Association of P53 and P21(CDKN1A/WAF1/CIP1) Polymorphisms with Oral Cancer in Taiwan Patients

Total Page:16

File Type:pdf, Size:1020Kb

Association of P53 and P21(CDKN1A/WAF1/CIP1) Polymorphisms with Oral Cancer in Taiwan Patients ANTICANCER RESEARCH 27: 1559-1564 (2007) Association of p53 and p21(CDKN1A/WAF1/CIP1) Polymorphisms with Oral Cancer in Taiwan Patients DA-TIAN BAU1,4*, MING-HSUI TSAI2*, YEN-LI LO6, CHIN-MOO HSU1, YUHSIN TSAI4, CHENG-CHUN LEE1 and FUU-JEN TSAI3,5 Departments of 1Medical Research, 2Otolaryngology and 3Pediatrics, China Medical University Hospital, Taichung; 4Graduate Institute of Chinese Medical Science and 5College of Chinese Medicine, China Medical University, Taichung; 6Division of Biostatistics and Bioinformatics, National Health Research Institutes, Taiwan, R.O.C. Abstract. Background: The tumor suppressor gene p53 and Oral cancer is one of the most commonly diagnosed cancers its downstream effector p21(CDKN1A/WAF1/CIP1) are in the world, the fourth most common in Taiwan. Its rising thought to play major roles in the development of human incidence and mortality poses a formidable challenge to malignancy. Polymorphic variants of p53, at codon 72, and oncologists. Early premalignant oral lesions, such as CDKN1A, at codon 31, have been associated with cancer leukoplakia, appear as a white patch in the oral cavity of susceptibility, but few studies have investigated their effect on betel and tobacco consumers; five to ten percent progress oral cancer risk. Materials and Methods: In this hospital-based toward malignancy (1). Therefore, the identification of a case-control study, the association of p53 codon 72 and biomarker for screening the high-risk population for CDKN1A codon 31 polymorphisms with oral cancer risk in a increased predisposition to cancer is of utmost importance Taiwanese population were investigated. In total, 137 patients for primary prevention and early anticancer intervention. with oral cancer and 105 age-matched controls recruited from The protein p21(Cdkn1a/Waf1/Cip1), encoded by the the Chinese Medical Hospital in Central Taiwan were CDKN1A locus, is a universal inhibitor of cyclin-dependent genotyped. Results: We found a significant difference in the kinases (Cdks), which suggests its widespread role in frequency of the p53 genotype, but not the CDKN1A genotype, regulating the cell cycle. The human CDKN1A gene consists between the oral cancer and control groups. Those who had of three exons of 68, 450 and 1600 bp (2). In normal cells, Arg/Arg at p53 codon 72 showed a 2.68-fold (95% confidence p21 exists predominantly in quaternary complexes with interval=1.19-6.01) increased risk of oral cancer compared to cyclins, Cdks, and PCNA to inhibit the activity of Cdks and those with Pro/Pro. The distribution of the combination of p53 control the G1- to S-phase transition (3). The CDKN1A codon 72 and CDKN1A codon 31 was different in the oral gene has a p53 transcriptional regulatory motif and cells cancer and control groups. The percentages of three subgroups lacking functional p53 tumor suppressor protein express with the p53 GG homozygote were all higher in the oral cancer very low levels of p21, suggesting that p53 regulates group, and the risky double homozygote, p53/CDKN1A CDKN1A expression directly (4). The expression of p21 GG/CC form, was almost 9-fold higher than the control group. induces differentiation of normal and transformed cells, and Conclusion: Our findings suggest that the homozygous Arg the involvement of p21 in terminal differentiation has been allele of the p53 codon 72 may be associated with the observed in several cell systems (5, 6). Differential development of oral cancer and be a useful marker for primary regulation of p21 by p53 and retinoblastoma has been prevention and anticancer intervention. reported in cellular response to oxidative stress (7). In addition, several recent studies suggest a role for p21 in apoptosis (8). Quercetin-induced apoptosis in hepatocytes was also associated with the regulation of p21 protein *Both