1
|
Titania Nanosheet Generates Peroxynitrite-Dependent S-Nitrosylation and Enhances p53 Function in Lung Cancer Cells. Pharmaceutics 2021; 13:pharmaceutics13081233. [PMID: 34452194 PMCID: PMC8401232 DOI: 10.3390/pharmaceutics13081233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/17/2022] Open
Abstract
Metal nanomaterials can enhance the efficacy of current cancer therapies. Here, we show that Ti0.8O2 nanosheets cause cytotoxicity in several lung cancer cells but not in normal cells. The nanosheet-treated cells showed certain apoptosis characteristics. Protein analysis further indicated the activation of the p53-dependent death mechanism. Transmission electron microscopy (TEM) and scanning electron microscopy (SEM) analyses revealed the cellular uptake of the nanosheets and the induction of cell morphological change. The nanosheets also exhibited a substantial apoptosis effect on drug-resistant metastatic primary lung cancer cells, and it was found that the potency of the nanosheets was dramatically higher than standard drugs. Ti0.8O2 nanosheets induce apoptosis through a molecular mechanism involving peroxynitrite (ONOO−) generation. As peroxynitrite is known to be a potent inducer of S-nitrosylation, we further found that the nanosheets mediated the S-nitrosylation of p53 at C182, resulting in higher protein-protein complex stability, and this was likely to induce the surrounding residues, located in the interface region, to bind more strongly to each other. Molecular dynamics analysis revealed that S-nitrosylation stabilized the p53 dimer with a ΔGbindresidue of <−1.5 kcal/mol. These results provide novel insight on the apoptosis induction effect of the nanosheets via a molecular mechanism involving S-nitrosylation of the p53 protein, emphasizing the mechanism of action of nanomaterials for cancer therapy.
Collapse
|
2
|
Cai Y, Lin Y, Xiong X, Lu J, Zhou R, Jin Y, You Z, Ye H, Li F, Cheng N. Knockdown expression of MECR, a novel gene of mitochondrial FAS II inhibits growth and colony-formation, promotes apoptosis of hepatocelluar carcinoma cells. Biosci Trends 2019; 13:234-244. [PMID: 31178528 DOI: 10.5582/bst.2019.01109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Mitochondrial trans-2-enoyl-CoA reductase (MECR) is a protein-coding gene, and the protein encoded by this gene is an oxidoreductase that catalyzes the last step in mitochondrial fatty acid synthesis (mtFASII). Numerous studies have shown disorder of lipid metabolism is closely related with malignance, especially in liver cancer. Through pre-experiment, we found that the expression of MECR gene was highly expressed in hepatocelluar carcinoma (HCC) cell lines in vitro. This suggests that the MECR gene may play a role of oncogene in HCC. Therefore, we conducted a preliminary experimental study on the role of MECR gene in HCC cells in vitro. Objective to explore whether the MECR gene can affect the malignant biological behavior of HCC. We selected HCC cell line BEL-7404 as experimental cell, which involves the highest expression of MECR in the pre-experiment. We constructed MECR knockdwon lentivirus vector, and then infected HCC cell lines to down-regulate MECR expression, and establish negative control group (NC). Through various experiments of cytology, our study showed that knockdown of MECR inhibited cell proliferation and colony formation, promoted apoptosis, and inhibited metastasis in HCC cell lines BEL-7404. MECR might serve as a novel gene therapeutic target for the treatment of HCC. Further study is needed to elucidate the signaling pathway through which MECR functions in HCC.
Collapse
Affiliation(s)
- Yulong Cai
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Yixin Lin
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Xianze Xiong
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Jiong Lu
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Rongxing Zhou
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Yanwen Jin
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Zhen You
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Hui Ye
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Fuyu Li
- Department of Biliary Surgery, West China Hospital, Sichuan University
| | - Nansheng Cheng
- Department of Biliary Surgery, West China Hospital, Sichuan University
| |
Collapse
|
3
|
Abstract
The p53 gene is pivotal for oncogenesis in a combination of mutations in oncogenes and antioncogenes. The ubiquitous loss of the p53 pathway in human cancers has generated considerable interest in developing p53-targeted cancer therapies, but current ideas and approaches targeting p53 are conflicting. Current researches focus on cancer-selective drugs with therapeutic strategies that both activate and inhibit p53. As p53 is ubiquitously lost in human cancers, the strategy of exogenous p53 addition is reasonable. However, p53 acts not equally in all cell types; thus, individualized p53 therapy is the direction of future research. To clarify the controversies on p53 for improvement of future antitumor studies, the review focuses on the available technological protocols, including their advantages and limitations in terms of future therapeutic use of p53 in the management of tumors.
