse
Affiliation(s)
- Sergiu Chira
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.
| | - Diana Gulei
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Amin Hajitou
- Cancer Phage Therapy Group, Division of Brain Sciences, Imperial College London, W12 0NN London, UK
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; Department of Functional Genomics and Experimental Pathology, Oncology Institute Prof. Dr. Ion Chiricuta, 400015 Cluj-Napoca, Romania
| |
Collapse
|
210
|
Cheng HW, Chein RJ, Cheng TJ, Wu PS, Wu HY, Hung PF, Wang CJ, Hsu YL, Wong JM, Yuan A, Wong CH, Yang PC, Pan SH. 2-anilino-4-amino-5-aroylthiazole-type compound AS7128 inhibits lung cancer growth through decreased iASPP and p53 interaction. Cancer Sci 2018; 109:832-842. [PMID: 29285847 PMCID: PMC5834782 DOI: 10.1111/cas.13489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/20/2017] [Accepted: 12/25/2017] [Indexed: 12/17/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Thus, developing novel therapeutic agents has become critical for lung cancer treatment. In this study, compound AS7128 was selected from a 2-million entry chemical library screening and identified as a candidate drug against non-small cell lung cancer in vitro and in vivo. Further investigation indicated that AS7128 could induce cell apoptosis and cell cycle arrest, especially in the mitosis stage. In addition, we also found that iASPP, an oncogenic protein that functionally inhibits p53, might be associated with AS7128 through mass identification. Further exploration indicated that AS7128 treatment could restore the transactivation ability of p53 and, thus, increase the expressions of its downstream target genes, which are related to cell cycle arrest and apoptosis. This occurs through disruption of the interactions between p53 and iASPP in cells. Taken together, AS7128 could bind to iASPP, disrupt the interaction between iASPP and p53, and result in cell cycle arrest and apoptosis. These findings may provide new insight for using iASPP as a therapeutic target for non-small cell lung cancer treatment.
Collapse
Affiliation(s)
- Hao-Wei Cheng
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | | | - Ting-Jen Cheng
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Pei-Shan Wu
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan
| | - Hsin-Yi Wu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Pei-Fang Hung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-Jen Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yuan-Ling Hsu
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
| | - Jau-Min Wong
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ang Yuan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Pan-Chyr Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Szu-Hua Pan
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan.,Doctoral Degree Program of Translational Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
211
|
Cruz-Gregorio A, Manzo-Merino J, Lizano M. Cellular redox, cancer and human papillomavirus. Virus Res 2018; 246:35-45. [DOI: 10.1016/j.virusres.2018.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/10/2018] [Accepted: 01/10/2018] [Indexed: 12/28/2022]
|
212
|
Kim DW, Kim KC, Kim KB, Dunn CT, Park KS. Transcriptional deregulation underlying the pathogenesis of small cell lung cancer. Transl Lung Cancer Res 2018. [PMID: 29535909 DOI: 10.21037/tlcr.2017.10.07] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The discovery of recurrent alterations in genes encoding transcription regulators and chromatin modifiers is one of the most important recent developments in the study of the small cell lung cancer (SCLC) genome. With advances in models and analytical methods, the field of SCLC biology has seen remarkable progress in understanding the deregulated transcription networks linked to the tumor development and malignant progression. This review will discuss recent discoveries on the roles of RB and P53 family of tumor suppressors and MYC family of oncogenes in tumor initiation and development. It will also describe the roles of lineage-specific factors in neuroendocrine (NE) cell differentiation and homeostasis and the roles of epigenetic alterations driven by changes in NFIB and chromatin modifiers in malignant progression and chemoresistance. These recent findings have led to a model of transcriptional network in which multiple pathways converge on regulatory regions of crucial genes linked to tumor development. Validation of this model and characterization of target genes will provide critical insights into the biology of SCLC and novel strategies for tumor intervention.
Collapse
Affiliation(s)
- Dong-Wook Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Keun-Cheol Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA.,Department of Biological Sciences, Kangwon National University, Chuncheon, Korea
| | - Kee-Beom Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Colin T Dunn
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Kwon-Sik Park
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
213
|
Bhattacharya B, Low SHH, Chong ML, Chia D, Koh KX, Sapari NS, Kaye S, Hung H, Benoukraf T, Soong R. Acquired resistance to combination treatment through loss of synergy with MEK and PI3K inhibitors in colorectal cancer. Oncotarget 2018; 7:29187-98. [PMID: 27081080 PMCID: PMC5045388 DOI: 10.18632/oncotarget.8692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/28/2016] [Indexed: 12/15/2022] Open
Abstract
Historically, understanding of acquired resistance (AQR) to combination treatment has been based on knowledge of resistance to its component agents. To test whether an altered drug interaction could be an additional factor in AQR to combination treatment, models of AQR to combination and single agent MEK and PI3K inhibitor treatment were generated. Combination indices indicated combination treatment of PI3K and MEK inhibitors remained synergistic in cells with AQR to single agent but not combination AQR cells. Differences were also observed between the models in cellular phenotypes, pathway signaling and drug cross-resistance. Genomics implicated TGFB2-EDN1 overexpression as candidate determinants in models of AQR to combination treatment. Supplementation of endothelin in parental cells converted synergism to antagonism. Silencing of TGFB2 or EDN1 in cells with AQR conferred synergy between PI3K and MEK inhibitor. These results highlight that AQR to combination treatment may develop through alternative mechanisms to those of single agent treatment, including a change in drug interaction.
Collapse
Affiliation(s)
- Bhaskar Bhattacharya
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Sarah Hong Hui Low
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Mei Ling Chong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Dilys Chia
- Department of Pharmacy, National University of Singapore, Singapore
| | - King Xin Koh
- Department of Pathology, National University of Singapore, Singapore
| | - Nur Sabrina Sapari
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Stanley Kaye
- Drug Development Unit, Royal Marsden NHS Trust, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Huynh Hung
- Laboratory of Molecular Endocrinology, National Cancer Centre of Singapore, Singapore
| | - Touati Benoukraf
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pathology, National University of Singapore, Singapore
| |
Collapse
|
214
|
Kong L, Murata MM, Digman MA. Absence of REV3L promotes p53-regulated cancer cell metabolism in cisplatin-treated lung carcinoma cells. Biochem Biophys Res Commun 2018; 496:199-204. [PMID: 29307819 DOI: 10.1016/j.bbrc.2018.01.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 11/25/2022]
Abstract
Lung cancer is one of the deadliest cancers in the world because of chemo-resistance to the commonly used cisplatin-based treatments. The use of low fidelity DNA polymerases in the translesional synthesis (TLS) DNA damage response pathway that repairs lesions caused by cisplatin also presents a mutational carcinogenic burden on cells that needs to be regulated by the tumor suppressor protein p53. However, there is much debate over the roles of the reversionless 3-like (REV3L) protein responsible for TLS and p53 in regulating cancer cell metabolism. In this study, the fluorescence lifetime of the metabolic coenzyme NADH reveals that the absence of REV3L can promote the p53-mediated upregulation of oxidative phosphorylation in cisplatin-treated H1299 lung carcinoma cells and increases cancer cell sensitivity to this platinum-based chemotherapy. These results demonstrate a previously unrecognized relationship between p53 and REV3L in cancer cell metabolism and may lead to improvements in chemotherapy treatment plans that reduce cisplatin resistance in lung cancer.
Collapse
Affiliation(s)
- Linghao Kong
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA; University High School, Irvine, CA 92612, USA
| | - Michael M Murata
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Michelle A Digman
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
215
|
|
216
|
Zhou X, Cao CY, Wan ATY, Yue GGL, Kwok FHF, Fung KP, Sun H, Lau CBS, Puno PT, Tsui SKW. Functional roles of eriocalyxin B in zebrafish revealed by transcriptome analysis. Mol Omics 2018; 14:156-169. [PMID: 29676772 DOI: 10.1039/c7mo00125h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Eriocalyxin B (EriB) is a naturalent-kaurane diterpenoid obtained fromIsodon eriocalyxvar.laxiflora(family Lamiaceae), which exerted multiple biological activities (e.g.anti-tumor and anti-inflammatory)viathe alteration of gene expression and signaling transduction.
