• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 72
  • 52
  • 18
  • 7
  • 7
  • 7
  • 7
  • 7
  • 7
  • Tagged with
  • 100
  • 100
  • 100
  • 50
  • 50
  • 43
  • 31
  • 24
  • 23
  • 21
  • 18
  • 18
  • 17
  • 17
  • 14
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
21

Histone deacetylase inhibitors are effective therapeutic agents in nasopharyngeal carcinoma cells.

January 2006 (has links)
Wong Yue Hang Albert. / Thesis submitted in: December 2005. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2006. / Includes bibliographical references (leaves 108-119). / Abstracts in English and Chinese. / Abstract --- p.i / Acknowledgements --- p.v / List of Figures --- p.x / List of Tables --- p.xi / Chapter Chapter 1 --- Introduction --- p.1 / Chapter Chapter 2 --- Literature Review --- p.4 / Chapter 2.1 --- Nasopharyngeal Carcinoma (NPC) --- p.4 / Chapter 2.1.1 --- Anatomy of Nasopharynx --- p.4 / Chapter 2.1.2 --- Histopathology of Nasopharyngeal Carcinoma --- p.5 / Chapter 2.1.3 --- Epidemiology and Etiology of Nasopharyngeal Carcinoma --- p.5 / Chapter 2.1.3.1 --- Endemic Regions of Nasopharyngeal Carcinoma --- p.5 / Chapter 2.1.3.2 --- Gender and Age Bias --- p.6 / Chapter 2.1.3.3 --- Nasopharyngeal Carcinoma in Hong Kong --- p.6 / Chapter 2.1.3.4 --- Environmental Factors and Diet --- p.7 / Chapter 2.1.3.5 --- HLA Haplotypes and Nasopharyngeal Carcinoma --- p.9 / Chapter 2.1.4 --- Epstein-Barr Virus (EBV) and Nasopharyngeal Carcinoma --- p.10 / Chapter 2.1.4.1 --- EBV and Human Cacners --- p.10 / Chapter 2.1.4.2 --- EBV Infection --- p.10 / Chapter 2.1.4.3 --- "Latent, Clonal EBV Infection" --- p.11 / Chapter 2.1.4.4 --- EBV Latency Form --- p.11 / Chapter 2.1.4.5 --- Reactivation of EBV --- p.12 / Chapter 2.1.5 --- Molecular Pathogenesis of Nasopharyngeal Carcinoma --- p.13 / Chapter 2.1.5.1 --- Genetic Changes --- p.13 / Chapter 2.1.5.2 --- Epigenetic Changes --- p.13 / Chapter 2.1.6 --- Therapy of Nasopharyngeal Carcinoma and its Deficiency --- p.14 / Chapter 2.1.6.1 --- Radiotherapy --- p.14 / Chapter 2.1.6.2 --- Concurrent Chemoradiotherapy --- p.16 / Chapter 2.1.6.3 --- Adjuvant and Neo-adjuvant Chemotherapy --- p.17 / Chapter 2.1.6.4 --- Chemotherapy in Metastatic Nasopharyngeal Carcinoma --- p.18 / Chapter 2.1.6.5 --- Novel Therapeutic Agents and Approach --- p.19 / Chapter 2.2 --- Histone Modification and Cancer --- p.20 / Chapter 2.2.1 --- Histone Modification and Transcription Regulation --- p.20 / Chapter 2.2.2 --- Carcinogenic Effect of Aberrant HAT and HDAC Activities --- p.21 / Chapter 2.2.3 --- Structural Classes of HDAC Inhibitors --- p.24 / Chapter 2.2.4 --- Anti-Cancer Mechanisms of HDAC Inhibitors --- p.25 / Chapter 2.3 --- Suberoylanilide Hydroxamic Acid (SAHA) --- p.27 / Chapter 2.3.1 --- Anti-tumor Effect of SAHA in Various Cancer Cell Lines --- p.27 / Chapter 2.3.2 --- SAHA Mediated Non-apoptotic Programmed Cell Death --- p.29 / Chapter 2.3.3 --- Anti-tumor and Preventive Effect of SAHA in Animal Model --- p.29 / Chapter 2.3.4 --- Clinical Trials of SAHA --- p.30 / Chapter 2.4 --- FK228 (Depsipeptide or FR901228) --- p.31 / Chapter 2.4.1 --- Anti-malignancy mechanism of FK228 --- p.31 / Chapter 2.4.2 --- Anti-angiogenesis --- p.32 / Chapter 2.4.3 --- Drug Resistance and FK228 --- p.33 / Chapter 2.4.4 --- Studies of FK228 on Animal Models --- p.33 / Chapter 2.4.5 --- Clinical Trials --- p.34 / Chapter 2.5 --- Histone Modification and Nasopharyngeal Carcinoma --- p.34 / Chapter Chapter 3 --- Materials and Methods --- p.36 / Chapter 3.1 --- Cell Lines --- p.36 / Chapter 3.2 --- EBER ish Hybridization (EBER ISH) --- p.37 / Chapter 3.3 --- HDAC Inhibitors --- p.38 / Chapter 3.4 --- Cellular Sensitivity of NPC Cell Lines to HDAC Inhibitors --- p.38 / Chapter 3.4.1 --- Drug Treatment --- p.38 / Chapter 3.4.2 --- Determining Relative Amount of Survival Cells (WST-1 Assay) --- p.39 / Chapter 3.5 --- Flow Cytometry Analysis --- p.40 / Chapter 3.5.1 --- Collecting Cells and Fixation --- p.40 / Chapter 3.5.2 --- Staining --- p.41 / Chapter 3.5.3 --- Flow Cytometry Analysis --- p.41 / Chapter 3.6 --- Protein Extraction --- p.41 / Chapter 3.6.1 --- Harvesting Samples --- p.41 / Chapter 3.6.2 --- Protein Extraction --- p.42 / Chapter 3.6.3 --- Protein Quantification --- p.42 / Chapter 3.7 --- Western Blotting --- p.43 / Chapter 3.7.1 --- SDS-Polyarcylamide Gel Electrophoresis (PAGE) (SDS-PAGE) --- p.43 / Chapter 3.7.2 --- Wet Transfer of Proteins --- p.43 / Chapter 3.7.3 --- Immunoblotting --- p.44 / Chapter 3.7.4 --- Signal Detection --- p.44 / Chapter 3.8 --- CodeLin´kёØ Oligonucleotide Microarray --- p.45 / Chapter 3.8.1 --- HDAC Inhibitor Treatment --- p.45 / Chapter 3.8.2 --- RNA Extraction --- p.45 / Chapter 3.8.3 --- Quality and Quantity Assessment of Total RNA Extracted --- p.46 / Chapter 3.8.4 --- CodeLinkIM Expression Bioarray System --- p.46 / Chapter 3.8.5 --- Data Analysis --- p.48 / Chapter 3.9 --- Real-time Reverse Transcription PCR (Real-time RT-PCR) --- p.48 / Chapter Chapter 4 --- Results --- p.50 / Chapter 4.1 --- Presence of EBV --- p.50 / Chapter 4.2 --- Anti-prolirative Effect of HDAC Inhibitors --- p.52 / Chapter 4.3 --- Histone Acetylation --- p.56 / Chapter 4.4 --- Induction of p21 Expression in NPC Cell Lines --- p.58 / Chapter 4.5 --- HDAC Inhibitors Induced Cell Cycle Arrest and Polyploidy Formation --- p.60 / Chapter 4.5.1 --- Trichostatin A Induced G2/M Arrest --- p.60 / Chapter 4.5.2 --- Suberoylanilide Hydroxamic Acid Induced G1 Arrest --- p.62 / Chapter 4.5.3 --- FK228 Mediated G2/M Arrest --- p.64 / Chapter 4.6 --- HDAC Inhibitors Altered the Expression of Cell Cycle Regulatory Proteins --- p.66 / Chapter 4.6.1 --- TSA Down-regulated Cyclin A and B --- p.66 / Chapter 4.6.2 --- Suppressed Expression of Cyclin D1 and B by SAHA --- p.69 / Chapter 4.6.3 --- Effect of FK228 on Expression of Different Cyclins in NPC Cell Lines --- p.71 / Chapter 4.7 --- Effect of HDAC Inhibitors on EBV Proteins --- p.73 / Chapter 4.8 --- HDAC Inhibitors Modulated Gene Expression Profile --- p.76 / Chapter 4.8.1 --- SAHA and FK228-Induced Gene Expression Profile --- p.76 / Chapter 4.8.2 --- Validation of Expression Profile of Selected Genes by Real-time RT-PCR --- p.83 / Chapter Chapter 5 --- Discussion --- p.87 / Chapter 5.1 --- Anti-proliferative Effect of SAHA and FK228 on NPC Cell Lines --- p.88 / Chapter 5.2 --- Resistance of SAHA or FK228 in NPC --- p.93 / Chapter 5.3 --- Growth Inhibitory Mechanism of SAHA and FK228 in NPC Cells --- p.94 / Chapter 5.4 --- Induction of Polyploidy Cells in NPC Cell Lines --- p.98 / Chapter 5.5 --- Does EBV play a Role in HDAC Inhibiotrs Induced Growth Arrest in NPC Cell Lines? --- p.99 / Chapter 5.6 --- Transcriptional Signature of SAHA and FK228 in NPC Cell Lines --- p.100 / Chapter 5.7 --- Combining SAHA or FK228 with other Anti-tumor Agents --- p.104 / Chapter 5.8 --- Future Prospectus --- p.105 / Chapter Chapter 6 --- Summary --- p.106 / References --- p.108 / Appendix 1 --- p.120 / Appendix 2 --- p.121
22

Anti-cancer effects of the products of Ganoderma lucidum, G. tsugae and their artificial hybrid on breast cancer cells.