Collapse
|
4
|
Variations within 3'-UTR of MDM4 gene contribute to clinical outcomes of advanced non-small cell lung cancer patients following platinum-based chemotherapy. Oncotarget 2017; 8:16313-16324. [PMID: 27462918 PMCID: PMC5369965 DOI: 10.18632/oncotarget.10771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/17/2016] [Indexed: 01/22/2023] Open
Abstract
Single-nucleotide polymorphism (SNPs) in microRNA (miRNA)-binding sites may modulate the posttranscriptional regulation of gene expression and explain individual sensitivity to platinum agents. This study aimed to investigate the impact of SNPs located at 3′-untranslated region (UTR) of MDM4 gene, on clinical outcomes of advanced non-small cell lung cancer (NSCLC) patients. Four SNPs were genotyped by using DNA from blood samples of advanced NSCLC patients (642 in the Discovery set and 330 in the Replication set) and were analyzed the relationships with clinical outcomes. Carriers with rs10900598 CC genotype and rs4245739 AC genotype showed increased overall survival (OS) than those with AA genotype (P = 0.017 and P = 0.037, respectively) in the Discovery set and after pooling results from the Replication set. A combined effect on survival of variant alleles was also concluded and validated. Stratification analysis revealed that the effect of MDM4 SNPs was more pronounced in lung adenocarcinoma (LAC) subgroups. A reduced expression of the reporter gene for the C allele of rs4245739 was observed in NSCLC cells using luciferase reporter gene assays. Taken together, our results demonstrate that genetic variations in 3′-UTR of MDM4 gene may influence outcomes of advanced NSCLC by miRNAs-mediated regulation.
Collapse
|
5
|
Giovannini C, Baglioni M, Baron Toaldo M, Cescon M, Bolondi L, Gramantieri L. Vidatox 30 CH has tumor activating effect in hepatocellular carcinoma. Sci Rep 2017; 7:44685. [PMID: 28322221 PMCID: PMC5359575 DOI: 10.1038/srep44685] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 12/02/2016] [Indexed: 12/12/2022] Open
Abstract
Complementary and alternative medicine (CAM) is the term used to describe many kinds of products, practices, and systems that are not part of conventional medicine. Cancer patients usually do everything they can to combat the disease, manage its symptoms, and cope with the side effects of treatment. Unfortunately, patients who use CAM underestimate the risk of interaction with cancer therapy or worse they omit conventional therapy thus reducing the possibility of cancer remission. Herein we analyzed the effects of Vidatox 30 CH (venom extracted from the Junceus Rhopalurus scorpion) on hepatocellular carcinoma (HCC), the second leading cause of cancer-related deaths. We found out that Vidatox increases HCC proliferation and invasion whereas it does not seem to interact with sorafenib, the orally active multikinase inhibitor approved for the treatment of advanced hepatocellular carcinoma. Our results suggest that the concentration of Vidatox used in the present study has not anti-neoplastic effects and care must be taken in hiring Vidatox in patients with HCC.
Collapse
Affiliation(s)
- Catia Giovannini
- Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi University Hospital, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Michele Baglioni
- Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Marco Baron Toaldo
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, Bologna, Italy
| | - Matteo Cescon
- Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Luigi Bolondi
- Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi University Hospital, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Laura Gramantieri
- Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
6
|
Kay LJ, Smulders-Srinivasan TK, Soundararajan M. Understanding the Multifaceted Role of Human Down Syndrome Kinase DYRK1A. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 105:127-71. [PMID: 27567487 DOI: 10.1016/bs.apcsb.2016.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The dual-specificity tyrosine (Y) phosphorylation-regulated kinase DYRK1A, also known as Down syndrome (DS) kinase, is a dosage-dependent signaling kinase that was originally shown to be highly expressed in DS patients as a consequence of trisomy 21. Although this was evident some time ago, it is only in recent investigations that the molecular roles of DYRK1A in a wide range of cellular processes are becoming increasingly apparent. Since initial knowledge on DYRK1A became evident through minibrain mnb, the Drosophila homolog of DYRK1A, this review will first summarize the scientific reports on minibrain and further expand on the well-established neuronal functions of mammalian and human DYRK1A. Recent investigations across the current decade have provided rather interesting and compelling evidence in establishing nonneuronal functions for DYRK1A, including its role in infection, immunity, cardiomyocyte biology, cancer, and cell cycle control. The latter part of this review will therefore focus in detail on the emerging nonneuronal functions of DYRK1A and summarize the regulatory role of DYRK1A in controlling Tau and α-synuclein. Finally, the emerging role of DYRK1A in Parkinson's disease will be outlined.