authors contributed equally to this work. expression in a p53-independent pathway (9). In view of the central role of p21 in inducing growth Correspondence to: Fuu-Jen Tsai, MD, Ph.D., Department of arrest, terminal differentiation, or apoptosis, alterations in Pediatrics, Medical Genetics and Medical Research, China Medical the CDKN1A gene and its expression may play a vital role in University Hospital, 2 Yuh-Der Road, Taichung, 404 Taiwan, R.O.C. Tel: +88 6422052121 Ext 7080, Fax: +88 6422033295, the pathogenesis of cancer. Alterations in CDKN1A e-mail: [email protected] expression have been observed in a wide variety of human cancers, including ovarian, uterine, cervical, colorectal, Key Words: p53, p21, polymorphism, oral cancer, carcinogenesis. hepatocellular, and head and neck carcinomas (10-12). 0250-7005/2007 $2.00+.40 1559 ANTICANCER RESEARCH 27: 1559-1564 (2007) Currently, knowledge of the biological and clinical relevance categorical variables. Information on these factors was identified of CDKN1A alterations in oral lesions is meager (13, 14). as established habits when occurring at least twice a week for years. Differential expression of p21 protein in tobacco-related Our study was approved by the Institutional Review Board of the China Medical University Hospital and written-informed consent oral tumorigenesis in the Indian population was reported was obtained from all participants. (15). As well known, in response to DNA damage, p21 is a key mediator of the G0-G1 cell cycle arrest induced by p53. Genotyping assays. Genomic DNA was prepared from peripheral Further study revealed that p21 also interacted with PCNA blood leukocytes using a QIAamp Blood Mini Kit (Blossom, Taipei, to cause both G1 and G2 cell cycle arrest in p53-deficient Taiwan) and further processed according to a previous paper (29). cells (5, 16, 17). A novel polymorphism of the CDKN1A Briefly, for p53 codon 72, the primers 5'-TCCCCCTT gene in codon 149 was found in the Indian population and GCCGTCCCAA-3' and 5'-CGTGCAAGTCACAGACTT-3' were used (30), and for CDKN1A codon 31, the primers 5'-GTCAGAA was considered as a marker of genetic susceptibility to CCGGCTGGGGATG-3' and 5'-CTCCTCCCAA CTCATCCCGG- esophageal and oral cancers (18, 19). Bioinformatics 3' were used (31). The following cycling conditions were performed: analysis revealed that the so-called polymorphism of codon one cycle at 94ÆC for 5 min; 35 cycles of 94ÆC for 20 sec, 58ÆC for 149 is not a susceptible site (20). 20 sec and 72ÆC for 20 sec; and a final extension at 72ÆC for 10 min. The p53 gene is a tumor suppressor gene, whose function The PCR products were studied after digestion with BstU1 restriction is partially mediated by transactivating the CDKN1A enzyme for p53 codon 72, or with BlpI for CDKN1A codon 31. promoter which controls the cell cycle and prevents tumor Statistical analyses. To ensure that the controls used were formation (21). It was found that premalignant lesions and representative of the general population and to exclude the squamous cell carcinomas (SCCs) with mutant p53 protein possibility of genotyping error, deviation of the genotype overexpressed p21 (19). p21 immuno-positive well- frequencies of p53 codon 72 or CDKN1A codon 31 single differentiated tumors with p53 missense mutations probably nucleotide polymorphism in the control subjects from those harbor a p21-dependent differentiation pathway activated expected under the Hardy-Weinberg equilibrium was assessed through a p53-independent mechanism (22). Taken using the goodness-of-fit test. Pearson's ¯2 test or Fisher's exact test together, these effects reflect the complexity of the p53/p21 (when the expected number in any cell was less than five) was used to compare the distribution of the p53 and CDKN1A genotypes pathways of cell cycle regulation and differentiation in the between cases and controls. Cancer risk associated with the pathogenesis of oral cancer. Mutations in either p53 or genotypes was estimated as odds ratio (ORs) and 95% confidence CDKN1A were detected in some tumor