January 2005 (has links)
Luk Wing Yan Vivien. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2005. / Includes bibliographical references (leaves 207-239). / Abstracts in English and Chinese. / Acknowledgment --- p.i / Abstract --- p.iii / 摘要 --- p.vi / Contents --- p.viii / List of Figures --- p.xiv / List of Table --- p.xxv / Abbreviations --- p.xxv / Chapter Chapter 1 --- Introduction --- p.1 / Chapter 1.1 --- Ganoderma spp --- p.1 / Chapter 1.2 --- Bioactive components of Ganoderma spp --- p.3 / Chapter 1.2.1 --- Lingzhi polysaccharide --- p.3 / Chapter 1.2.2 --- Terpenes --- p.4 / Chapter 1.3 --- Ganoderma spp. as Chinese traditional medicine --- p.5 / Chapter 1.4 --- Artificial hybridisation of Ganoderma luciudm and G. tsugae --- p.6 / Chapter 1.4.1 --- Protoplast isolation and fusion of Ganoderma tsugae and G. lucidum --- p.8 / Chapter 1.5 --- Breast Cancer --- p.8 / Chapter 1.5.1 --- Anti-tumor effects of natural substances against breast cancer cell MCF-7 --- p.9 / Chapter 1.5.2 --- Anti-tumor effects of natural substances against breast cancer cell MDA-MB-231 --- p.11 / Chapter 1.5.3 --- Anti-proliferation of cancer --- p.12 / Chapter 1.5.3.1 --- Cell cycle arrest --- p.12 / Chapter 1.5.3.2 --- Cell death --- p.13 / Chapter 1.5.4 --- Anti-proliferation assays --- p.17 / Chapter 1.5.4.1 --- MTT assay --- p.17 / Chapter 1.5.4.2 --- Trypan blue cell viability assay --- p.18 / Chapter 1.5.4.3 --- BrdU assay --- p.18 / Chapter 1.6 --- Endocrine system and hormones --- p.19 / Chapter 1.6.1 --- Estrogen --- p.23 / Chapter 1.6.2 --- Estrogen receptors --- p.24 / Chapter 1.6.3 --- Estrogen action --- p.29 / Chapter 1.6.4 --- Estrogenicity assays --- p.32 / Chapter 1.6.4.1 --- Recombinant yeast assay --- p.33 / Chapter 1.6.4.2 --- E-screen assay --- p.35 / Chapter 1.6.4.3 --- Estrogen receptor competitor binding assay --- p.36 / Chapter 1.6.4.4 --- Endogenous estrogen-regulated gene expression assay --- p.39 / Chapter 1.6.4.4.1 --- Transforming growth factor --- p.39 / Chapter 1.6.4.4.2 --- Monoamine oxidase A --- p.40 / Chapter 1.6.4.4.3 --- pS2 --- p.40 / Chapter 1.6.4.5 --- Uterotrophic assay --- p.41 / Chapter 1.6.4.6 --- Comparison of in vitro and in vivo assay --- p.42 / Chapter 1.7 --- Aim of study --- p.45 / Chapter 1.7.1 --- Objectives --- p.45 / Chapter Chapter 2 --- Materials and Methods --- p.47 / Chapter 2.1 --- Fungal culture --- p.47 / Chapter 2.2 --- Artificial hybridization of Ganoderma tsugae and G. lucidum --- p.47 / Chapter 2.2.1 --- Protoplast isolation of Ganoderma tsugae and G. lucidum --- p.47 / Chapter 2.2.2 --- Protoplast fusion of Ganoderma tsugae and G. lucidum --- p.48 / Chapter 2.3 --- Screening and selection of hybrid ´Ø --- p.49 / Chapter 2.3.1 --- Temperature screening --- p.49 / Chapter 2.3.2 --- DNA fingerprint by Arbitarily-primed polymerase chain reaction --- p.49 / Chapter 2.3.2.1 --- Extraction of genomic DNA --- p.49 / Chapter 2.3.2.2 --- Arbitrarily-primed polymerase chain reaction --- p.50 / Chapter 2.3.2.3 --- Gel electrophoresis --- p.51 / Chapter 2.4 --- Confirmation test --- p.51 / Chapter 2.4.1 --- Somatic incompatibility test --- p.51 / Chapter 2.4.2 --- DNA fingerprinting