Collapse
Affiliation(s)
- L J Kay
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - T K Smulders-Srinivasan
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - M Soundararajan
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
7
|
Shahera U, Munshi S, Jahan M, Nessa A, Alam S, Tabassum S. IP-10, p53, and Foxp3 Expression in Hepatocytes of Chronic Hepatitis B Patients with Cirrhosis and Hepatocellular Carcinoma. Euroasian J Hepatogastroenterol 2016; 6:149-153. [PMID: 29201748 PMCID: PMC5578584 DOI: 10.5005/jp-journals-10018-1188] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022] Open
Abstract
AIM Elucidating differences in gene expression may be useful in understanding the molecular pathogenesis and for developing specific markers for the outcome of hepatitis B virus (HBV) infection. In the present study, expressions of host gene interferon gamma-inducible protein (IP-10), p53, and Foxp3 were studied in hepatocytes of patients with chronic HBV infection to determine a possible link between selected host gene expression and the outcome of HBV infection. MATERIALS AND METHODS The study was conducted in 60 patients with chronic HBV infection and they were divided into four groups: HBV-positive cirrhosis (n = 15), HBV-negative cirrhosis (n = 15), HBV-positive hepatocellular carcinoma (HCC) (n = 15) and HBV-negative HCC (n = 15). Total messenger ribonucleic acid (mRNA) extraction was done followed by complementary deoxyribonucleic acid (cDNA) synthesis, and finally gene expression was performed using real-time polymerase chain reaction (PCR) technique. RESULTS IP-10 and p53 gene expressions were lower in HBV-positive cirrhosis, and Foxp3 gene expression was upregulated in HBV-positive cirrhosis in comparison to HBV-negative cirrhosis. The expressions of all the three genes were upregulated among HBV-positive HCC in comparison to HBV-negative HCC. The expression of IP-10, p53, and Foxp3 genes was upregulated in HBV-positive HCC in comparison to HBV-positive cirrhosis. CONCLUSION This study indicates that there are variations in the expression of the selected genes among cirrhosis and HCC patients with or without HBV. All the three selected genes were more or less upregulated in HBV-positive HCC patients, but only Foxp3 expression was upregulated in HBV-positive cirrhosis. These three particular genes may have a role in the molecular pathogenesis and clinical outcome of HBV-positive cirrhosis and HCC patients. These aspects need further evaluation by studies with larger numbers of cirrhosis and HCC patients. HOW TO CITE THIS ARTICLE Shahera U, Munshi S, Jahan M, Nessa A, Alam S, Tabassum S. IP-10, p53, and Foxp3 Expression in Hepatocytes of Chronic Hepatitis B Patients with Cirrhosis and Hepatocellular Carcinoma. Euroasian J Hepato-Gastroenterol 2016;6(2):149-153.
Collapse
Affiliation(s)
- Umme Shahera
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Shahabag, Dhaka, Bangladesh
| | - Saifullah Munshi
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Shahabag, Dhaka, Bangladesh
| | - Munira Jahan
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Shahabag, Dhaka, Bangladesh
| | - Afzalun Nessa
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Shahabag, Dhaka, Bangladesh
| | - Shahinul Alam
- Department of Hepatology Bangabandhu Sheikh Mujib Medical University, Shahabag, Dhaka, Bangladesh
| | - Shahina Tabassum
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Shahabag, Dhaka, Bangladesh
| |
Collapse
|
8
|
Kaur S, Sambyal V, Guleria K, Manjari M, Sudan M, Uppal MS, Singh NR, Singh G, Singh H. Analysis of TP53 polymorphisms in North Indian sporadic esophageal cancer patients. Asian Pac J Cancer Prev 2014; 15:8413-22. [PMID: 25339039 DOI: 10.7314/apjcp.2014.15.19.8413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To investigate the relationship of five TP53 polymorphisms (p.P47S, p.R72P, PIN3 ins16bp, p.R213R and r.13494g>a) with the esophageal cancer (EC) risk in North Indians. MATERIALS AND METHODS Genotyping of p.P47S, p.R72P, PIN3 ins16bp, p.R213R and r.13494g>a polymorphisms of TP53 in 136 sporadic EC patients and 136 controls using polymerase chain reaction and PCR-RFLP. RESULTS The frequencies of genotype RR, RP and PP of p.R72P polymorphism were 16.91 vs 26.47%, 58.82 vs 49.27% and 24.27 vs 24.27% among patients and controls respectively. We observed significantly increased frequency of RP genotype in cases as compared to controls (OR=1.87, 95% CI, 1.01-3.46, p=0.05). The frequencies of genotype A1A1, A1A2 and A2A2 of PIN3 ins16bp polymorphism were 69.12 vs 70.59%, 27.20 vs 25% and 3.68 vs 4.41% among patients and controls. There was no significant difference among genotype and allele distribution between patients and controls. The frequencies of genotype GG, GA and AA of r.13494g>a polymorphism were 62.50 vs 64.70%, 34.56 vs 30.15% and 2.94 vs 5.15% among patients and controls respectively. No significant difference between genotype and allele frequency was observed in the patients and controls. For p.P47S and p.R213R polymorphisms, all the cases and controls had homozygous wild type genotype. The RP-A1A1-GG genotype combination shows significant risk for EC (OR=2.01, 95%CI: 1.01-3.99, p=0.05). CONCLUSIONS Among the five TP53 polymorphisms investigated, only p.R72P polymorphism may contributes to EC susceptibility.