cells (21, 23) and intervals (CIs) using unconditional logistic regression with polymorphisms of p53 codon 72 or CDKN1A codon 31 were adjustment for age, smoking, alcohol consumption and betel found to be associated with many tumors (24-27). chewing habits. Data was recognized as significant when the Since p53 gene mutations are the most common cancer- statistical p-value was less than 0.05. related genetic alterations, found in ~50% of human cancer (28), and p53 regulates p21 expression (4), we were Results interested in looking for a susceptible site in p53 and CDKN1A genes together in Taiwan population, especially in The mean ages of the oral cancer patients and the controls oral cancer patients. Thus, the main goal of the study was were 53.27 (SD=12.27) and 53.02 (SD=10.08) years, to examine the joint effect of polymorphisms in codon 72 of respectively. The proportion of men and women were the p53 gene and codon 31 of the CDKN1A gene with oral almost the same in the patient and control groups (i.e., 117 cancer in a Central Taiwan population. men and 20 women). Characteristically, the most common sites of primary tumor were the tongue and buccal mucosa Materials and Methods in this Central Taiwan population. Approximately 38% (52 of 137) of the study cohort consisted of patients diagnosed Study population and sample collection. One hundred and thirty- with TNM late stage (III and IV) oral cancer (data not seven patients diagnosed with oral cancer were recruited at the shown). outpatient clinics of general surgery between 1997-2005 at the China Medical University Hospital, Taichung, Taiwan, Republic of The frequency of the alleles for the p53 codon 72 and China. All patients voluntarily participated, completed a self- CDKN1A codon 31 between oral cancer and control groups administered questionnaire and provided peripheral blood samples. is shown in Table I. The Arg allele at p53 codon 72 was One hundred and five non-oral cancer healthy people as controls significantly associated with oral cancer risk (p=0.02).
Recommended publications
  • Gene Therapy of Prostate Cancer: Current and Future Directions
    Endocrine-Related Cancer (2002) 9 115–139 Gene therapy of prostate cancer: current and future directions N J Mabjeesh, H Zhong and J W Simons Winship Cancer Institute, Department of Hematologyand Oncology,EmoryUniversitySchool of Medicine, 1365 Clifton Road, Suite B4100, Atlanta, Georgia 30322, USA (Requests for offprints should be addressed to J W Simons; Email: jonathan—[email protected]) Abstract Prostate cancer (PCA) is the second most common cause of death from malignancyin American men. Developing new approaches for gene therapyfor PCA is critical as there is no effective treatment for patients in the advanced stages of this disease. Current PCA gene therapyresearch strategies include cytoreductive approaches (immunotherapy and cytolytic/pro-apoptotic) and corrective approaches (replacing deleted or mutated genes). The prostate is ideal for gene therapy. It is an accessoryorgan, offers unique antigens (prostate-specific antigen, prostate-specific membrane antigen, human glandular kallikrein 2 etc.) and is stereotacticallyaccessible for in situ treatments. Viral and non-viral means are being used to transfer the genetic material into tumor cells. The number of clinical trials utilizing gene therapymethods for PCA is increasing. We review the multiple issues involved in developing effective gene therapystrategies for human PCA and earlyclinical results. Endocrine-Related Cancer (2002) 9 115–139 Introduction stration of the impact of chemotherapy (mitoxantrone+ prednisone) on quality of life as compared with prednisone New therapeutics like gene therapy are needed urgently for alone in advanced hormone-refractory PCA. At the present advanced prostate cancer (PCA). PCA remains the most time, chemotherapy should be considered as a palliative or common solid tumor and the second leading cause of cancer- investigational treatment in patients with symptomatic andro- related deaths among men in the USA.