by specific polymerase chain reaction --- p.52 / Chapter 2.4.2.1 --- Specific Polymerase Chain Reaction (PCR) --- p.52 / Chapter 2.4.2.2 --- Purification of PCR products --- p.52 / Chapter 2.4.2.3 --- Cycle-sequencing --- p.53 / Chapter 2.3.2.4 --- Sequencing --- p.54 / Chapter 2.3.2.5 --- Sequence analysis --- p.54 / Chapter 2.5 --- Characterization of the selected hybrid --- p.56 / Chapter 2.5.1 --- Scanning electron microscopy (SEM) --- p.56 / Chapter 2.5.1.1 --- Preparation of specimens for scanning electron microscopy --- p.56 / Chapter 2.5.1.2 --- "Cytological studies of pileus, stipe and spores of G. lucidum, G. tsugae and hybrid" --- p.57 / Chapter 2.5.2 --- Temperature effect --- p.57 / Chapter 2.5.3 --- Submerged fermentation --- p.57 / Chapter 2.5.4 --- Fruiting test --- p.58 / Chapter 2.6 --- "Bioactive components of G. lucidum, G. tsugae and hybrid" --- p.58 / Chapter 2.6.1 --- Sample preparation --- p.58 / Chapter 2 6.2 --- Lingzhi polysaccharide --- p.59 / Chapter 2.6.3 --- Terpenes --- p.59 / Chapter 2.7 --- Effect of extracts against breast cancer cell lines --- p.60 / Chapter 2.7.1 --- Cell culture --- p.60 / Chapter 2.7.2 --- Lingzhi Extract preparation --- p.61 / Chapter 2.7.3 --- Optimization of cell density --- p.61 / Chapter 2.7.3.1 --- MTT assay --- p.61 / Chapter 2 7.3.2 --- Trypan blue cell viability assay --- p.62 / Chapter 2.7.3.3 --- BrdU assay --- p.62 / Chapter 2.7.3.4 --- Growth curve of MCF-7 --- p.63 / Chapter 2.7.3.5 --- Growth curve of MDA-MB-231 --- p.64 / Chapter 2.7.4 --- Anti-proliferative effect of extracts on MCF-7 cells --- p.69 / Chapter 2.7.4.1 --- MTT assay --- p.69 / Chapter 2 7.4.2 --- Trypan blue cell viability assay --- p.69 / Chapter 2.7.4.3 --- BrdU assay --- p.70 / Chapter 2.7.5 --- Study of cultured medium effect of biomass and pileus extracts on MCF-7 cells --- p.71 / Chapter 2.7.5.1 --- Cultured medium effect ofbiomass and pileus extracts --- p.71 / Chapter 2.7.6 --- mRNA expression assay (RT-PCR) --- p.71 / Chapter 2.7.6.1 --- Effect of extract on gene expression --- p.71 / Chapter 2.7.6.2 --- Time effect of extract on gene expression --- p.72 / Chapter 2.7.6.3 --- Isolation of RNA --- p.72 / Chapter 2.7.6.4 --- Quantification and qualification of DNA and RNA by spectrophotometry --- p.73 / Chapter 2.7.6.5 --- First strand cDNA synthesis --- p.73 / Chapter 2.7.6.6 --- Amplification of cDNA --- p.74 / Chapter 2.7.7 --- Effect of biomass and pileus lingzhi polysacchandes and terpenes on MCF-7 cells --- p.75 / Chapter 2.7.7.1 --- Effect of reconstitution of lingzhi polysacchande and terpenes on MCF-7 cells --- p.75 / Chapter 2.7.8 --- Effect of biomass and pileus extracts on MDA-MB-231 cells --- p.76 / Chapter 2.8 --- Estrogenicigy assay --- p.76 / Chapter 2 8.1 --- E-screen test --- p.76 / Chapter 2.8.2 --- Estrogen receptor competitor binding assay --- p.77 / Chapter 2.8.3 --- pS2 mRNA expression assay --- p.78 / Chapter 2.9 --- DNA microarray analysis --- p.79 / Chapter 2.9.1 --- mRNA purification --- p.79 / Chapter 2.9.2 --- RT and LPR (Linear Polymerase Reaction) labeling --- p.80 / Chapter 2 9.3 --- pre-hybridization --- p.81 / Chapter 2.9.4 --- Hybridization --- p.82 / Chapter 2.9.5 --- Detection --- p.82 / Chapter 2.9.6 --- Image acquisition and analysis --- p.83 / Chapter Chapter 3 --- Result --- p.84 / Chapter 3.1 --- Artificial hybndization of Ganoderma tsugae and G. lucidum --- p.84 / Chapter 3.1.1 --- protoplast isomation and fusion of Ganoderma tsugae and G. lucidum --- p.84 / Chapter 3.2 --- Screening and selection of hybrid --- p.84 / Chapter 3.2.1 --- Temperature screening --- p.84 / Chapter 3.2.2 --- DNA fingerprint by Arbitrarily-primed polymerase chain reaction --- p.86 / Chapter 3.3 --- Confirmation tests --- p.88 / Chapter 3.3.1 --- Somatic incompatibility test --- p.88 / Chapter 3.3.2 --- DNA fingerprinting by specific polymerase chain reaction --- p.90 / Chapter 3.4 --- Characterization of selected hybrid --- p.100 / Chapter 3.4.1 --- Scanning electron micscropy --- p.100 / Chapter 3.4.2 --- Temperature effect --- p.103 / Chapter 3.4.3 --- Submerged fermentation --- p.105 / Chapter 3.4.4 --- Fruiting test --- p.107 / Chapter 3.5 --- "Bioactive components of G. lucidum, G. tsugae and hybrid" --- p.109 / Chapter 3.5.1 --- Lingzhi polysaccharide --- p.109 / Chapter 3.5.2 --- Terpenes --- p.109 / Chapter 3.6 --- Effect of extracts against breast cancer cell lines --- p.119 / Chapter 3.6.1 --- Anti-proliferative effect of extracts on MCF-7 cells --- p.119 / Chapter 3.6.2 --- Study of medium effect of biomass and pileus extracts on MCF-7 cells --- p.139 / Chapter 3.6.3 --- mRNA expression assay (RT-PCR) --- p.143 / Chapter 3.6.4 --- Effect of biomass and pileus lingzhi polysaccharides and terpenes on MCF-7 cells --- p.150 / Chapter 3.6.5 --- Effect of biomass and pileus extracts on MDA-MB231- cells --- p.159 / Chapter 3.7 --- Estrogenicity assay --- p.166 / Chapter 3.7.1 --- E-screen assay on biomass and pileus extracts --- p.166 / Chapter 3.7.2 --- E-screen assay on biomass and pileus terpenes and lingzhi polysaccharide --- p.166 / Chapter 3.7.3 --- Estrogen receptor competitor binding assay --- p.169 / Chapter 3.7.4 --- pS2 mRNA expression assay --- p.175 / Chapter 3.8 --- DNA microarray analysis --- p.177 / Chapter Chapter 4 --- Discussion --- p.184 / Chapter 4.1 --- Artificial hybridization of Ganoderma tsugae and G. lucidum --- p.184 / Chapter 4.1.1 --- Protoplast isolation and fusion of Ganoderma tsugae and G. luciudm --- p.184 / Chapter 4.1.2 --- Screening and selection of hybrid --- p.184 / Chapter 4.1.3 --- Characterization of the selected hybrid --- p.185 / Chapter 4.1.4 --- "Nature of hybrid, mutant and variant" --- p.189 / Chapter 4.2 --- Effect of extracts against breast cancer cell lines --- p.190 / Chapter 4.2.1 --- Anti-proliferative effect of extracts on MCF-7 cells --- p.190 / Chapter 4.2.2 --- Study of effect of cultured medium of biomass and pileus extracts on MCF-7 cells --- p.193 / Chapter 4.2.3 --- Effect of biomass and pileus extracts on MDA-MB231- cells --- p.194 / Chapter 4.2.4 --- mRNA expression assay (RT-PCR) --- p.195 / Chapter 4.3 --- Estrogenicity --- p.198 / Chapter 4.3.1 --- E-screen assay --- p.198 / Chapter 4.3.2 --- Estrogen receptor competitor binding assay --- p.199 / Chapter 4.3.3 --- pS2 mRNA expression assay --- p.200 / Chapter 4.3.4 --- Ganoderma spp. As hormonal therapy --- p.201 / Chapter 4.4 --- DNA microarray analysis --- p.201 / Chapter 4.5 --- Further investigation --- p.204 / Chapter Chapter 5 --- Conclusion --- p.205 / Chapter Chapter 6 --- Reference --- p.207