Collapse
Affiliation(s)
- Sukhpreet Kaur
- Human Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Heng BC, Aubel D, Fussenegger M. An overview of the diverse roles of G-protein coupled receptors (GPCRs) in the pathophysiology of various human diseases. Biotechnol Adv 2013; 31:1676-94. [DOI: 10.1016/j.biotechadv.2013.08.017] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/19/2013] [Accepted: 08/19/2013] [Indexed: 12/23/2022]
|
10
|
Chen WJ, Xiong ZA, Tang Y, Dong PT, Li P, Wang ZG. Feasibility and effect of ultrasound microbubble-mediated wild-type p53 gene transfection of HeLa cells. Exp Ther Med 2012; 3:999-1004. [PMID: 22970006 DOI: 10.3892/etm.2012.528] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/20/2012] [Indexed: 11/06/2022] Open
Abstract
Gene therapy holds great promise for the treatment of diseases. The key problem of gene therapy is the choice of an effective vector. Ultrasound-mediated microbubble technique (UMMT) has already shown promising applications in numerous types of tumors apart from cervical carcinoma. In the present study, according to the results of an MTT assay, we initially chose an ultrasound intensity of 0.5 W/cm(2), an ultrasound exposure time of 30 sec and a microbubble concentration of 10% as the optimum experimental condition for wtp53 plasmid transfection into HeLa cells. To further investigate the transfection efficiency of ultrasound combined with microbubbles, RT-PCR analysis was used to examine the mRNA level of p53. The transfection efficiency in the plasmid plus microbubbles and ultrasound group was significantly higher than that of the other groups. Following transfection of the wtp53 gene, flow cytometric analysis showed that the cell cycle of HeLa cells was arrested in the G1 phase. The results of the present study suggest that UMMT, a new gene delivery system, increases the transfection efficiency of the wtp53 gene. Moreover, the growth of HeLa cells was arrested by introducing wtp53. This study may afford a new trend for the gene therapy of cervical carcinoma.
Collapse
|
11
|
Nadler-Milbauer M, Apter L, Haupt Y, Haupt S, Barenholz Y, Minko T, Rubinstein A. Synchronized release of Doxil and Nutlin-3 by remote degradation of polysaccharide matrices and its possible use in the local treatment of colorectal cancer. J Drug Target 2012; 19:859-73. [PMID: 22082104 DOI: 10.3109/1061186x.2011.622401] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A novel approach to the prevention of colorectal cancer (CRC) recurrence by the local, luminal application of the combined therapies: Nutlin-3 (NUT) and the liposomal preparation of doxorubicin, Doxil(*) (Doxil) is presented here. The two drug entities were loaded into calcium alginate beads, engineered to erode upon exposure to a de-crosslinking agent, to allow for the controlled, concomitant release of the two. The synchronized release-driven improved cytotoxicity of NUT and Doxil was tested in vitro in RKO (wild-type p53) and HT-29 (mutant p53) CRC cells, by measuring intracellular expression of p53, p21 and Mdm2, as well as monitoring cell proliferation and viable cell numbers. NUT treatment alone was identified to be cytotoxic exclusively towards RKO cells. However, coadministration of NUT enhanced Doxil's anti-proliferative effects and cell death induction in a synergistic manner in both cell types. It was also identified that combinatorial treatment in a wt p53 context affected the p53 pathway by elevating the expression of p53 and its target p21. The capability of the formulation to erode in the presence of a de-crosslinking agent was demonstrated in vivo in the cecum of the anesthetized rat using indomethacin as a poorly water-soluble PK probe.
Collapse
Affiliation(s)
- Mirela Nadler-Milbauer
- The Hebrew University of Jerusalem, School of Pharmacy, Research Institute for Drug Research, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
G protein-coupled receptors (GPCRs) belong to a superfamily of cell surface signalling proteins that have a pivotal role in many physiological functions and in multiple diseases, including the development of cancer and cancer metastasis. Current drugs that target GPCRs - many of which have excellent therapeutic benefits - are directed towards only a few GPCR members. Therefore, huge efforts are currently underway to develop new GPCR-based drugs, particularly for cancer. We review recent findings that present unexpected opportunities to interfere with major tumorigenic signals by manipulating GPCR-mediated pathways. We also discuss current data regarding novel GPCR targets that may provide promising opportunities for drug discovery in cancer prevention and treatment.
Collapse
|
13
|
Luo J, Zhou X, Diao L, Wang Z. Experimental Research on Wild-type p53 Plasmid Transfected into Retinoblastoma Cells and Tissues using an Ultrasound Microbubble Intensifier. J Int Med Res 2010; 38:1005-15. [PMID: 20819437 DOI: 10.1177/147323001003800327] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The transfection efficiency of wild-type p53 ( wtp53) was investigated in retinoblastoma (RB) Y79 cells using an ultrasound microbubble technique. A human RB nude mouse xenograft tumour model was also used to investigate whether this technique could deliver wtp53 into solid tumours. Reverse transcription–polymerase chain reaction (RT–PCR) demonstrated that wtp53 was successfully transfected into Y79 cells in the plasmid with microbubbles and ultrasound group and in the plasmid with liposomes group, but not in the plasmid with ultrasound group or in the untreated control group. Flow cytometry showed that apoptosis was highest in the microbubbles and ultrasound group (25.58%) compared with the plasmid with liposomes group (19.50%), and the other two groups (< 10%). RT–PCR also showed that the wtp53 gene was successfully transfected into solid tumours in the plasmid with microbubbles and ultrasound group. This study provides preliminary evidence in support of a potential new approach to RB gene therapy.