    [Show full text]
  • RESEARCH ARTICLE MCBS Mol Cell Biomed Sci
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Molecular and Cellular Biomedical Sciences (E-Journal) Widowati W, et al. Effect of TNFa and IFNg Toward Apoptosis in Breast Cancer Cells RESEARCH ARTICLE MCBS Mol Cell Biomed Sci. 2018; 2(2): 60-9 DOI: 10.21705/mcbs.v2i2.21 Direct and Indirect Effect of TNFa and IFNg Toward Apoptosis in Breast Cancer Cells Wahyu Widowati1, Diana Krisanti Jasaputra1, Sutiman Bambang Sumitro2, Mochamad Aris Widodo3, Ervi Afifah4, Rizal Rizal4, Dwi Davidson Rihibiha4, Hanna Sari Widya Kusuma4, Harry Murti5, Indra Bachtiar5, Ahmad Faried6 1Medical Research Center, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia 2Department of Biology, Faculty of Science, Brawijaya University, Malang, East Java, Indonesia 3Pharmacology Laboratory, Faculty of Medicine, Brawijaya University, Malang, East Java, Indonesia 4Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung West Java, Indonesia 5Stem Cell and Cancer Institute, Jakarta, Indonesia 6Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia Background: Breast cancer (BC) is the leading cause of death cancer in women. Cancer therapies using TNFα and IFNγ have been recently developed by direct effects and activation of immune responses. This study was performed to evaluate the effects of TNFα and IFNγ directly, and TNFα and IFNγ secreted by Conditioned Medium-human Wharton’s Jelly Mesenchymal Stem Cells (CM-hWJMSCs) toward apoptosis of BC cells (MCF7). Materials and Methods: BC cells were induced by TNFα and IFNγ in 175 and 350ng/mL, respectively. CM-hWJMSCs were produced by co-culture hWJMSCs and NK cells that secreted TNFα, IFNγ, perforin (Prf1), granzyme B (GzmB) for treating BC cells.
    [Show full text]
  • Survivin, Versatile Modulation of Cell Division and Apoptosis in Cancer
    Oncogene (2003) 22, 8581–8589 & 2003 Nature Publishing Group All rights reserved 0950-9232/03 $25.00 www.nature.com/onc Survivin, versatile modulation of cell division and apoptosis in cancer Dario C Altieri*,1 1Department of Cancer Biology and the Cancer Center, LRB-428, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA Survivin is a member of the inhibitor of apoptosis (IAP) spliced survivin transcripts (Figure 1). In addition to gene family that has attracted attention from several wild-type survivin (142 amino acids), two survivin viewpoints of basic and translational research. Its cell isoforms are generated by insertion of an alternative cycle-regulated expression at mitosis and association with exon 2 (survivin-2B, 165 amino acids) or removal of the mitotic apparatus have been of interest to cell exon 3 (survivin-DEx-3, 137 amino acids) (Mahotka biologists studying faithful segregation of sister chroma- et al., 1999). In survivin-DEx-3, the splicing event tids and timely separation of daughter cells. Investigators introduces a frame shift that generates a unique – interested in mechanisms of apoptosis have found survivin COOH terminus of potential functional significance an evolving challenge:while survivin inhibits apoptosis in (Mahotka et al., 2002) (Figure 1). vitro and in vivo, this pathway may be more selective as A unique property of survivin is a sharp cell-cycle- compared to cytoprotection mediated by other IAPs. dependent expression at mitosis. This is largely, but not Finally, basic and translational researchers in cancer exclusively, controlled at the level of gene transcription biology have converged on survivin as a pivotal cancer and involves canonical CDE/CHR boxes (Badie et al., gene, not simply for its sharp expression in tumors and not 2000) in the survivin promoter (Kobayashi et al., 1999; in normal tissues, but also for the potential exploitation of Li and Altieri, 1999b), acting as potential G1-repressor this pathway in cancer diagnosis and therapy.