Collapse
Affiliation(s)
- Xunian Zhou
- School of Biomedical Sciences
- The Chinese University of Hong Kong
- China
- Institute of Chinese Medicine
- The Chinese University of Hong Kong
| | - Cyanne Ye Cao
- School of Biomedical Sciences
- The Chinese University of Hong Kong
- China
| | - Angel Tsz-Yau Wan
- School of Biomedical Sciences
- The Chinese University of Hong Kong
- China
| | - Grace Gar-Lee Yue
- Institute of Chinese Medicine
- The Chinese University of Hong Kong
- China
- State Key Laboratory of Phytochemistry and Plant Resources in West China
- The Chinese University of Hong Kong
| | - Frankie Hin-Fai Kwok
- Institute of Chinese Medicine
- The Chinese University of Hong Kong
- China
- State Key Laboratory of Phytochemistry and Plant Resources in West China
- The Chinese University of Hong Kong
| | - Kwok-Pui Fung
- School of Biomedical Sciences
- The Chinese University of Hong Kong
- China
- Institute of Chinese Medicine
- The Chinese University of Hong Kong
| | - Handong Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China
- Kunming Institute of Botany
- Chinese Academy of Sciences
- Kunming 650201
- China
| | - Clara Bik-San Lau
- Institute of Chinese Medicine
- The Chinese University of Hong Kong
- China
- State Key Laboratory of Phytochemistry and Plant Resources in West China
- The Chinese University of Hong Kong
| | - Pema-Tenzin Puno
- State Key Laboratory of Phytochemistry and Plant Resources in West China
- Kunming Institute of Botany
- Chinese Academy of Sciences
- Kunming 650201
- China
| | | |
Collapse
|
217
|
Fann LY, Chen Y, Chu DC, Weng SJ, Chu HC, Wu ATH, Lee JF, Ali AAA, Chen TC, Huang HS, Ma KH. Identification and preclinical evaluation of the small molecule, NSC745887, for treating glioblastomas via suppressing DcR3-associated signaling pathways. Oncotarget 2017; 9:11922-11937. [PMID: 29552282 PMCID: PMC5844718 DOI: 10.18632/oncotarget.23714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/11/2017] [Indexed: 11/25/2022] Open
Abstract
The small-molecule naphtha [2,3-f]quinoxaline-7,12-dione (NSC745887) can effectively inhibit the proliferation of various cancers by trapping DNA-topoisomerase cleavage. The aim of this study was to elucidate cellular responses of NSC745887 in human glioblastoma multiforme (GBM, U118MG and U87MG cells) and investigate the underlying molecular mechanisms. NSC745887 reduced the cell survival rate and increased the sub-G1 population in dose- and time-dependent manners in GBM cells. Moreover, NSC745887 increased expression of γH2AX and caused DNA fragmentation leading to DNA damage. Furthermore, Annexin V/propidium iodide and Br-dTP staining showed the apoptotic effect of NSC745887 in GBM cells. DNA repair proteins of ataxia-telangiectasia mutated (ATM), ATM and Rad3-related, and decoy receptor 3 also decreased with NSC745887 treatment. In addition, NSC745887 caused apoptosis by the caspase-8/9-caspase-3-poly(ADP-ribose) polymerase cascade. An in vivo study indicated that NSC745887 suppressed the [18F]-FDG-specific uptake value in brain tumors. Histological staining also indicated a decrease in Ki-67 and increases in γH2AX and cleaved caspase-3 in the brain tumor area. These data provide preclinical evidence for NSC745887 as a potential new small molecule drug for managing glioblastomas.
Collapse
Affiliation(s)
- Li-Yun Fann
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Nursing and Department of Neurosurgery, Taipei City Hospital, Taipei, Taiwan, ROC.,Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Ying Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Da-Chen Chu
- Department of Nursing and Department of Neurosurgery, Taipei City Hospital, Taipei, Taiwan, ROC
| | - Shao-Ju Weng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Heng-Cheng Chu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Alexander T H Wu
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Jiann-Fong Lee
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Ahmed Atef Ahmed Ali
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Tsung-Chih Chen
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hsu-Shan Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC.,Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Kuo-Hsing Ma
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
218
|
Revisiting tumor patterns and penetrance in germline TP53 mutation carriers: temporal phases of Li-Fraumeni syndrome. Curr Opin Oncol 2017; 30:23-29. [PMID: 29076966 DOI: 10.1097/cco.0000000000000423] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Germline pathogenic TP53 mutation may predispose to multiple cancers but penetrance and cancer patterns remain incompletely documented. We have analyzed international agency for research on cancer TP53 database to reevaluate age and variant-dependent tumor patterns. RECENT FINDINGS Genome-wide studies suggest that germline variants are more frequent than estimated prevalence of Li-Fraumeni syndrome (LFS), suggesting that many carriers of potentially pathogenic mutations may not develop the syndrome. Carriers of a germline TP53 mutation who are detected in a clinical context have a penetrance of 80% at age 70. Penetrance varies according to age, sex and mutation type. Temporal tumor patterns show distinct phases, with childhood phase (0-15 years, 22% of all cancers) characterized by adrenal cortical carcinoma, choroid plexus carcinoma, rhabdomyosarcoma and medulloblastoma; early adulthood phase (16-50 years, 51%) including breast cancer, osteosarcoma, soft tissue sarcomas, leukemia, astrocytoma and glioblastoma, colorectal and lung cancer; late adulthood phase (51-80 years, 27%) including pancreatic and prostate cancer. SUMMARY Germline pathogenic variants in TP53 gene have different consequences according to cell, tissue, context and age. The occurrence of frequent variants in patients with no criteria suggestive of LFS calls for attention in predicting individual risk and highlights the need of additional predictors for assigning carriers to appropriate surveillance programs.
Collapse
|
219
|
Guo KY, Han L, Li X, Yang AV, Lu J, Guan S, Li H, Yu Y, Zhao Y, Yang J, Zhang H. Novel proteasome inhibitor delanzomib sensitizes cervical cancer cells to doxorubicin-induced apoptosis via stabilizing tumor suppressor proteins in the p53 pathway. Oncotarget 2017; 8:114123-114135. [PMID: 29371974 PMCID: PMC5768391 DOI: 10.18632/oncotarget.23166] [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] [Received: 10/17/2017] [Accepted: 11/28/2017] [Indexed: 02/06/2023] Open
Abstract
Cervical cancer, the third most commonly occurring cancer, is the second leading cause of cancer related mortality among women. Aberrant ubiquitination and proteasome activity, both human papillomavirus and tumor derived, have been shown to contribute to tumor angiogenesis, proliferation, and invasion in many cancers, including cervical cancer. Thus, small molecule proteasome inhibitors are a potential and strategic treatment option for cervical cancer. In this study, novel proteasome inhibitor delanzomib (CEP-18770) exhibited potent pro-apoptotic and cytotoxic effects on a panel of cervical cancer cell lines by blocking proteasomal activity. Delanzomib also significantly sensitized cervical cancer cells to treatment of doxorubicin (Dox), a traditional chemotherapeutic agent. Furthermore, proteasome inhibition revealed stabilization of p53 and p53 transcriptional targets and induction of p38/JNK phosphorylation. Additionally, delanzomib worked synergistically with Dox to further upregulate p53 and its downstream targets and enhanced Dox-induced p38 phosphorylation. Our study strongly supports the 26S proteasome as a potential therapeutic target in cervical cancer and proteasome inhibition by delanzomib may be a potential treatment strategy for cervical cancer patients.
Collapse
Affiliation(s)
- Kevin Y Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lili Han
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Gynecology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830001, China
| | - Xinyu Li
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrew V Yang
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jiaxiong Lu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shan Guan
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui Li
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yang Yu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yanling Zhao
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianhua Yang
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hong Zhang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| |
Collapse
|
220
|
Cheng Y, Li Y, Ma C, Song Y, Xu H, Yu H, Xu S, Mu Q, Li H, Chen Y, Zhao G. Arsenic trioxide inhibits glioma cell growth through induction of telomerase displacement and telomere dysfunction. Oncotarget 2017; 7:12682-92. [PMID: 26871293 PMCID: PMC4914314 DOI: 10.18632/oncotarget.7259] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/24/2016] [Indexed: 12/22/2022] Open
Abstract
Glioblastomas are resistant to many kinds of treatment, including chemotherapy, radiation and other adjuvant therapies. As2O3 reportedly induces ROS generation in cells, suggesting it may be able to induce telomerase suppression and telomere dysfunction in glioblastoma cells. We show here that As2O3 induces ROS generation as well as telomerase phosphorylation in U87, U251, SHG4 and C6 glioma cells. It also induces translocation of telomerase from the nucleus to the cytoplasm, thereby decreasing total telomerase activity. These effects of As2O3 trigger an extensive DNA damage response at the telomere, which includes up-regulation of ATM, ATR, 53BP1, γ-H2AX and Mer11, in parallel with telomere fusion and 3′-overhang degradation. This ultimately results in induction of p53- and p21-mediated cell apoptosis, G2/M cell cycle arrest and cellular senescence. These results provide new insight into the antitumor effects of As2O3 and can perhaps contribute to solving the problem of glioblastoma treatment resistance.