Collapse
Affiliation(s)
- J Luo
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing University of Medical Sciences, Chongqing, China
- Department of Ophthalmology, Zhong Shan Hospital, Chongqing, China
| | - X Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing University of Medical Sciences, Chongqing, China
| | - L Diao
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing University of Medical Sciences, Chongqing, China
| | - Z Wang
- Institute of Ultrasonic Imaging, The Second Affiliated Hospital of Chongqing University of Medical Sciences, Chongqing, China
| |
Collapse
|
14
|
Grochola LF, Zeron-Medina J, Mériaux S, Bond GL. Single-nucleotide polymorphisms in the p53 signaling pathway. Cold Spring Harb Perspect Biol 2010; 2:a001032. [PMID: 20452958 PMCID: PMC2857176 DOI: 10.1101/cshperspect.a001032] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The p53 tumor suppressor pathway is central both in reducing cancer frequency in vertebrates and in mediating the response of commonly used cancer therapies. This article aims to summarize and discuss a large body of evidence suggesting that the p53 pathway harbors functional inherited single-nucleotide polymorphisms (SNPs) that affect p53 signaling in cells, resulting in differences in cancer risk and clinical outcome in humans. The insights gained through these studies into how the functional p53 pathway SNPs could help in the tailoring of cancer therapies to the individual are discussed. Moreover, recent work is discussed that suggests that many more functional p53 pathway SNPs are yet to be fully characterized and that a thorough analysis of the functional human genetics of this important tumor suppressor pathway is required.
Collapse
Affiliation(s)
- Lukasz F Grochola
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | | | | | | |
Collapse
|
15
|
Novel amino-modified silica nanoparticles as efficient vector for hepatocellular carcinoma gene therapy. Med Oncol 2009; 27:1200-7. [PMID: 19949900 DOI: 10.1007/s12032-009-9359-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 11/04/2009] [Indexed: 01/03/2023]
Abstract
Due to the ineffective conventional treatment for hepatocellular carcinoma (HCC), the nonviral gene delivery system has been proved to be an attractive alternative to HCC therapy. In this work, we have developed a kind of new self-assembled nanoparticles, which were named as amino-modified silica nanoparticles (AMSNs). Scanning electron microscopy and zeta potential results demonstrated that AMSNs had a diameter of 20-30 nm and positive surface charges of +11.3 mV, respectively. The AMSNs could bind DNA strongly and protect DNA from degradation, which was confirmed by DNA-binding assay and serum protection assay. Furthermore, AMSNs could transfer foreign DNA into targeted cells with high transfection efficiency and little cytotoxicity. Combined with the p53 gene, AMSNs could transfect pp53-EGFP in HepG2 cells and result in a high-level of p53 mRNA and protein expressions. The nude mice treated with AMSNs/pp53-EGFP complexes showed significant tumor growth inhibition. Our results showed the AMSNs, an efficient gene vector, had the potential of gene therapy for HCC.
Collapse
|
16
|
Solyakov L, Sayan E, Riley J, Pointon A, Tobin AB. Regulation of p53 expression, phosphorylation and subcellular localization by a G-protein-coupled receptor. Oncogene 2009; 28:3619-30. [PMID: 19648965 PMCID: PMC2875175 DOI: 10.1038/onc.2009.225] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 03/24/2009] [Accepted: 06/05/2009] [Indexed: 12/31/2022]
Abstract
G-protein-coupled receptors (GPCRs) have been extremely successful drug targets for a multitude of diseases from heart failure to depression. This superfamily of cell surface receptors have not, however, been widely considered as a viable target in cancer treatment. In this study we show that a classical G(q/11)-coupled GPCR, the M(3)-muscarinic receptor, was able to regulate apoptosis through receptors that are endogenously expressed in the human neuroblastoma cell line, SH-SY5Y, and when ectopically expressed in Chinese hamster ovary (CHO) cells. Stimulation of the M(3)-muscarinic receptor was shown to inhibit the ability of the DNA-damaging chemotherapeutic agent, etoposide, from mediating apoptosis. This protective response in CHO cells correlated with the ability of the receptor to regulate the expression levels of p53. In contrast, stimulation of endogenous muscarinic receptors in SH-SY5Y cells did not regulate p53 expression but rather was able to inhibit p53 translocation to the mitochondria and p53 phosphorylation at serine 15 and 37. This study suggests the possibility that a GPCR can regulate the apoptotic properties of a chemotherapeutic DNA-damaging agent by regulating the expression, subcellular trafficking and modification of p53 in a manner that is, in part, dependent on the cell type.
Collapse
Affiliation(s)
- L Solyakov
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | | | | | | | | |
Collapse
|
17
|
Panigrahi K, Eggen M, Maeng JH, Shen Q, Berkowitz DB. The alpha,alpha-difluorinated phosphonate L-pSer-analogue: an accessible chemical tool for studying kinase-dependent signal transduction. CHEMISTRY & BIOLOGY 2009; 16:928-36. [PMID: 19778720 PMCID: PMC2766077 DOI: 10.1016/j.chembiol.2009.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 07/24/2009] [Accepted: 08/03/2009] [Indexed: 11/21/2022]
Abstract
This overview focuses on the (alpha,alpha-difluoromethylene)phosphonate mimic of phosphoserine (pCF(2)Ser) and its application to the study of kinase-mediated signal transduction-pathways of great interest to drug development. The most versatile modes of access to these chemical biological tools are discussed, organized by method of PCF(2)-C bond formation. The pCF(2)-Ser mimic may be site-specifically incorporated into peptides (SPPS) and proteins (expressed protein ligation). This isopolar, dianionic pSer mimic results in a "constitutive phosphorylation" phenotype and is seen to support native protein-protein interactions that depend on serine phosphorylation. Signal transduction pathways studied with this chemical biological approach include the regulation of p53 tumor suppressor protein activity and of melatonin production. Given these successes, the future is bright for the use of such "teflon phospho-amino acid mimics" to map kinase-based signaling pathways.
