Cancer Gene Therapy Using a Pro-Apoptotic Gene, Caspase-3

Total Page:16

File Type:pdf, Size:1020Kb

Cancer Gene Therapy Using a Pro-Apoptotic Gene, Caspase-3 Gene Therapy (1999) 6, 1952–1959 1999 Stockton Press All rights reserved 0969-7128/99 $15.00 http://www.stockton-press.co.uk/gt Cancer gene therapy using a pro-apoptotic gene, caspase-3 K Yamabe1, S Shimizu1,2, T Ito1, Y Yoshioka1, M Nomura1, M Narita1, I Saito3, Y Kanegae3 and H Matsuda1 1The First Department of Surgery, 2Department of Medical Genetics, Biomedical Research Center, Osaka University Medical School, Suita; and 3Laboratory of Molecular Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan Caspase-3 is a member of the cysteine protease family, 3, but not the empty vector, induced extensive apoptosis which plays a crucial role in apoptosis. We applied the and reduced tumor volume when combined with etoposide human caspase-3 gene as a novel form of anticancer gene administration. However, this effect was not observed with therapy. Overexpression of human caspase-3 alone could a Bcl-2 overexpressing tumor. In conclusion, caspase-3 not induce apoptosis of tumor cell lines, but apoptosis was gene transduction accompanied by an additional death markedly enhanced by the addition of etoposide. In an stimulus may be a useful method of anticancer gene AH130 liver tumor model, transduction of human caspase- therapy, except for Bcl-2 overexpressing tumors. Keywords: caspase-3 gene; anticancer gene therapy; Bcl-2; recombinant adenovirus vector Introduction advantages of high transduction efficiency, loss of episo- mal adenoviral DNA after several cycles of cell prolifer- 1–3 Caspase-3 (also called CPP32 or apopain) is a member ation without any effect on other foreign genes, and the of the cysteine protease family, which plays an important ability to prepare high titer (Ͼ108 p.f.u./ml) viral stock 4 role in apoptosis. Apoptotic signals activated by various solutions. stimuli converge towards a common pathway, in which We previously showed that transduction of human caspases have a signaling role. More than 10 caspases caspase-3 gene into HeLa cells induced little apoptosis, 5 have been identified in mammals. Among them, cas- whereas apoptosis was substantially induced by sub- pase-3 has been implicated in cell death, based upon sequent addition of anti-Fas antibody.7 Accordingly, the 6 abnormalities found in knockout mice and the inhibition present study investigated the effect of human caspase-3 7,8 of cell death by a caspase-3 inhibitor. Caspase-3 is syn- gene transduction combined with exposure to chemo- thesized as an inactive precursor of 32 kDa that is first therapy using several cancer cell lines and a liver tumor processed into subunits (P20 and P12), and finally into model. Our results suggested the possibility of using the active subunits (P17 and P12) when a death-inducing sig- human caspase-3 gene for anticancer gene therapy. nal is applied.1–3 Therefore, expression of caspase-3 pre- cursor (pro-caspase-3) alone cannot induce apoptosis and its activation requires a death-inducing stimulus, such as Results cancer chemotherapy agents.7 Several death-inducing genes have been reported to be Effect of caspase-3 gene transduction on apoptosis in useful for anticancer gene therapy, and some of them, vitro including the thymidine kinase gene, require additional To evaluate the effect of human caspase-3 (hcasp-3) and stimuli to kill cancer cells. Such regulation could have human caspase-1 (hcasp-1) gene transduction on apoptosis, great advantages in cancer therapy, since targeting of HepG2 cells (a human hepatoma cell line) were transi- additional stimuli could allow these genes selectively to ently transduced with an hcasp-3- and hcasp-1-containing kill transduced cells. In this context, the caspase-3 gene adenovirus vector (Figure 1A) or with the empty vector should be eminently suitable for anticancer gene therapy, (as a control) together with a ␤-gal-containing adenovirus because (1) it has a strong effect when activated, and (2) vector (as a reporter gene). As shown in Figure 1B (a–c), cell death is not induced by gene transduction alone. almost all the HepG2 cells were positively stained with As the gene delivery system we used a recombinant X-gal at 10 multiplicities of infection (MOI), indicating 9,10 inactivated adenovirus vector with an expression efficient expression of the transduced genes. There were ␤ cosmid consisting of the chicken -actin promoter and some variations in the intensity of staining, possibly 11 the cytomegalovirus enhancer. This method has the caused by differences in the number of copies of the transduced genes. The extent of apoptosis was estimated by TUNEL staining. As shown in Figure 1B (d–f) and C, Correspondence: S Shimizu, The First Department of Surgery, Osaka the proportion of apoptotic cells rapidly increased to 80% University Medical School, 2-2 Yamada-oka, Suita 565-0871, Japan on 1 day after transduction with 10 MOI of the hcasp-1 Received 1 February 1999; accepted 15 July 1999 adenovirus, whereas transduction of