Collapse
Affiliation(s)
- Ye Cheng
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Yunqian Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Chengyuan Ma
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Yang Song
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Haiyang Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Hongquan Yu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Songbai Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Qingchun Mu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Haisong Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Yong Chen
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| | - Gang Zhao
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P. R. China
| |
Collapse
|
221
|
Shakambari G, Sameer Kumar R, Ashokkumar B, Varalakshmi P. Agro Waste Utilization for Cost-Effective Production of l-Asparaginase by Pseudomonas plecoglossicida RS1 with Anticancer and Acrylamide Mitigation Potential. ACS OMEGA 2017; 2:8108-8117. [PMID: 30023574 PMCID: PMC6044496 DOI: 10.1021/acsomega.7b01429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 10/17/2017] [Indexed: 05/06/2023]
Abstract
Agricultural wastes such as the peels of onion and garlic were used as a supplement along with l-asparagine for the very first time to produce increased yield of l-asparaginase by Pseudomonas plecoglossicida RS1. Statistical optimization strategies such as response surface methodology were used to generate a medium composition containing extracts of 0.9 (v/v) of garlic peel waste and 0.5% (v/v) onion peel waste along with 0.2% (w/w) l-asparagine, which yielded a twofold increase in the enzyme activity compared to the unsupplemented minimal (M-9) medium. The presence of l-asparagine content in the peel extract was confirmed by high-performance liquid chromatography. Further, l-asparaginase was purified to homogeneity, and identity was confirmed by matrix-assisted laser desorption ionization time-of-flight analysis. The application of the purified l-asparaginase as a therapeutic was studied in HeLa cells which showed a p53-mediated G2 cell cycle arrest. Moreover, the purified l-asparaginase showed effective acrylamide mitigation in vitro, at 6 IU, and its effective degradation was also demonstrated by the effect on chemotactic index of Caenorhabditis elegans and the restoration of the cognitive abilities of C. elegans which was coexposed to acrylamide and l-asparaginase compared to that exposed to acrylamide alone. Thus, l-asparaginase, with multipotent applications, was produced by effective waste utilization for economical commercial production.
Collapse
Affiliation(s)
- Ganeshan Shakambari
- Department
of Molecular Microbiology, School of Biotechnology, and Department of
Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, Tamil Nadu 625021, India
| | - Rai Sameer Kumar
- Department
of Molecular Microbiology, School of Biotechnology, and Department of
Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, Tamil Nadu 625021, India
| | - Balasubramaniem Ashokkumar
- Department
of Molecular Microbiology, School of Biotechnology, and Department of
Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, Tamil Nadu 625021, India
| | - Perumal Varalakshmi
- Department
of Molecular Microbiology, School of Biotechnology, and Department of
Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, Tamil Nadu 625021, India
- E-mail: (P.V.)
| |
Collapse
|
222
|
Mitkin NA, Muratova AM, Sharonov GV, Korneev KV, Sviriaeva EN, Mazurov D, Schwartz AM, Kuprash DV. p63 and p73 repress CXCR5 chemokine receptor gene expression in p53-deficient MCF-7 breast cancer cells during genotoxic stress. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:1169-1178. [PMID: 29107083 DOI: 10.1016/j.bbagrm.2017.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/02/2017] [Accepted: 10/24/2017] [Indexed: 12/17/2022]
Abstract
Many types of chemotherapeutic agents induce of DNA-damage that is accompanied by activation of p53 tumor suppressor, a key regulator of tumor development and progression. In our previous study we demonstrated that p53 could repress CXCR5 chemokine receptor gene in MCF-7 breast cancer cells via attenuation of NFkB activity. In this work we aimed to determine individual roles of p53 family members in the regulation of CXCR5 gene expression under genotoxic stress. DNA-alkylating agent methyl methanesulfonate caused a reduction in CXCR5 expression not only in parental MCF-7 cells but also in MCF-7-p53off cells with CRISPR/Cas9-mediated inactivation of the p53 gene. Since p53 knockout was associated with elevated expression of its p63 and p73 homologues, we knocked out p63 using CRISPR/Cas9 system and knocked down p73 using specific siRNA. The CXCR5 promoter activity, CXCR5 expression and CXCL13-directed migration in MCF-7 cells with inactivation of all three p53 family genes were completely insensitive to genotoxic stress, while pairwise p53+p63 or p53+p73 inactivation resulted in partial effects. Using deletion analysis and site-directed mutagenesis, we demonstrated that effects of NFkB on the CXCR5 promoter inversely correlated with p63 and p73 levels. Thus, all three p53 family members mediate the effects of genotoxic stress on the CXCR5 promoter using the same mechanism associated with attenuation of NFkB activity. Understanding of this mechanism could facilitate prognosis of tumor responses to chemotherapy.
Collapse
Affiliation(s)
- Nikita A Mitkin
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia
| | - Alisa M Muratova
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia; Department of Immunology, Lomonosov Moscow State University, Leninskye gory 1, 119234 Moscow, Russia
| | - George V Sharonov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; Faculty of Medicine, Lomonosov Moscow State University, Leninskye gory 1, 119234 Moscow, Russia
| | - Kirill V Korneev
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia; Department of Immunology, Lomonosov Moscow State University, Leninskye gory 1, 119234 Moscow, Russia
| | - Ekaterina N Sviriaeva
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia
| | - Dmitriy Mazurov
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., Moscow 119334, Russia
| | - Anton M Schwartz
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia
| | - Dmitry V Kuprash
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia; Department of Immunology, Lomonosov Moscow State University, Leninskye gory 1, 119234 Moscow, Russia.
| |
Collapse
|
223
|
Wu M, Ye H, Tang Z, Shao C, Lu G, Chen B, Yang Y, Wang G, Hao H. p53 dynamics orchestrates with binding affinity to target genes for cell fate decision. Cell Death Dis 2017; 8:e3130. [PMID: 29048401 PMCID: PMC5682658 DOI: 10.1038/cddis.2017.492] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/27/2017] [Accepted: 08/29/2017] [Indexed: 01/15/2023]
Abstract
Emerging evidence support that temporal dynamics is pivotal for signaling molecules in orchestrating smart responses to diverse stimuli. p53 is such a signaling molecule that employs temporal dynamics for the selective activation of downstream target genes and ultimately for cell fate decision. Yet how this fine-tuned p53 machinery is quantitatively decoded remains largely unclear. Here we report a quantitative mechanism defining how p53 dynamics orchestrates with binding affinity to target genes for cell fate decision. Treating cells with a genotoxic drug doxorubicin at various doses and durations, we found that a mild and prolonged challenge triggered sequential p53 pulses and ultimately resulted in a terminal pulse enacting apoptosis in a comparable rate with that induced by an acute and high-dose treatment. To transactivate proapoptotic genes and thereafter executing apoptosis, p53 must exceed a certain threshold and accumulate for sufficient time at levels above it. Effective cumulative levels above the threshold, defined as E∫p53, but not the total accumulation levels of p53, precisely discriminate survival and apoptotic cells. p53 accumulation below this threshold, even with prolonging time to reach a total level comparable to that from the accumulation over the threshold, could not transactivate proapoptotic genes to which the binding affinity of p53 is lower than that of proarrest genes, and this property is independent of dynamic features. Our findings indicate that the dynamic feature per se does not directly control cell fate, but rather it orchestrates with the binding affinity to target genes to confer an appropriate time window for cell fate choice. Our study provides a quantitative mechanism unifying p53 dynamics and binding affinity to target genes, providing novel insights to understand how p53 can respond quantitatively to chemotherapeutic drugs, and guiding the design of metronomic regimens for chemotherapeutic drugs.