    [Show full text]
  • Tumor Necrosis Factor-Related Apoptosis Inducing Ligand Overexpression and Taxol Treatment Suppresses the Growth of Cervical Cancer Cells in Vitro and in Vivo
    5744 ONCOLOGY LETTERS 15: 5744-5750, 2018 Tumor necrosis factor-related apoptosis inducing ligand overexpression and Taxol treatment suppresses the growth of cervical cancer cells in vitro and in vivo XIAOJIE SUN1, MANHUA CUI2, DING WANG3, BAOFENG GUO1 and LING ZHANG3 1Department of Plastic Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033; 2Department of Gynaecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin 130022; 3Department of Pathophysiology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021 P.R. China Received May 23, 2017; Accepted January 17, 2018 DOI: 10.3892/ol.2018.8071 Abstract. Tumor necrosis factor-related apoptosis inducing Introduction ligand (TRAIL) is a member of tumor necrosis factor (TNF) superfamily and functions to promote apoptosis by binding to Cervical cancer is a common malignancy with an estimated cell surface death receptor (DR)4 and DR5. Cancer cells are 485000 new cases and 236000 deaths annually worldwide (1). more sensitive than normal cells to TRAIL-induced apoptosis, Nearly all cases of cervical cancer are caused by human and TRAIL-based therapeutic strategies have shown promise papillomavirus infection (2). Advances in novel diagnostic for the treatment of cancer. The present study investigated and therapeutic technologies have led to a considerable whether enforced overexpression of TRAIL in cervical cancer decline in cervical cancer morbidity and mortality over the cells promoted cell death in the presence or absence of Taxol, last decade (3,4). Current treatments for cervical cancer are an important first‑line cancer chemotherapeutic drug. Hela surgery, radiotherapy and chemotherapy (5); however, addi- human cervical cancer cells were transfected with a TRAIL tional therapies will be required to increase survival rates and expression plasmid, and the effects of the combination treat- reduce the need for surgery.
    [Show full text]
  • The Anticancer Gene ORCTL3 Targets Stearoyl-Coa Desaturase-1 for Tumour-Specific Apoptosis
    Oncogene (2015) 34, 1718–1728 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc ORIGINAL ARTICLE The anticancer gene ORCTL3 targets stearoyl-CoA desaturase-1 for tumour-specific apoptosis G AbuAli1, W Chaisaklert1, E Stelloo1, E Pazarentzos1, M-S Hwang1, D Qize1, SV Harding2, A Al-Rubaish3, AJ Alzahrani3,5, A Al-Ali3, TAB Sanders2, EO Aboagye4 and S Grimm1 ORCTL3 is a member of a group of genes, the so-called anticancer genes, that cause tumour-specific cell death. We show that this activity is triggered in isogenic renal cells upon their transformation independently of the cells’ proliferation status. For its cell death effect ORCTL3 targets the enzyme stearoyl-CoA desaturase-1 (SCD1) in fatty acid metabolism. This is caused by transmembrane domains 3 and 4, which are more efficacious in vitro than a low molecular weight drug against SCD1, and critically depend on their expression level. SCD1 is found upregulated upon renal cell transformation indicating that its activity, while not impacting proliferation, represents a critical bottleneck for tumourigenesis. An adenovirus expressing ORCTL3 leads to growth inhibition of renal tumours in vivo and to substantial destruction of patients’ kidney tumour cells ex vivo. Our results indicate fatty acid metabolism as a target for tumour-specific apoptosis in renal tumours and suggest ORCTL3 as a means to accomplish this. Oncogene (2015) 34, 1718–1728; doi:10.1038/onc.2014.93; published online 28 April 2014 INTRODUCTION the therapy of renal tumours relies mainly on surgery and there is Recent studies have led to the emergence of a new class of genes hardly any systemic drug treatment that can be used against 11 with a specific anticancer activity.