Collapse
Affiliation(s)
- Kaushik Panigrahi
- Department of Chemistry, University of Nebraska, Lincoln, NE, 68588
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - MariJean Eggen
- Department of Chemistry, University of Nebraska, Lincoln, NE, 68588
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Jun-Ho Maeng
- Department of Chemistry, University of Nebraska, Lincoln, NE, 68588
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Quanrong Shen
- Department of Chemistry, University of Nebraska, Lincoln, NE, 68588
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | | |
Collapse
|
18
|
Grochola LF, Vazquez A, Bond EE, Würl P, Taubert H, Müller TH, Levine AJ, Bond GL. Recent natural selection identifies a genetic variant in a regulatory subunit of protein phosphatase 2A that associates with altered cancer risk and survival. Clin Cancer Res 2009; 15:6301-8. [PMID: 19773383 DOI: 10.1158/1078-0432.ccr-09-0797] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE A regulated p53-dependent stress response is crucial in suppressing tumor formation and mediating the response to commonly used cancer therapeutics. However, little is known about the human, inherited genetics of this important signaling pathway. EXPERIMENTAL DESIGN Studies of human genetic variants in the p53 tumor suppressor gene and MDM2 oncogene have shown that single nucleotide polymorphisms (SNP) can affect p53 signaling, confer cancer risk, and alter outcome, and also suggest that the pathway is under evolutionary selective pressure. Here, we attempt to accelerate the identification of functional p53 pathway SNPs by incorporating these characteristics into an analysis of 142 genes that are known to affect p53 signaling. RESULTS We report that a genomic scan for recent natural selection denotes that of the 142 genes studied, the PPP2R5E gene that encodes a regulatory subunit of the tumor suppressing protein phosphatase 2A resides in a naturally selected genomic region. We go on to show that a selected SNP in PPP2R5E (epsilon-SNP2) associates with significant allelic differences in the onset (up to 19.2 years; P = 0.0002) and risk (odds ratio, up to 8.1; P = 0.0009) of soft tissue sarcoma development, as well as overall survival (relative risk, up to 3.04; P = 0.026). CONCLUSIONS The PPP2R5E gene is identified as harboring genetic variants that can affect human cancer and are possibly under evolutionary selection pressure.
Collapse
Affiliation(s)
- Lukasz F Grochola
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Altered tumor formation and evolutionary selection of genetic variants in the human MDM4 oncogene. Proc Natl Acad Sci U S A 2009; 106:10236-41. [PMID: 19497887 DOI: 10.1073/pnas.0901298106] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
A large body of evidence strongly suggests that the p53 tumor suppressor pathway is central in reducing cancer frequency in vertebrates. The protein product of the haploinsufficient mouse double minute 2 (MDM2) oncogene binds to and inhibits the p53 protein. Recent studies of human genetic variants in p53 and MDM2 have shown that single nucleotide polymorphisms (SNPs) can affect p53 signaling, confer cancer risk, and suggest that the pathway is under evolutionary selective pressure (1-4). In this report, we analyze the haplotype structure of MDM4, a structural homolog of MDM2, in several different human populations. Unusual patterns of linkage disequilibrium (LD) in the haplotype distribution of MDM4 indicate the presence of candidate SNPs that may also modify the efficacy of the p53 pathway. Association studies in 5 different patient populations reveal that these SNPs in MDM4 confer an increased risk for, or early onset of, human breast and ovarian cancers in Ashkenazi Jewish and European cohorts, respectively. This report not only implicates MDM4 as a key regulator of tumorigenesis in the human breast and ovary, but also exploits for the first time evolutionary driven linkage disequilibrium as a means to select SNPs of p53 pathway genes that might be clinically relevant.
Collapse
|
20
|
Shaked H, Shiff I, Kott-Gutkowski M, Siegfried Z, Haupt Y, Simon I. Chromatin immunoprecipitation-on-chip reveals stress-dependent p53 occupancy in primary normal cells but not in established cell lines. Cancer Res 2009; 68:9671-7. [PMID: 19047144 DOI: 10.1158/0008-5472.can-08-0865] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The p53 tumor suppressor protein is a transcription factor that plays a key role in the cellular response to stress and cancer prevention. Upon activation, p53 regulates a large variety of genes causing cell cycle arrest, apoptosis, or senescence. We have developed a p53-focused array, which allows us to investigate, simultaneously, p53 interactions with most of its known target sequences using the chromatin immunoprecipitation (ChIP)-on-chip methodology. Applying this technique to multiple cell types under various growth conditions revealed a profound difference in p53 activity between primary cells and established cell lines. We found that, in peripheral blood mononuclear cells, p53 exists in a form that binds only a small subset of its target regions. Upon exposure to genotoxic stress, the extent of targets bound by p53 significantly increased. By contrast, in established cell lines, p53 binds to essentially all of its targets irrespective of stress and cellular fate (apoptosis or arrest). Analysis of gene expression in these established lines revealed little correlation between DNA binding and the induction of gene expression. Our results suggest that nonactivated p53 has limited binding activity, whereas upon activation it binds to essentially all its targets. Additional triggers are most likely required to activate the transcriptional program of p53.