Collapse
Affiliation(s)
- Mengqiu Wu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang #24, Nanjing, Jiangsu 210009, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Hui Ye
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang #24, Nanjing, Jiangsu 210009, China
| | - Zhiyuan Tang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Chang Shao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang #24, Nanjing, Jiangsu 210009, China
| | - Gaoyuan Lu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang #24, Nanjing, Jiangsu 210009, China
| | - Baoqiang Chen
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang #24, Nanjing, Jiangsu 210009, China
| | - Yuyu Yang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang #24, Nanjing, Jiangsu 210009, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang #24, Nanjing, Jiangsu 210009, China
| | - Haiping Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang #24, Nanjing, Jiangsu 210009, China
| |
Collapse
|
224
|
Inflammation and the chemical carcinogen benzo[a]pyrene: Partners in crime. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 774:12-24. [DOI: 10.1016/j.mrrev.2017.08.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/02/2017] [Accepted: 08/19/2017] [Indexed: 12/12/2022]
|
225
|
Sabapathy K, Lane DP. Therapeutic targeting of p53: all mutants are equal, but some mutants are more equal than others. Nat Rev Clin Oncol 2017; 15:13-30. [DOI: 10.1038/nrclinonc.2017.151] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
226
|
Functional significance and therapeutic implication of ring-type E3 ligases in colorectal cancer. Oncogene 2017; 37:148-159. [PMID: 28925398 PMCID: PMC5770599 DOI: 10.1038/onc.2017.313] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/29/2017] [Accepted: 07/31/2017] [Indexed: 02/07/2023]
Abstract
Accumulative studies revealed that E3 ubiquitin ligases have important roles in colorectal carcinogenesis. The pathogenic mechanisms of colorectal cancer (CRC) initiation and progression are complex and heterogeneous, involving somatic mutations, abnormal gene fusion, deletion or amplification and epigenetic alteration, which may cause aberrant expression or altered function of E3 ligases in CRC. Defects of E3 ligases have been reported to be involved in the molecular etiology and pathogenesis of CRC. The aberrant expressed E3 ligases can function as either oncogenes or tumor suppressors depending on ubiquiting target substrates in CRC. Recently, considerable progress has been made in our understanding of the potential roles of E3 ligase-mediated ubiquitylation in colorectal carcinogenesis. There are mainly two subtypes of E3 ubiquitin ligases in humans, as defined by the presence of either a HECT domain or a RING finger domain on the basis of structural similitude. Most cancer-associated E3 ligases participate in regulating the cell cycle, apoptosis, gene transcription, cell signaling and DNA repair, the critical parts of CRC tumorigenesis. In this review, we have provided a comprehensive summary of abnormally expressed E3 ligases and their related pivotal mechanistic effects in CRC. In particular, we have highlighted the function of RING-type E3 ubiquitin enzymes in modulating cancer signaling pathways, immunity and tumor microenvironment in CRC development and progression; their mechanism(s) of action in CRC involving both ubiquitylation-dependent and ubiquitylation-independent effects; and the potential of RING E3 ligases as molecular biomarkers for predicting patient prognosis and as therapeutic targets in CRC. A better understanding of E3 ligase-mediated substrates' ubiquitylation involved in the development of CRC will provide new insights into the pathophysiology mechanisms of CRC, and unravel novel prognostic markers and therapeutic strategies for CRC.
Collapse
|
227
|
Spoletini M, Taurone S, Tombolini M, Minni A, Altissimi G, Wierzbicki V, Giangaspero F, Parnigotto PP, Artico M, Bardella L, Agostinelli E, Pastore FS. Trophic and neurotrophic factors in human pituitary adenomas (Review). Int J Oncol 2017; 51:1014-1024. [PMID: 28902350 DOI: 10.3892/ijo.2017.4120] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/17/2017] [Indexed: 11/06/2022] Open
Abstract
The pituitary gland is an organ that functionally connects the hypothalamus with the peripheral organs. The pituitary gland is an important regulator of body homeostasis during development, stress, and other processes. Pituitary adenomas are a group of tumors arising from the pituitary gland: they may be subdivided in functional or non-functional, depending on their hormonal activity. Some trophic and neurotrophic factors seem to play a key role in the development and maintenance of the pituitary function and in the regulation of hypothalamo-pituitary-adrenocortical axis activity. Several lines of evidence suggest that trophic and neurotrophic factors may be involved in pituitary function, thus suggesting a possible role of the trophic and neurotrophic factors in the normal development of pituitary gland and in the progression of pituitary adenomas. Additional studies might be necessary to better explain the biological role of these molecules in the development and progression of this type of tumor. In this review, in light of the available literature, data on the following neurotrophic factors are discussed: ciliary neurotrophic factor (CNTF), transforming growth factors β (TGF‑β), glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), vascular endothelial growth inhibitor (VEGI), fibroblast growth factors (FGFs) and epidermal growth factor (EGF) which influence the proliferation and growth of pituitary adenomas.
Collapse
Affiliation(s)
- Marialuisa Spoletini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, 'Sapienza' University of Rome, Rome, Italy
| | - Samanta Taurone
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Mario Tombolini
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Antonio Minni
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | | | | | - Felice Giangaspero
- Department of Radiology, Oncology and Anatomic Pathology, 'Sapienza' University of Rome, Rome, Italy
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (TES) Onlus, Padua, Italy
| | - Marco Artico
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Lia Bardella
- Department of Neurology and Psychiatry, 'Sapienza' University of Rome, Rome, Italy
| | - Enzo Agostinelli
- Department of Biochemical Sciences 'A. Rossi Fanelli', 'Sapienza' University of Rome, Rome, Italy
| | - Francesco Saverio Pastore
- Department of Systems' Medicine, Division of Neurosurgery, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
228
|
Rupp M, Hagenbuchner J, Rass B, Fiegl H, Kiechl-Kohlendorfer U, Obexer P, Ausserlechner MJ. FOXO3-mediated chemo-protection in high-stage neuroblastoma depends on wild-type TP53 and SESN3. Oncogene 2017; 36:6190-6203. [PMID: 28869600 PMCID: PMC5671944 DOI: 10.1038/onc.2017.288] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/21/2017] [Accepted: 07/13/2017] [Indexed: 12/12/2022]
Abstract
Forkhead box O class transcription factors are homeostasis regulators that control cell death, longevity and therapy-resistance. In neuroblastoma (NB), nuclear FOXO3 correlates with stage M disease and poor prognosis. To analyze whether FOXO3 contributes to drug-resistance in this childhood cancer, we investigated how different high-stage-derived NB cells respond to the activation of an ectopic FOXO3 allele. We found endogenous FOXO3 mostly localized to the nucleus—upon activation of an ectopic, 4OHT-activated FOXO3(A3)ER fusion protein two of the cell lines underwent apoptosis, whereas in the others FOXO3-activation even increased survival during drug-treatment. In the latter cell type, FOXO3 did not induce the BH3-only protein BCL2L11/BIM due to impaired binding of FOXO3 to the BIM-promoter, but still activated other FOXO3 targets. It was shown before that FOXO3 and TP53 physically interact with each other at two different regions—the TP53-N-terminus binds to the FOXO3-DNA binding domain (DBD) and the FOXO3-C-terminus interacts with the TP53-DBD. Interestingly, cell lines that undergo FOXO3-induced cell death carry homozygous point mutations in the TP53-DBD near the structural hotspot-mutation-site R175H, which abrogated FOXO3–TP53 interaction. In contrast, in FOXO3-death-resistant cells no point mutations in the TP53-DBD were found—in these cells FOXO3–TP53 complexes are formed and FOXO3-binding to the BIM-promoter, but not the induction of the detoxifying protein SESN3, were prevented, which in turn increased chemo-protection in this type of high-stage-derived NB cells. Our combined data suggest that FOXO3 steps in as a death inducer in case of TP53-mutation, whereas functional TP53 alters FOXO3-target-promoter-recognition, which prevents death induction by FOXO3 and instead increases chemo-protection and survival of NB cells. This novel mechanism may explain the low incidence of TP53 mutation in high-stage NB at diagnosis and suggests FOXO3 as a therapeutic target for this childhood malignancy.