    [Show full text]
  • Progress in Cancer Gene Therapy
    © 2000 Nature America, Inc. 0929-1903/00/$15.00/ϩ0 www.nature.com/cgt Progress in cancer gene therapy David T. Curiel,1 Winald R. Gerritsen,2 and Mark R. L. Krul3 1Division of Human Gene Therapy, Departments of Medicine, Pathology, and Surgery, Gene Therapy Center, University of Alabama, Birmingham, Alabama 35294; 2Academic Hospital Vrije Universiteit Amsterdam, Department of Medical Oncology, Amsterdam, The Netherlands; and 3NDDO Oncology, Section of Drug Development Strategy, Amsterdam, The Netherlands. The “First International Symposium on Genetic Anticancer Agents,” which took place in Amsterdam on March 8–9, 2000, served as a forum to review the results of preclinical and clinical gene therapy studies for cancer endeavored to date. Despite the fact that gene therapy was initially conceptualized as an approach for inherited genetic disease, it is currently finding its widest employ for treating neoplastic disorders. In this regard, more than 70% of patients treated to date in human clinical gene therapy protocols have been in the context of anticancer regimens.1 Of note, the application of gene therapy for cancer has proceeded from the same rational basis as was originally conceptualized for inherited genetic disorders. Specifically, the molecular basis of these disorders is increasingly being understood, therapeutic genes are available, and alternative therapies are often lacking. Most recently, the field of gene therapy has enjoyed the realization of the first incontrovertible evidence of clinical benefit, for hemophilia and cardiovascular disease, in its first 15 years of human application.2 This recent recognition of the potential power of gene therapy, and the current lack of realizing such ends for neoplastic disease, has led to a reassessment of the field.
    [Show full text]
  • Anticancer Gene Transfer for Cancer Gene Therapy
    1 Anticancer Gene Transfer for Cancer Gene Therapy Evangelos Pazarentzos, Ph.D1, & Nicholas D. Mazarakis Ph.D2 1 Postoctoral Fellow, University of California-San Francisco (UCSF) 600 16th st, Genentech Hall, San Francisco CA, 94158, USA 2 Lucas-Lee Chair of Molecular BioMedicine & Head of Gene Therapy, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, E402 Burlington Danes Building, Du Cane Road, London W12 0NN ABSTRACT Gene therapy vectors are among the treatments currently used to treat malignant tumors. Gene therapy vectors use a specific therapeutic transgene that causes death in cancer cells. In early attempts at gene therapy, therapeutic transgenes were driven by non- specific vectors which induced toxicity to normal cells in addition to the cancer cells. Recently, novel cancer specific viral vectors have been developed that target cancer cells leaving normal cells unharmed. Here we review such cancer specific gene therapy systems currently used in the treatment of cancer and discuss the major challenges and future directions in this field. INTRODUCTION CANCER GENE THERAPY – ONCOLYSIS Viruses were discovered more than a century ago but from early times diseases like cancer and especially leukemia were attempted to be treated with viruses. Throughout the recorded history of diseases, there have been observations of cancer regression upon natural co-infection with viruses (Sinkovics and Horvath, 1993; Kelly and Russell, 2007). During the early twentieth century, based on these observations, several clinical trials were conducted via fluid transfer from animal or human bodies that were infected with viruses to infect patients with cancer (Hoster et al., 1949).