Collapse
Affiliation(s)
- Helena Shaked
- Department of Molecular Biology, Microarray Service Laboratory, The Core Research Facility, and Lautenberg Center for General and Tumor Immunology, Hebrew University Medical School, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
21
|
Vazquez A, Bond EE, Levine AJ, Bond GL. The genetics of the p53 pathway, apoptosis and cancer therapy. Nat Rev Drug Discov 2008; 7:979-87. [PMID: 19043449 DOI: 10.1038/nrd2656] [Citation(s) in RCA: 511] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The p53 pathway has been shown to mediate cellular stress responses; p53 can initiate DNA repair, cell-cycle arrest, senescence and, importantly, apoptosis. These responses have been implicated in an individual's ability to suppress tumour formation and to respond to many types of cancer therapy. Here we focus on how best to use knowledge of this pathway to tailor current therapies and develop novel ones. Studies of the genetics of p53 pathway components - in particular p53 itself and its negative regulator MDM2 - in cancer cells has proven useful in the development of targeted therapies. Furthermore, inherited single nucleotide polymorphisms in p53 pathway genes could serve a similar purpose.
Collapse
Affiliation(s)
- Alexei Vazquez
- The Institute for Advanced Study, Einstein Drive, Princeton, New Jersey, 08540, USA
| | | | | | | |
Collapse
|
22
|
Affiliation(s)
- Abigail Walton
- Palliative Care Hope Healthcare, Greenwich Hospital, Sydney, New South Wales, Australia
| | - Andrew L. Broadbent
- Palliative Care Hope Healthcare, Greenwich Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
23
|
Bruyere F, Corcoran NM, Berdjis N, Namdarian B, Pedersen J, Ockrim J, Voelzke BB, Costello AJ, Hovens CM. Aurora kinase B is an independent protective factor in superficial bladder tumours with a dysfunctional G1 checkpoint. BJU Int 2008; 102:247-52. [DOI: 10.1111/j.1464-410x.2008.07572.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
24
|
Liu W, Ip MM, Podgorsak MB, Das GM. Disruption of estrogen receptor alpha-p53 interaction in breast tumors: a novel mechanism underlying the anti-tumor effect of radiation therapy. Breast Cancer Res Treat 2008; 115:43-50. [PMID: 18481172 DOI: 10.1007/s10549-008-0044-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 04/28/2008] [Indexed: 01/08/2023]
Abstract
Inactivation of tumor suppressor p53 is one of the most frequent events in cancer. Unlike many other cancers, however, p53 gene mutations are infrequent in breast cancers, as about 80% of breast tumors contain wild type p53. The mechanisms underlying functional inactivation of wild type p53 in breast cancer have remained elusive. Besides, how p53 gets activated in breast tumors subjected to radiation therapy remains unknown. We recently reported that in MCF-7 breast cancer cells, estrogen receptor alpha (ERalpha) directly binds to p53 and represses its function. Furthermore, the ERalpha-p53 interaction was disrupted by ionizing radiation. These observations have important translational implications especially as there are no reliable cellular or molecular criteria for rational radiotherapy for breast cancer. Here we report our studies towards addressing this important issue, using an MCF-7 breast cancer xenograft model in mice. Radiation effectively inhibits growth of these tumors and stabilizes p53, but has no observable effect on ERalpha protein level. Importantly, chromatin immunoprecipitation (ChIP) assays demonstrated that ERalpha interacts with p53 bound to endogenous target gene promoters in tumors in vivo, and this interaction is considerably reduced in response to radiotherapy although p53 level is increased. Concomitant with its effect on ERalpha-p53 interaction, radiation increases p53-mediated transcriptional activation of several target genes and increases p53-mediated transcriptional repression of survivin. Our studies show that disruption of ERalpha-p53 interaction in vivo resulting in restoration of functional p53 is a cellular response to radiation. Radiation could be affecting ERalpha and/or p53 directly or it could be influencing other proteins associated with the ERalpha-p53 complex. To the best of our knowledge, this is the first report on analysis of DNA-protein-protein interaction occurring on endogenous gene promoters in vivo in breast tumor tissues. These findings suggest that alleviating the inhibitory effect of ERalpha on p53 could be one of the molecular mechanisms underlying activation of p53 by radiation in breast tumors, and therefore, could be exploited to develop more effective ways of combining radiation therapy with systemic therapies such as hormonal therapy and chemotherapy.