Collapse
Affiliation(s)
- M Rupp
- Department of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria.,Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - J Hagenbuchner
- Department of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria
| | - B Rass
- Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria
| | - H Fiegl
- Department of Obstetrics and Gynecology, Medical University Innsbruck, Innsbruck, Austria
| | | | - P Obexer
- Department of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - M J Ausserlechner
- Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
229
|
Sotillo WS, Villagomez R, Smiljanic S, Huang X, Malakpour A, Kempengren S, Rodrigo G, Almanza G, Sterner O, Oredsson S. Anti-cancer stem cell activity of a sesquiterpene lactone isolated from Ambrosia arborescens and of a synthetic derivative. PLoS One 2017; 12:e0184304. [PMID: 28863191 PMCID: PMC5581169 DOI: 10.1371/journal.pone.0184304] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/21/2017] [Indexed: 01/06/2023] Open
Abstract
New regimens are constantly being pursued in cancer treatment, especially in the context of treatment-resistant cancer stem cells (CSCs) that are assumed to be involved in cancer recurrence. Here, we investigated the anti-cancer activity of sesquiterpene lactones (SLs) isolated from Ambrosia arborescens and of synthetic derivatives in breast cancer cell lines, with a specific focus on activity against CSCs. The breast cancer cell lines MCF-7, JIMT-1, and HCC1937 and the normal-like breast epithelial cell line MCF-10A were treated with the SLs damsin and coronopilin, isolated from A. arborescens, and with ambrosin and dindol-01, synthesized using damsin. Inhibitory concentration 50 (IC50) values were obtained from dose-response curves. Based on IC50 values, doses in the μM range were used for investigating effects on cell proliferation, cell cycle phase distribution, cell death, micronuclei formation, and cell migration. Western blot analysis was used to investigate proteins involved in cell cycle regulation as well as in the NF-κB pathway since SLs have been shown to inhibit this transcription factor. Specific CSC effects were investigated using three CSC assays. All compounds inhibited cell proliferation; however, damsin and ambrosin were toxic at single-digit micromolar ranges, while higher concentrations were required for coronopilin and dindol-01. Of the four cell lines, the compounds had the least effect on the normal-like MCF-10A cells. The inhibition of cell proliferation can partly be explained by downregulation of cyclin-dependent kinase 2. All compounds inhibited tumour necrosis factor-α-induced translocation of NF-κB from the cytoplasm to the nucleus. Damsin and ambrosin treatment increased the number of micronuclei; moreover, another sign of DNA damage was the increased level of p53. Treatment with damsin and ambrosin decreased the CSC subpopulation and inhibited cell migration. Our results suggest that these compounds should be further investigated to find efficient CSC-inhibiting compounds.
Collapse
Affiliation(s)
- Wendy Soria Sotillo
- Department of Biology, Lund University, Lund, Sweden.,Molecular Biology and Biotechnology Institute, University Major of San Andrés, La Paz, Bolivia
| | - Rodrigo Villagomez
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | | | - Xiaoli Huang
- Department of Biology, Lund University, Lund, Sweden
| | | | | | - Gloria Rodrigo
- Molecular Biology and Biotechnology Institute, University Major of San Andrés, La Paz, Bolivia
| | - Giovanna Almanza
- Chemical Research Institute, University Major of San Andres, La Paz, Bolivia
| | - Olov Sterner
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | | |
Collapse
|
230
|
Tomono T, Yano K, Ogihara T. Snail-Induced Epithelial-to-Mesenchymal Transition Enhances P-gp-Mediated Multidrug Resistance in HCC827 Cells. J Pharm Sci 2017; 106:2642-2649. [DOI: 10.1016/j.xphs.2017.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 12/12/2022]
|
231
|
Heterogeneity of p53 dependent genomic responses following ethanol exposure in a developmental mouse model of fetal alcohol spectrum disorder. PLoS One 2017; 12:e0180873. [PMID: 28723918 PMCID: PMC5516996 DOI: 10.1371/journal.pone.0180873] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 06/22/2017] [Indexed: 11/28/2022] Open
Abstract
Prenatal ethanol exposure can produce structural and functional deficits in the brain and result in Fetal Alcohol Spectrum Disorder (FASD). In rodent models acute exposure to a high concentration of alcohol causes increased apoptosis in the developing brain. A single causal molecular switch that signals for this increase in apoptosis has yet to be identified. The protein p53 has been suggested to play a pivotal role in enabling cells to engage in pro-apoptotic processes, and thus figures prominently as a hub molecule in the intracellular cascade of responses elicited by alcohol exposure. In the present study we examined the effect of ethanol-induced cellular and molecular responses in primary somatosensory cortex (SI) and hippocampus of 7-day-old wild-type (WT) and p53-knockout (KO) mice. We quantified apoptosis by active caspase-3 immunohistochemistry and ApopTag™ labeling, then determined total RNA expression levels in laminae of SI and hippocampal subregions. Immunohistochemical results confirmed increased incidence of apoptotic cells in both regions in WT and KO mice following ethanol exposure. The lack of p53 was not protective in these brain regions. Molecular analyses revealed a heterogeneous response to ethanol exposure that varied depending on the subregion, and which may go undetected using a global approach. Gene network analyses suggest that the presence or absence of p53 alters neuronal function and synaptic modifications following ethanol exposure, in addition to playing a classic role in cell cycle signaling. Thus, p53 may function in a way that underlies the intellectual and behavioral deficits observed in FASD.
Collapse
|
232
|
Takafuji T, Kayama K, Sugimoto N, Fujita M. GRWD1, a new player among oncogenesis-related ribosomal/nucleolar proteins. Cell Cycle 2017; 16:1397-1403. [PMID: 28722511 DOI: 10.1080/15384101.2017.1338987] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Increasing attention has been paid to certain ribosomal or ribosome biosynthesis-related proteins involved in oncogenesis. Members of one group are classified as "tumor suppressive factors" represented by RPL5 and RPL11; loss of their functions leads to cancer predisposition. RPL5 and RPL11 prevent tumorigenesis by binding to and inhibiting the MDM2 ubiquitin ligase and thereby up-regulating p53. Many other candidate tumor suppressive ribosomal/nucleolar proteins have been suggested. However, it remains to be experimentally clarified whether many of these factors can actually prevent tumorigenesis and if so, how they do so. Conversely, some ribosomal/nucleolar proteins promote tumorigenesis. For example, PICT1 binds to and anchors RPL11 in nucleoli, down-regulating p53 and promoting tumorigenesis. GRWD1 was recently identified as another such factor. When overexpressed, GRWD1 suppresses p53 and transforms normal human cells, probably by binding to RPL11 and sequestrating it from MDM2. However, other pathways may also be involved.
Collapse
Affiliation(s)
- Takuya Takafuji
- a Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences , Kyushu University , Higashi-ku, Fukuoka , Japan
| | - Kota Kayama
- a Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences , Kyushu University , Higashi-ku, Fukuoka , Japan
| | - Nozomi Sugimoto
- a Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences , Kyushu University , Higashi-ku, Fukuoka , Japan
| | - Masatoshi Fujita
- a Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences , Kyushu University , Higashi-ku, Fukuoka , Japan
| |
Collapse
|
233
|
Saeki Y, Abe M, Kono F, Nakazato A, Ishihara Y, Abe H. DNA methylation analysis of cancer-related genes from cell-free DNA of patients with cancer. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.pmu.2017.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
234
|
Biazi BI, Zanetti TA, Baranoski A, Corveloni AC, Mantovani MS. Cis-Nerolidol Induces Endoplasmic Reticulum Stress and Cell Death in Human Hepatocellular Carcinoma Cells through Extensive CYP2C19 and CYP1A2 Oxidation. Basic Clin Pharmacol Toxicol 2017; 121:334-341. [DOI: 10.1111/bcpt.12772] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 02/21/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Bruna Isabela Biazi
- Laboratory of Toxicological Genetics; Department of General Biology; Biological Sciences Center; State University of Londrina - UEL; Londrina Paraná Brazil
| | - Thalita Alves Zanetti
- Laboratory of Toxicological Genetics; Department of General Biology; Biological Sciences Center; State University of Londrina - UEL; Londrina Paraná Brazil
| | - Adrivanio Baranoski
- Laboratory of Toxicological Genetics; Department of General Biology; Biological Sciences Center; State University of Londrina - UEL; Londrina Paraná Brazil
| | - Amanda Cristina Corveloni
- Laboratory of Toxicological Genetics; Department of General Biology; Biological Sciences Center; State University of Londrina - UEL; Londrina Paraná Brazil
| | - Mário Sérgio Mantovani
- Laboratory of Toxicological Genetics; Department of General Biology; Biological Sciences Center; State University of Londrina - UEL; Londrina Paraná Brazil
| |
Collapse
|
235
|
Chaudhary R, Gryder B, Woods WS, Subramanian M, Jones MF, Li XL, Jenkins LM, Shabalina SA, Mo M, Dasso M, Yang Y, Wakefield LM, Zhu Y, Frier SM, Moriarity BS, Prasanth KV, Perez-Pinera P, Lal A. Prosurvival long noncoding RNA PINCR regulates a subset of p53 targets in human colorectal cancer cells by binding to Matrin 3. eLife 2017; 6. [PMID: 28580901 PMCID: PMC5470874 DOI: 10.7554/elife.23244] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 05/20/2017] [Indexed: 12/19/2022] Open
Abstract