    [Show full text]
  • Knockdown of ZFPL1 Results in Increased Autophagy and Autophagy‑Related Cell Death in NCI‑N87 and BGC‑823 Human Gastric Carcinoma Cell Lines
    MOLECULAR MEDICINE REPORTS 15: 2633-2642, 2017 Knockdown of ZFPL1 results in increased autophagy and autophagy‑related cell death in NCI‑N87 and BGC‑823 human gastric carcinoma cell lines YONG-ZHENG XIE, WAN-LI MA, JI-MING MENG and XUE-QUN REN Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China Received October 1, 2015; Accepted September 28, 2016 DOI: 10.3892/mmr.2017.6300 Abstract. Macroautophagy, which will hereafter be referred Introduction to as autophagy, is an evolutionarily conserved process, during which cells recycle and remove damaged organelles Gastric cancer (GC) is the fifth most common cancer and proteins in response to cellular stress. However, the worldwide, and is currently the third leading cause of mechanisms underlying the regulation of autophagy remain cancer-associated mortality. GC is particularly prevalent in to be fully elucidated. The present study demonstrated that Asia (1). Unfortunately, the majority of patients with GC present knockdown of zinc finger protein like 1 (ZFPL1) induces with late stage cancer, and therefore require palliative chemo- autophagy and increases autophagic cell death in NCI-N87 therapy (2). Treatment of GC continues to present a challenge, and BGC-823 human gastric carcinoma cell lines. To particularly with regards to the high mortality-to-incidence examine the role of ZFPL1 in gastric carcinoma cells, ZFPL1 ratio, despite significant progress being made in early detection expression was downregulated by lentiviral infection. Zinc and treatment (3). Genetic alterations are thought to be impor- finger domain-FLAG was used to compete with ZFPL1 tant factors in the vast majority of solid tumors.
    [Show full text]
  • Antitumor Effects of Chi-Shen Extract from Salvia Miltiorrhiza and Paeoniae Radix on Human Hepatocellular Carcinoma Cells
    Acta Pharmacol Sin 2007 Aug; 28 (8): 1215–1223 Full-length article Antitumor effects of Chi-Shen extract from Salvia miltiorrhiza and Paeoniae radix on human hepatocellular carcinoma cells Sheng HU, Shi-min CHEN, Xiao-kuan LI, Rui QIN, Zhi-nan MEI1 Institute of Materia Media, South-Central University for Nationalities, Wuhan 430074, China Key words Abstract human hepatocellular carcinoma cells; Aim: To investigate the antihepatocellular carcinoma effects of Chi-Shen extract apoptosis; Bcl-2; Bax; caspase-3; Salvia (CSE) from the water-soluble compounds of Salvia miltiorrhiza and Paeoniae miltiorrhiza; Paeoniae radix radix. Methods: The effect of CSE on the growth of HepG2 cells (hepatocellular carcinoma cell line) was studied by 3-(4,5)-2,5-diphenyltetrazolium bromide assay. Apoptosis were detected through acridine orange (AO) and ethylene dibromide 1 Correspondence to Dr Zhi-nan MEI. Phn 86-27-6784-3237. (EB) staining and DNA fragmentation assay. The effect of CSE on the cell cycle of Fax 86-27-6784-3220. HepG2 cells was studied by the propidium iodide staining method. The activation E-mail [email protected] of caspases-3, -8 and -9 was examined by immunoassay kits. The transcription of Received 2006-10-31 the Bcl-2 family and p53 was detected by RT-PCR. Results: Our data revealed that Accepted 2007-02-28 CSE strongly induced HepG2 cell death in a dose- and time-dependent manner. CSE-induced cell death was considered to be apoptotic by observing the typical doi: 10.1111/j.1745-7254.2007.00606.x apoptotic morphological change by AO/EB staining and DNA fragmentation assay.
    [Show full text]
  • Imperial College London Department of Medicine
    Imperial College London Department of Medicine A Genetic Screening for the Tumour Suppressive Anticancer Genes A thesis submitted for the degree of Doctor of Philosophy Qize Ding December 2016 Supervisor: Professor Eric Lam 1 Declaration of Originality I, Qize Ding, hereby declare that I am the sole author of this thesis, the whole of which contains my original research work conducted at the Department of Medicine, Imperial College London from 2012 to 2015. Contents such as figures, data and material from other sources are appropriately cited and acknowledged in the text and a full list of reference is shown at the end of my thesis. I declare that this is the true copy of my thesis and the content of this thesis has not been and will not be submitted in any form for a higher degree from any other university or institution. 2 Acknowledgements I would like to express my sincere gratitude to my current main supervisor Professor Eric Lam, as well as my former main supervisor Professor Stefan Grimm who supervised my project for the first two and half years. Their knowledge on the research areas and valuable inputs, directions and guidance have been of great value to me throughout the project. I would also like to say many thanks to my co-supervisor Dr Mona A El-Bahrawy, who advised me on the thesis write-up and corrections. Many thanks go to my colleagues, the PhD student Motasim Masood, who was helpful and supported the MTT, clonogenic assay and apoptosis assay. Another former PhD student, Ming Hwang who helped with some Western blotts.