Collapse
Affiliation(s)
- Wensheng Liu
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | |
Collapse
|
25
|
Bensaad K, Vousden KH. p53: new roles in metabolism. Trends Cell Biol 2007; 17:286-91. [PMID: 17481900 DOI: 10.1016/j.tcb.2007.04.004] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 03/07/2007] [Accepted: 04/23/2007] [Indexed: 11/22/2022]
Abstract
Virtually all cancers show metabolic changes that result in upregulation of glycolysis and glucose consumption. Although discovered in the 1920s, how this glycolytic switch happens, and whether it is a cause or a consequence of the malignant process, has remained a matter of debate. The p53 tumor suppressor gene, discovered some 30 years ago, has a key role in preventing cancer development. Recent discoveries revealing new functions for p53 in the regulation of glucose metabolism and oxidative stress have brought together these two venerable fields of cancer biology. These activities of p53 appear to be key in tumor suppression, and shed some light on the pathways that underlie the metabolic changes in cancer cells.
Collapse
Affiliation(s)
- Karim Bensaad
- The Beatson Institute for Cancer Research, Glasgow, UK
| | | |
Collapse
|
26
|
Lee JM, Mao JT, Krysan K, Dubinett SM. Significance of cyclooxygenase-2 in prognosis, targeted therapy and chemoprevention of NSCLC. Future Oncol 2007; 3:149-53. [PMID: 17381414 DOI: 10.2217/14796694.3.2.149] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Evaluation of: Tsubochi H, Nobuyuki S, Hiyama M et al.: Combined analysis of cyclooxygenase-2 expression with p53 and Ki-67 in nonsmall cell lung cancer. Ann. Thorac. Surg. 82(4), 1198–1204 (2006).The report by Tsubochi and colleagues adds to the growing evidence indicating that tumor cyclo-oxygenase (Cox)-2 has a multifaceted role in conferring the malignant phenotype of non-small-cell lung cancer (NSCLC), and provides insight into the use of markers to provide prognostic information. Cox-2 has been implicated in apoptosis resistance, angiogenesis, decreased host immunity and enhanced invasion and metastasis, and, thus, has a critical involvement in carcinogenesis. This study, as well as others, has contributed to providing an insight into opportunities for targeted therapies in NSCLC. Cox-2 is one of the novel targets being studied for lung cancer therapy and chemoprevention.
Collapse
Affiliation(s)
- Jay M Lee
- Division of Cardiothoracic Surgery, Department of Surgery, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | | | | | | |
Collapse
|
27
|
Shu KX, Li B, Wu LX. The p53 network: p53 and its downstream genes. Colloids Surf B Biointerfaces 2007; 55:10-8. [PMID: 17188467 DOI: 10.1016/j.colsurfb.2006.11.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Revised: 10/17/2006] [Accepted: 11/03/2006] [Indexed: 12/13/2022]
Abstract
The tumor-suppressor gene p53 and its downstream genes consist of a complicated gene network. p53 is a key molecular node in the network, which is activated in response to several cellular signals resulting in the maintenance of genetic stability. Several cellular signals may activate the p53 network. When the expression of P53 is elevated, P53-MDM2 module and the ubiquitin system can accurately regulate the expression level of P53. P53 can bind to specific DNA sequence, activate its downstream genes expression, and control cell-cycle arrest, DNA repair, and apoptosis. Elucidating the function of p53 gene network will help understand the interaction mechanisms of p53 and its downstream genes.
Collapse
Affiliation(s)
- Kun-Xian Shu
- College of Bioinformation, Chongqing University of Posts and Telecommunications, Chongqing 400065, China.
| | | | | |
Collapse
|
28
|
Guan YS, La Z, Yang L, He Q, Li P. p53 gene in treatment of hepatic carcinoma: status quo. World J Gastroenterol 2007; 13:985-992. [PMID: 17373730 PMCID: PMC4146884 DOI: 10.3748/wjg.v13.i7.985] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 12/12/2006] [Accepted: 01/16/2007] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the 10 most common cancers worldwide. There is no ideal treatment for HCC yet and many researchers are trying to improve the effects of treatment by changing therapeutic strategies. As the majority of human cancers seem to exhibit either abnormal p53 gene or disrupted p53 gene activation pathways, intervention to restore wild-type p53 (wt-p53) activities is an attractive anti-cancer therapy including HCC. Abnormalities of p53 are also considered a predisposition factor for hepatocarcinogenesis. p53 is frequently mutated in HCC. Most HCCs have defects in the p53-mediated apoptotic pathway although they carry wt-p53. High expression of p53 in vivo may exert therapeutic effects on HCC in two aspects: (1) High expression of exogenous p53 protein induces apoptosis of tumor cells by inhibiting proliferation of cells through several biologic pathways and (2) Exogenous p53 renders HCC more sensitive to some chemotherapeutic agents. Several approaches have been designed for the treatment of HCC via the p53 pathway by restoring the tumor suppression function from inactivation, rescuing the mutated p53 gene from instability, or delivering therapeutic exogenous p53. Products with p53 status as the target have been studied extensively in vitro and in vivo. This review elaborates some therapeutic mechanisms and advances in using recombinant human adenovirus p53 and oncolytic virus products for the treatment of HCC.
Collapse
Affiliation(s)
- Yong-Song Guan
- Department of Radiology and Oncology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China.
| | | | | | | | | |
Collapse
|