Thousands of long noncoding RNAs (lncRNAs) have been discovered, yet the function of the vast majority remains unclear. Here, we show that a p53-regulated lncRNA which we named PINCR (p53-induced noncoding RNA), is induced ~100-fold after DNA damage and exerts a prosurvival function in human colorectal cancer cells (CRC) in vitro and tumor growth in vivo. Targeted deletion of PINCR in CRC cells significantly impaired G1 arrest and induced hypersensitivity to chemotherapeutic drugs. PINCR regulates the induction of a subset of p53 targets involved in G1 arrest and apoptosis, including BTG2, RRM2B and GPX1. Using a novel RNA pulldown approach that utilized endogenous S1-tagged PINCR, we show that PINCR associates with the enhancer region of these genes by binding to RNA-binding protein Matrin 3 that, in turn, associates with p53. Our findings uncover a critical prosurvival function of a p53/PINCR/Matrin 3 axis in response to DNA damage in CRC cells. DOI:http://dx.doi.org/10.7554/eLife.23244.001 Though DNA contains the information needed to build the proteins that keep cells alive, only 2% of the DNA in a human cell codes for proteins. The remaining 98% is referred to as non-coding DNA. The information in some of these non-coding regions can still be copied into molecules of RNA, including long molecules called lncRNAs. Little is known about what lncRNAs actually do, but growing evidence suggests that these molecules are important for a number of vital processes including cell growth and survival. When the DNA in an animal cell gets damaged, the cell needs to decide whether to pause growth and repair the damage, or to kill itself if the harm is too great. One of the best-studied proteins guiding this decision is the p53 protein, which increases the number of protein-coding genes needed to carry out either option in this decision. That is to say that, p53 regulates the genes needed to kill the cell and the genes needed to temporarily pause its growth and repair the damage, which instead keeps the cell alive. So, how does the p53 protein guide the decision, and are lncRNA molecules involved? Using human colon cancer cells, Chaudhary et al. now report that when DNA is damaged, the levels of a specific lncRNA increase 100-fold. Further experiments showed that this lncRNA – named PINCR, which refers to p53-induced noncoding RNA – promotes the survival of cells. Chaudhary et al. showed that PINCR molecules do this by recruiting a protein called Matrin 3 to a certain region in the DNA called an enhancer and then links it to promoter region in the DNA of specific genes that temporarily pause cell growth but keep the cell alive. This in turn activates these ‘pro-survival genes’. In further experiments, when the PINCR molecules were essentially deleted, p53 was not able to fully activate these genes and as a result more of the cells died. Together these findings increase our knowledge of how lncRNAs can work, especially in the context of DNA damage in cancer cells. A next important step will be to uncover other roles for the PINCR molecule in both cancer and healthy cells. DOI:http://dx.doi.org/10.7554/eLife.23244.002
Collapse
Affiliation(s)
- Ritu Chaudhary
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Berkley Gryder
- Oncogenomics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Wendy S Woods
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Murugan Subramanian
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Matthew F Jones
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Xiao Ling Li
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Lisa M Jenkins
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Svetlana A Shabalina
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Min Mo
- Laboratory of Gene Regulation and Development, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Mary Dasso
- Laboratory of Gene Regulation and Development, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Yuan Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Lalage M Wakefield
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Yuelin Zhu
- Molecular Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | | | - Branden S Moriarity
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Twin Cities, United States
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Pablo Perez-Pinera
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Ashish Lal
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| |
Collapse
|
236
|
Vangimalla SS, Ganesan M, Kharbanda KK, Osna NA. Bifunctional Enzyme JMJD6 Contributes to Multiple Disease Pathogenesis: New Twist on the Old Story. Biomolecules 2017; 7:biom7020041. [PMID: 28587176 PMCID: PMC5485730 DOI: 10.3390/biom7020041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 02/05/2023] Open
Abstract
Jumonji domain-containing protein 6 (JMJD6) is a non-heme Fe(II) 2-oxoglutarate (2OG)-dependent oxygenase with arginine demethylase and lysyl hydroxylase activities. Its initial discovery as a dispensable phosphatidylserine receptor (PSR) in the cell membrane of macrophages for phagocytosis was squashed by newer studies which revealed its nuclear localization and bifunctional enzymatic activity. Though its interaction with several nuclear and cytoplasmic target proteins has been demonstrated, the exact mechanisms and clinical significance of these various biologic interplays are not yet well established. Recent investigations have shed the light on the multiple pathways by which JMJD6 can regulate cell proliferation and cause tumorigenesis. Clinically, JMJD6 has been associated with more aggressive and metastatic disease, poorer prognosis, and lower overall survival rates-particularly in lung colon and oral cancers. JMJD6 is a novel biomarker for predicting future disease outcomes and is a target for new therapeutic treatments in future studies. Aberrant expression and dysregulation of JMJD6 are implicated in various other processes such as impaired T-cell proliferation and maturation, inoculation, and virulence of foot-and-mouth disease virus (FMDV), and impaired methylation of innate immunity factor. This article reviews the association of JMJD6 with various pathological processes-particularly, its role in tumorigenesis and virological interactions.
Collapse
Affiliation(s)
- Shiva Shankar Vangimalla
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE 68105, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE 68105, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE 68105, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Natalia A Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE 68105, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
237
|
Abstract
Innumerable breakthroughs in many fundamental areas of biology have come from unbiased screens and selections for mutations, either across the genome or within a gene. However, long-standing hurdles to key elements of mutant hunts (mutagenesis, phenotypic characterization, and linkage of phenotype to genotype) have limited the organisms in which mutant hunts could be used. These hurdles are now being eliminated by an explosion of new technologies. We believe that a renewed emphasis on unbiased mutant hunts, in both existing model systems and in those where genetics is just now becoming feasible, will lead to new seminal discoveries and surprises.
Collapse
|
238
|
R M, P HA, Mahadevan V. HDAC inhibitors show differential epigenetic regulation and cell survival strategies on p53 mutant colon cancer cells. J Biomol Struct Dyn 2017; 36:938-955. [PMID: 28264628 DOI: 10.1080/07391102.2017.1302820] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Besides inactivating tumour suppressor activity in cells, mutations in p53 confer significant oncogenic functions and promote metastasis and resistance to anticancer therapy. A variety of therapies involving genetic and epigenetic signalling events regulate tumorogenesis and progression in such cases. Pharmacological interventions with HDAC inhibitors have shown promise in therapy. This work explores the changes in efficacy of the four HDAC inhibitors SAHA, MS-275, valproic acid and sodium butyrate on a panel of colon cancer cell lines - HCT116 (p53 wt), HCT116 p53-/-, HT29 and SW480 (with mutations in p53). Clonogenic assays, gene profiling and epigenetic expression done on these cells point to p53 dependent differential activity of the 4 HDAC inhibitors which also elevate methylation levels in p53 mutant cell lines. In silico modelling establishes the alterations in interactions that lead to such differential activity of valproic acid, one of the inhibitors considered for the work. Molecular Dynamic simulations carried out on the valproic acid complex ensure stability of the complex. This work establishes a p53 dependent epigenetic signalling mechanism triggered by HDAC inhibition expanding the scope of HDAC inhibitors in adjuvant therapy for p53 mutant tumours.
Collapse
Affiliation(s)
- Mahalakshmi R
- a Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical & Biotechnology , SASTRA University , Thanjavur 613401 , India
| | - Husayn Ahmed P
- b Institute of Bioinformatics and Applied Biotechnology (IBAB) , Bangalore 560100 , India
| | - Vijayalakshmi Mahadevan
- a Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical & Biotechnology , SASTRA University , Thanjavur 613401 , India.,b Institute of Bioinformatics and Applied Biotechnology (IBAB) , Bangalore 560100 , India
| |
Collapse
|
239
|
Ciccarese C, Massari F, Blanca A, Tortora G, Montironi R, Cheng L, Scarpelli M, Raspollini MR, Vau N, Fonseca J, Lopez-Beltran A. Tp53 and its potential therapeutic role as a target in bladder cancer. Expert Opin Ther Targets 2017; 21:401-414. [PMID: 28281901 DOI: 10.1080/14728222.2017.1297798] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Despite more than 30 years of research on p53 resulting in >50,000 publications, we are now beginning to figure out the complexity of the p53 pathway, gene ontology and conformational structure of the molecule. Recent years brought great advances in p53 related drugs and the potencial ways in which p53 is inactivated in cancer. Areas covered: We searched for related publications on Pubmed and ClinicalTrial.gov using the following keywords 'p53, Tp53, p53 and bladder cancer, p53 and therapeutic target'. Relevant articles improved the understanding on p53 pathways and their potential as candidate to targeted therapy in bladder cancer. Expert opinion: Novel strategies developed to restore the function of mutants with chemical chaperones or by using compounds to improved pharmacokinetic properties are in development with potential to be applied in the oncology clinic. Other strategies targeting aberrantly overexpressed p53 regulators with wild-type p53 are also an active area of research. In particular, studies inhibiting the interaction of p53 with its negative regulators MDMX and MDM2 are an important field in drug discovery. Small molecules for inhibition of MDM2 are now in clinical trials process. However, personalized anticancer therapy might eventually advance through analyses of p53 status in cancer patients.