23

Growth inhibitory effects of chlorophyllin on human breast carcinoma MCF-7 cells.

January 2005 (has links)
Kong Ka-lai. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2005. / Includes bibliographical references (leaves 126-149). / Abstracts in English and Chinese. / Acknowledgements --- p.i / Abstract --- p.ii / Abstract (Chinese Version) --- p.vi / Table of Contents --- p.ix / List of Figures/Table --- p.xiii / List of Abbreviations --- p.xvi / Chapter Chapter 1 --- General Introduction / Chapter 1.1 --- An Overview on Cancer --- p.1 / Chapter 1.2 --- Biological Effects of Chlorophyllin --- p.7 / Chapter 1.2.1 --- CHL as Photosensitizer --- p.7 / Chapter 1.2.2 --- CHL as Antioxidant --- p.8 / Chapter 1.2.3 --- CHL as Anticarcinogenic Agent --- p.9 / Chapter 1.3 --- Regulation of Cell Cycle --- p.13 / Chapter 1.3.1 --- Cell-Cycle Checkpoints --- p.13 / Chapter 1.3.2 --- Cell-Cycle Regulatory Proteins --- p.15 / Chapter 1.4 --- Regulation of Mitogen-Activated Protein Kinase (MAPK) Signaling Cascade --- p.21 / Chapter 1.5 --- Programmed Cell Death (or Apoptosis) --- p.27 / Chapter 1.5.1 --- Regulation of Caspase-Dependent Apoptosis --- p.28 / Chapter 1.5.2 --- Regulation of Caspase-Independent Cell Death --- p.32 / Chapter 1.5.3 --- Bcl-2 Family Proteins in Modulation of Cell Death --- p.32 / Chapter 1.6 --- In Vivo Antitumor Screening System --- p.37 / Chapter 1.7 --- Aims of the Present Study --- p.38 / Chapter Chapter 2 --- In Vitro Studies of the Anticancer Effect of Chlorophyllin / Chapter 2.1 --- Introduction --- p.39 / Chapter 2.1.1 --- DNA-Flow Cytometric Analysis --- p.51 / Chapter 2.1.2 --- Western Blot Analysis --- p.54 / Chapter 2.2 --- Materials and Methods --- p.56 / Chapter 2.2.1 --- Maintenance of Cell Lines --- p.56 / Chapter 2.2.2 --- Cytotoxic and Cytostatic Effects on the Cancer Cells --- p.56 / Chapter 2.2.3 --- DNA-Flow Cytometric Analysis --- p.60 / Chapter 2.2.4 --- Western Blot Analysis --- p.61 / Chapter 2.2.5 --- JC-1 Mitochondrial Potential Sensor --- p.64 / Chapter 2.2.6 --- Caspase Inhibitors --- p.65 / Chapter 2.2.7 --- Statistical Analysis --- p.66 / Chapter 2.2.8 --- Densitometric Analysis --- p.66 / Chapter 2.3 --- Results --- p.67 / Chapter 2.3.1 --- Effects of CHL on the Growth of Human Cancer Cells by MTT Assay --- p.67 / Chapter 2.3.2 --- Effect of CHL on the Proliferation of MCF-7 Cells by Chemi-BrdU Incorporation --- p.69 / Chapter 2.3.3 --- Effect of CHL on Cell Cycle of MCF-7 Cells --- p.71 / Chapter 2.3.4 --- Effect of CHL on the Cyclin D1 Expression in MCF-7 Cells --- p.74 / Chapter 2.3.5 --- Effects of CHL on JNK and c-Jun Expressions and Their Phosphorylations in MCF-7 Cells --- p.76 / Chapter 2.3.6 --- Effect of CHL on DNA fragmentation in MCF-7 Cells --- p.78 / Chapter 2.3.7 --- Effect of CHL on Mitochondrial Membrane Potential of MCF-7 Cells --- p.80 / Chapter 2.3.8 --- Effects of CHL on the PARP Expression and Cleavage in