    [Show full text]
  • Is CRISPR/Cas9 an Option?
    G C A T T A C G G C A T genes Review Therapeutic Editing of the TP53 Gene: Is CRISPR/Cas9 an Option? Regina Mirgayazova 1, Raniya Khadiullina 1, Vitaly Chasov 1, Rimma Mingaleeva 1, Regina Miftakhova 1, Albert Rizvanov 1 and Emil Bulatov 1,2,* 1 Kazan Federal University, 420008 Kazan, Russia; [email protected] (R.M.); [email protected] (R.K.); [email protected] (V.C.); [email protected] (R.M.); [email protected] (R.M.); [email protected] (A.R.) 2 Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia * Correspondence: [email protected] Received: 6 May 2020; Accepted: 23 June 2020; Published: 25 June 2020 Abstract: The TP53 gene encodes the transcription factor and oncosuppressor p53 protein that regulates a multitude of intracellular metabolic pathways involved in DNA damage repair, cell cycle arrest, apoptosis, and senescence. In many cases, alterations (e.g., mutations of the TP53 gene) negatively affect these pathways resulting in tumor development. Recent advances in genome manipulation technologies, CRISPR/Cas9, in particular, brought us closer to therapeutic gene editing for the treatment of cancer and hereditary diseases. Genome-editing therapies for blood disorders, blindness, and cancer are currently being evaluated in clinical trials. Eventually CRISPR/Cas9 technology is expected to target TP53 as the most mutated gene in all types of cancers. A majority of TP53 mutations are missense which brings immense opportunities for the CRISPR/Cas9 system that has been successfully used for correcting single nucleotides in various models, both in vitro and in vivo.
    [Show full text]
  • Saporin As a Novel Suicide Gene in Anticancer Gene Therapy
    Cancer Gene Therapy (2007) 14, 165–173 r 2007 Nature Publishing Group All rights reserved 0929-1903/07 $30.00 www.nature.com/cgt ORIGINAL ARTICLE Saporin as a novel suicide gene in anticancer gene therapy N Zarovni1, R Vago1,2, T Solda`3, L Monaco4 and MS Fabbrini5 Department of Biological and Technological Research and Cancer Immunotherapy and Gene Therapy Program, San Raffaele H Scientific Institute, Milan, Italy We used a non-viral gene delivery approach to explore the potential of the plant saporin (SAP) gene as an alternative to the currently employed suicide genes in cancer therapy. Plasmids expressing cytosolic SAP were generated by placing the region encoding the mature plant ribosome-inactivating protein under the control of cytomegalovirus (CMV) or simian virus 40 (SV40) promoters. Their ability to inhibit protein synthesis was first tested in cultured tumor cells co-transfected with a luciferase reporter gene. In particular, SAP expression driven by CMV promoter (pCI-SAP) demonstrated that only 10 ng of plasmid per 1.6 Â 104 B16 cells drastically reduced luciferase activity to 18% of that in control cells. Direct intratumoral injection of pCI-SAP complexed with either lipofectamine or N-(2,3-dioleoyloxy-1-propyl) trimethylammonium methyl sulfate (DOTAP) in B16 melanoma-bearing mice resulted in a noteworthy attenuation of tumor growth. This antitumor effect was increased in mice that received repeated intratumoral injections. A SAP catalytic inactive mutant (SAP-KQ) failed to exert any antitumor effect demonstrating that this was specifically owing to the SAP N-glycosidase activity. Our overall data strongly suggest that the gene encoding SAP, owing to its rapid and effective action and its independence from the proliferative state of target cells might become a suitable candidate suicide gene for oncologic applications.
    [Show full text]