Collapse
Affiliation(s)
- Chiara Ciccarese
- a Medical Oncology, Azienda Ospedaliera Universitaria Integrata , University of Verona , Verona , Italy
| | - Francesco Massari
- b Medical Oncology , Azienda Ospedaliera Universitaria Integrata (A.O.U.I.) , Verona , Italy
| | - Ana Blanca
- c Maimonides Biomedical Research Institute of Cordoba, Spain - Urology Department , Reina Sofía Hospital , Córdoba , Spain
| | - Giampaolo Tortora
- d Medical Oncology dU, Policlinico 'G.B. Rossi' , University of Verona , Verona , Italy
| | - Rodolfo Montironi
- e Pathological Anatomy , Polytechnic University of the Marche Region, School of Medicine, United Hospitals , Ancona , Italy
| | - Liang Cheng
- f Department of Pathology and Laboratory Medicine , Indiana University School of Medicine , Indianapolis , IN 46202 , USA
| | - Marina Scarpelli
- e Pathological Anatomy , Polytechnic University of the Marche Region, School of Medicine, United Hospitals , Ancona , Italy
| | - Maria R Raspollini
- g Histopathology and Molecular Diagnostics Service , Careggi University Hospital Florence , Florence , Italy
| | - Nuno Vau
- h Medical Oncology , Champalimaud Clinical Center , Lisbon , Portugal
| | - Jorge Fonseca
- i Urology service , Champalimaud Clinical Center , Lisbon , Portugal
| | - Antonio Lopez-Beltran
- j Department of Surgery and Pathology , Cordoba University Medical School, Cordoba, Spain and Champalimaud Clinical Center , Lisbon , Portugal
| |
Collapse
|
240
|
Squillaro T, Antonucci I, Alessio N, Esposito A, Cipollaro M, Melone MAB, Peluso G, Stuppia L, Galderisi U. Impact of lysosomal storage disorders on biology of mesenchymal stem cells: Evidences from in vitro silencing of glucocerebrosidase (GBA) and alpha-galactosidase A (GLA) enzymes. J Cell Physiol 2017; 232:3454-3467. [PMID: 28098348 DOI: 10.1002/jcp.25807] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 12/27/2022]
Abstract
Lysosomal storage disorders (LDS) comprise a group of rare multisystemic diseases resulting from inherited gene mutations that impair lysosomal homeostasis. The most common LSDs, Gaucher disease (GD), and Fabry disease (FD) are caused by deficiencies in the lysosomal glucocerebrosidase (GBA) and alpha-galactosidase A (GLA) enzymes, respectively. Given the systemic nature of enzyme deficiency, we hypothesized that the stem cell compartment of GD and FD patients might be also affected. Among stem cells, mesenchymal stem cells (MSCs) are a commonly investigated population given their role in hematopoiesis and the homeostatic maintenance of many organs and tissues. Since the impairment of MSC functions could pose profound consequences on body physiology, we evaluated whether GBA and GLA silencing could affect the biology of MSCs isolated from bone marrow and amniotic fluid. Those cell populations were chosen given the former's key role in organ physiology and the latter's intriguing potential as an alternative stem cell model for human genetic disease. Our results revealed that GBA and GLA deficiencies prompted cell cycle arrest along with the impairment of autophagic flux and an increase of apoptotic and senescent cell percentages. Moreover, an increase in ataxia-telangiectasia-mutated staining 1 hr after oxidative stress induction and a return to basal level at 48 hr, along with persistent gamma-H2AX staining, indicated that MSCs properly activated DNA repair signaling, though some damages remained unrepaired. Our data therefore suggest that MSCs with reduced GBA or GLA activity are prone to apoptosis and senescence due to impaired autophagy and DNA repair capacity.
Collapse
Affiliation(s)
- Tiziana Squillaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy.,Institute of Bioscience and Bioresources, National Research Council, Naples, Italy
| | - Ivana Antonucci
- Laboratory of Molecular Genetics, Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, G. d'Annunzio University, Chieti-Pescara, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Anna Esposito
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marilena Cipollaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mariarosa Anna Beatrice Melone
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging; Division of Neurology and InterUniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gianfranco Peluso
- Institute of Bioscience and Bioresources, National Research Council, Naples, Italy
| | - Liborio Stuppia
- Laboratory of Molecular Genetics, Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, G. d'Annunzio University, Chieti-Pescara, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
241
|
Tan BX, Liew HP, Chua JS, Ghadessy FJ, Tan YS, Lane DP, Coffill CR. Anatomy of Mdm2 and Mdm4 in evolution. J Mol Cell Biol 2017; 9:3-15. [PMID: 28077607 PMCID: PMC6372010 DOI: 10.1093/jmcb/mjx002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/24/2016] [Accepted: 01/10/2017] [Indexed: 01/09/2023] Open
Abstract
Mouse double minute (Mdm) genes span an evolutionary timeframe from the ancient eukaryotic placozoa Trichoplax adhaerens to Homo sapiens, implying a significant and possibly conserved cellular role throughout history. Maintenance of DNA integrity and response to DNA damage involve many key regulatory pathways, including precise control over the tumour suppressor protein p53. In most vertebrates, degradation of p53 through proteasomal targeting is primarily mediated by heterodimers of Mdm2 and the Mdm2-related protein Mdm4 (also known as MdmX). Both Mdm2 and Mdm4 have p53-binding regions, acidic domains, zinc fingers, and C-terminal RING domains that are conserved throughout evolution. Vertebrates typically have both Mdm2 and Mdm4 genes, while analyses of sequenced genomes of invertebrate species have identified single Mdm genes, suggesting that a duplication event occurred prior to emergence of jawless vertebrates about 550-440 million years ago. The functional relationship between Mdm and p53 in T. adhaerens, an organism that has existed for 1 billion years, implies that these two proteins have evolved together to maintain a conserved and regulated function.
Collapse
Affiliation(s)
- Ban Xiong Tan
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #06-06, Singapore138648, Singapore
| | - Hoe Peng Liew
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #06-06, Singapore138648, Singapore
| | - Joy S. Chua
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #06-06, Singapore138648, Singapore
| | - Farid J. Ghadessy
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #06-06, Singapore138648, Singapore
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis St, #07-01,Singapore138671, Singapore
| | - David P. Lane
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #06-06, Singapore138648, Singapore
| | - Cynthia R. Coffill
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #06-06, Singapore138648, Singapore
| |
Collapse
|
242
|
Sepuri NBV, Tammineni P, Mohammed F, Paripati A. Nuclear Transcription Factors in the Mitochondria: A New Paradigm in Fine-Tuning Mitochondrial Metabolism. Handb Exp Pharmacol 2017; 240:3-20. [PMID: 27417432 DOI: 10.1007/164_2016_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Noncanonical functions of several nuclear transcription factors in the mitochondria have been gaining exceptional traction over the years. These transcription factors include nuclear hormone receptors like estrogen, glucocorticoid, and thyroid hormone receptors: p53, IRF3, STAT3, STAT5, CREB, NF-kB, and MEF-2D. Mitochondria-localized nuclear transcription factors regulate mitochondrial processes like apoptosis, respiration and mitochondrial transcription albeit being nuclear in origin and having nuclear functions. Hence, the cell permits these multi-stationed transcription factors to orchestrate and fine-tune cellular metabolism at various levels of operation. Despite their ubiquitous distribution in different subcompartments of mitochondria, their targeting mechanism is poorly understood. Here, we review the current status of mitochondria-localized transcription factors and discuss the possible targeting mechanism besides the functional interplay between these factors.
Collapse
Affiliation(s)
- Naresh Babu V Sepuri
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Telangana, 500046, India.
| | - Prasad Tammineni
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Telangana, 500046, India
| | - Fareed Mohammed
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Telangana, 500046, India
| | - Arunkumar Paripati
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Telangana, 500046, India
| |
Collapse
|
243
|
Min-Wen JC, Yan-Jiang BC, Mishra S, Dai X, Magae J, Shyh-Chang N, Kumar AP, Sethi G. Molecular Targets of Ascochlorin and Its Derivatives for Cancer Therapy. STRESS AND INFLAMMATION IN DISORDERS 2017; 108:199-225. [DOI: 10.1016/bs.apcsb.2017.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
244
|
Carrà G, Crivellaro S, Taulli R, Guerrasio A, Saglio G, Morotti A. Mechanisms of p53 Functional De-Regulation: Role of the IκB-α/p53 Complex. Int J Mol Sci 2016; 17:ijms17121997. [PMID: 27916821 PMCID: PMC5187797 DOI: 10.3390/ijms17121997] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/23/2016] [Accepted: 11/24/2016] [Indexed: 02/06/2023] Open
Abstract
TP53 is one of the most frequently-mutated and deleted tumor suppressors in cancer, with a dramatic correlation with dismal prognoses. In addition to genetic inactivation, the p53 protein can be functionally inactivated in cancer, through post-transductional modifications, changes in cellular compartmentalization, and interactions with other proteins. Here, we review the mechanisms of p53 functional inactivation, with a particular emphasis on the interaction between p53 and IκB-α, the NFKBIA gene product.