hcasp-3 adenovirus Caspase-3 gene therapy K Yamabe et al 1953 Figure 1 Low induction of apoptosis in hcasp-3-transduced HepG2 cells. (A) Structure of recombinant adenovirus with the caspase-1 and caspase-3 expression units (AxCAOYCaspase-1, and -3). AxCAOYCaspase-1 and -3 have the cytomegalovirus enhancer (CMV.E.), chicken ␤-actin promotor, and human caspase-1 and -3, respectively, at the E1 region. (B) Representative appearance of transduced HepG2 cells. HepG2 cells were transiently transduced with hcasp-3, hcasp-1, or the empty vector (10 MOI) together with the lacZ gene-containing adenovirus vector (10 MOI). Cells were fixed and stained with X-gal buffer (X-gal), the TUNEL method (TUNEL), and Hoechst33342 (Hoechst) at 1 day after transduction. (C and D) Induction of apoptosis in hcasp-1-transduced HepG2 cells, but little effect of hcasp-3. Similar experiments to those in (B) were performed, and the proportion of apoptotic cells was estimated by TUNEL staining (C) or nuclear fragmentation (D) at the indicated times (C) and at 1 day after transduction (D). Data are expressed as the mean ± s.d. of four independent experiments. Caspase-3 gene therapy K Yamabe et al 1954 or the vector alone induced a slight and no apoptosis and AH130 cells were treated with hcasp-3 and etoposide after 3 days, respectively, findings consistent with pre- (data not shown). These findings indicated that addition vious observations.7 Similar results were also obtained by of etoposide caused marked caspase-3 activation in hcasp- assessing cells with nuclear fragmentation after staining 3 gene-transduced cells, resulting in the induction of using Hoechst33342 (Figure 1B (g–i) and D). Further- apoptosis. more, similar results were noted when HeLa cells (a human cervical carcinoma cell line) or AH130 cells (a rat Effect of hcasp-3 gene transduction plus etoposide on hepatoma cell line) were used (data not shown). These solid tumors findings indicated that hcasp-1 expression, but not hcasp- To determine whether hcasp-3 transduction was also 3 expression, could induce apoptosis by itself. The clinical effective against solid tumors, we used a liver tumor application of hcasp-1 seemed to be difficult because it is model that was created by injecting AH130 cells (a Don- not easy to regulate its apoptosis-inducing ability, so we ryu rat ascites hepatoma cell line) into the livers of Don- focused on hcasp-3. ryu rats. At 3 days after injection, tumors with a volume of about 1 cm3 were formed. Then adenovirus (5 × 108 Efficient expression of hcasp-3 in an inactive form p.f.u.) transduction was done by the direct injection Since hcasp-3 transduction caused little apoptosis, inef- method. Four days after transduction, etoposide (0.2 ficient expression of caspase-3 was a possibility. As mg/100 g body weight) was injected via the dorsal vein shown in Figure 2, however, expression of caspase-3 pro- of the penis. As shown in Figure 4A (a–d), caspase-3 was form (P32) in hcasp-3-transduced cells was four-fold efficiently expressed in hcasp-3-transduced, but not vec- higher than in non-transduced or vector-transduced cells. tor-transduced tumors, as assessed by immunohisto- We could not detect P20 (active P17 plus the prodomain), chemistry using an anti-human caspase-3 antibody that which is the product of caspase activation, in hcasp-3- did not cross-react with rat caspase-3. Consistent with the transduced cells after culture for 3 days, indicating that results obtained using HepG2 cells (Figure 1), hcasp-3 hcasp-3 gene transduction alone did not cause caspase transduction alone could not induce apoptosis as activation, a result that was consistent with the slight detected by the TUNEL assay (Figure 4Ae), whereas sub- induction of apoptosis (Figure 1). These findings indi- sequent addition of etoposide induced extensive cated that hcasp-3 gene transduction using an adenovirus apoptosis of hcasp-3-transduced tumor cells (Figure 4Ag), vector at 10 MOI resulted in efficient production of but not of vector-transduced cells (Figure 4Ah). At this inactive caspase-3 and did not induce apoptosis. dose of etoposide, there was rapid growth of the vector- transduced tumors, but growth was significantly reduced Effect of etoposide on hcasp-3-transduced cells in hcasp-3-transduced tumors (Figure 4B), possibly due to Since caspase-3 activation requires a death-inducing the induction of apoptosis. Transduction of hcasp-3 alone stimulus, we added 100 ␮m etoposide (a chemotherapy did not reduce the tumor volume (Figure 4B). These agent) to cultures from 1 day after adenovirus transduc- findings indicated that transduction of human caspase-3 tion. At this concentration, production of processed cas- was effective for inducing apoptosis and reducing tumor pase-3 (P20) was observed in hcasp-3-transduced cells but volume in a liver tumor model when applied together not in vector-transduced cells (Figure 3A).
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]
  • 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.
    [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]