Collapse
Affiliation(s)
- Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Sabrina Crivellaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Riccardo Taulli
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Angelo Guerrasio
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| |
Collapse
|
245
|
Wang Q, Zou Y, Nowotschin S, Kim SY, Li QV, Soh CL, Su J, Zhang C, Shu W, Xi Q, Huangfu D, Hadjantonakis AK, Massagué J. The p53 Family Coordinates Wnt and Nodal Inputs in Mesendodermal Differentiation of Embryonic Stem Cells. Cell Stem Cell 2016; 20:70-86. [PMID: 27889317 DOI: 10.1016/j.stem.2016.10.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 09/07/2016] [Accepted: 10/02/2016] [Indexed: 01/01/2023]
Abstract
In this study, we outline a regulatory network that involves the p53 tumor suppressor family and the Wnt pathway acting together with the TGF-β pathway in mesendodermal differentiation of mouse and human embryonic stem cells. Knockout of all three members, p53, p63, and p73, shows that the p53 family is essential for mesendoderm specification during exit from pluripotency in embryos and in culture. Wnt3 and its receptor Fzd1 are direct p53 family target genes in this context, and induction of Wnt signaling by p53 is critical for activation of mesendodermal differentiation genes. Globally, Wnt3-activated Tcf3 and nodal-activated Smad2/3 transcription factors depend on each other for co-occupancy of target enhancers associated with key differentiation loci. Our results therefore highlight an unanticipated role for p53 family proteins in a regulatory network that integrates essential Wnt-Tcf and nodal-Smad inputs in a selective and interdependent way to drive mesendodermal differentiation of pluripotent cells.
Collapse
Affiliation(s)
- Qiong Wang
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yilong Zou
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sonja Nowotschin
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sang Yong Kim
- Rodent Genetic Engineering Core, Langone Medical Center, New York University, New York, NY 10016, USA
| | - Qing V Li
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, New York, NY 10065, USA
| | - Chew-Li Soh
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jie Su
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chao Zhang
- Department of Medicine and Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Weiping Shu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Qiaoran Xi
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Danwei Huangfu
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Joan Massagué
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
246
|
Songock WK, Kim SM, Bodily JM. The human papillomavirus E7 oncoprotein as a regulator of transcription. Virus Res 2016; 231:56-75. [PMID: 27818212 DOI: 10.1016/j.virusres.2016.10.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022]
Abstract
High-risk human papillomaviruses (HPVs) encode oncoproteins which manipulate gene expression patterns in the host keratinocytes to facilitate viral replication, regulate viral transcription, and promote immune evasion and persistence. In some cases, oncoprotein-induced changes in host cell behavior can cause progression to cancer, but a complete picture of the functions of the viral oncoproteins in the productive HPV life cycle remains elusive. E7 is the HPV-encoded factor most responsible for maintaining cell cycle competence in differentiating keratinocytes. Through interactions with dozens of host factors, E7 has an enormous impact on host gene expression patterns. In this review, we will examine the role of E7 specifically as a regulator of transcription. We will discuss mechanisms of regulation of cell cycle-related genes by E7 as well as genes involved in immune regulation, growth factor signaling, DNA damage responses, microRNAs, and others pathways. We will also discuss some unanswered questions about how transcriptional regulation by E7 impacts the biology of HPV in both benign and malignant conditions.
Collapse
Affiliation(s)
- William K Songock
- Department of Microbiology and Immunology and Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Seong-Man Kim
- Department of Microbiology and Immunology and Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Jason M Bodily
- Department of Microbiology and Immunology and Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
247
|
Benzyl isothiocyanate promotes apoptosis of oral cancer cells via an acute redox stress-mediated DNA damage response. Food Chem Toxicol 2016; 97:336-345. [DOI: 10.1016/j.fct.2016.09.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/19/2016] [Accepted: 09/26/2016] [Indexed: 11/24/2022]
|
248
|
Kozłowska E, Puszynski K. Application of bifurcation theory and siRNA-based control signal to restore the proper response of cancer cells to DNA damage. J Theor Biol 2016; 408:213-221. [DOI: 10.1016/j.jtbi.2016.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 07/17/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
|
249
|
Canale M, Petracci E, Delmonte A, Chiadini E, Dazzi C, Papi M, Capelli L, Casanova C, De Luigi N, Mariotti M, Gamboni A, Chiari R, Bennati C, Calistri D, Ludovini V, Crinò L, Amadori D, Ulivi P. Impact of TP53 Mutations on Outcome in EGFR-Mutated Patients Treated with First-Line Tyrosine Kinase Inhibitors. Clin Cancer Res 2016; 23:2195-2202. [PMID: 27780855 DOI: 10.1158/1078-0432.ccr-16-0966] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/30/2016] [Accepted: 10/17/2016] [Indexed: 11/16/2022]
Abstract
Purpose: To analyze the impact of TP53 mutations on response to first-line tyrosine kinase inhibitors (TKI) in patients with EGFR-mutated non-small cell lung cancer (NSCLC).Experimental Design: 136 EGFR-mutated NSCLC patients receiving first-line TKIs were analyzed. TP53 mutations were evaluated in 123 patients in relation to disease control rate (DCR), objective response rate (ORR), progression-free survival (PFS), and overall survival (OS).Results:TP53 mutations were observed in 37 (30.1%), 10 (27.0%), 6 (16.2%), 9 (24.3%), and 12 (32.4%) patients in exons 5, 6, 7, and 8, respectively. DCR was 70% in TP53-mutated patients compared with 88% in TP53-wild type (wt) patients [relative risk, RR, of disease progression: 3.17 (95% CI, 1.21-8.48), P = 0.019]. In particular, a 42% DCR was observed in patients with TP53 exon 8 mutation versus 87% in exon 8 wt patients [RR of disease progression 9.6 (2.71-36.63), P < 0.001]. Shorter median PFS and OS were observed in patients with TP53 exon 8 mutations compared with others (4.2 vs. 12.5, P = 0.058, and 16.2 vs. 32.3, P = 0.114, respectively); these differences became significant in the subgroup with EGFR exon 19 deletion (4.2 vs. 16.8, P < 0.001, and 7.6 vs. not reached, P = 0.006, respectively), HR 6.99 (95% CI, 2.34-20.87, P < 0.001) and HR 4.75 (95% CI, 1.38-16.29, P = 0.013), respectively.Conclusions:TP53 mutations, especially exon 8 mutations, reduce responsiveness to TKIs and worsen prognosis in EGFR-mutated NSCLC patients, mainly those carrying exon 19 deletions. Clin Cancer Res; 23(9); 2195-202. ©2016 AACR.
Collapse
Affiliation(s)
- Matteo Canale
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Angelo Delmonte
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Elisa Chiadini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Claudio Dazzi
- Medical Oncology Unit, S.Maria delle Croci Hospital, Ravenna, Italy
| | | | - Laura Capelli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Claudia Casanova
- Medical Oncology Unit, S.Maria delle Croci Hospital, Ravenna, Italy
| | - Nicoletta De Luigi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Marita Mariotti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Rita Chiari
- Division of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Chiara Bennati
- Division of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Daniele Calistri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Vienna Ludovini
- Division of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Lucio Crinò
- Division of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Dino Amadori
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| |
Collapse
|
250
|
Abstract
As an indispensable process of cell life, apoptosis is essential for keeping homeostasis at cell level. Dysregulation of apoptosis is usually involved in the pathological processes of many complex diseases including cancer. With the properties such as high affinity and specificity to their targets, easy of synthesis and modification and good biocompatibility, aptamers have been attractive molecules applied in basic research, diagnostics and therapeutics. This review mainly focuses on the recent researches on application of aptamers in interference of cell apoptosis. Key targets along the intrinsic and extrinsic apoptosis pathways were respectively dissected using aptamers as a tool, providing an insight into the pathological processes, especially for cancer.
Collapse
|