MCF-7 Cells --- p.83 / Chapter 2.3.9 --- "Effects of CHL on Bcl-2, Bcl-xL and Bad Expressions in MCF-7 Cells" --- p.85 / Chapter 2.3.10 --- Effects of CHL on Caspase Activations in MCF-7 Cells --- p.88 / Chapter 2.3.11 --- Effects of Caspase Inhibitors on the CHL-Induced Apoptosis in MCF-7 Cells --- p.90 / Chapter 2.4 --- Discussion --- p.93 / Chapter Chapter 3 --- In Vivo Studies of the Anticancer Effect of Chlorophyllin / Chapter 3.1 --- Introduction --- p.104 / Chapter 3.2 --- Materials and Methods --- p.106 / Chapter 3.2.1 --- Transplantation of MCF-7 Cells into the Nude Mice and Treatment --- p.106 / Chapter 3.2.2 --- Western Blot Analysis --- p.107 / Chapter 3.2.3 --- Statistical Analysis --- p.107 / Chapter 3.3 --- Results --- p.108 / Chapter 3.3.1 --- In Vivo Antitumor Activity of CHL --- p.108 / Chapter 3.3.2 --- In Vivo Effects of CHL on Cyclin D1 and Bcl-2 Expressions in MCF-7 Solid Tumor --- p.111 / Chapter 3.4 --- Discussion --- p.113 / Chapter Chapter 4 --- General Discussion --- p.115 / References --- p.126
24

Antiangiogenic agents from tripterygium wilfordii for cancer treatment. / 雷公藤中的抗血管新生劑 / CUHK electronic theses & dissertations collection / Lei gong teng zhong de kang xue guan xin sheng ji

January 2009 (has links)
Five traditional Chinese medicines were screened for their antiangiogenic activities through zebrafish angiogenic assay. Two of them, Tripterygium wilfordii and Rheum palmatum showed potential in the primary screening. T. wilfordii was selected in further study. / In the further investigation of antiangiogenic activity of triptolide on mammal systems, triptolide showed potent activity in human umbilical vein endothelial cells (HUVECs) assays including proliferation, migration and tube formation assay. It inhibited HUVEC proliferation with IC50 as low as 34 nM. It also showed more potency in HUVEC migration and tube formation assay at as low concentration as nanomolar level than SU5416, a putative VEGFR-2 inhibitor currently in clinic trials. RT-PCR and western blotting analysis showed that the underlying mechanism of triptolide correlated with down-regulation of VEGFR-2 and Tie2 expression and production. Tie2 inhibition appeared to be a later event as compared with VEGFR-2. Tie2 overexpression in HUVEC could attenuate the inhibitory effect of triptolide on HUVEC proliferation. Tie2 knockdown mimicked the inhibition activity of triptolide in tube formation assay. These phenomemon revealed that Tie2 signaling pathway plays a crucial role in triptolide-mediated angiogenesis inhibition. In in vivo Matrigel Plug assay, triptolide showed inhibition effect at as low as 100 nM. / T. wilfordii is an immune-suppressive, anti-inflammatory herb used in China for centuries. Through bioassay-guided purification, three antiangiogenic terpenoids were isolated from the ethyl acetate fraction, namely, celastrol, cangoronine and triptolide. Among them, triptolide manifested the most potent antiangiogenic activities against vessel formation. As low as 0.31microM, triptolide inhibited 20% of vessel formation, and the inhibition reached a plateau of 50% at 1.2 microM. Celatrol reduced vessel formation by more than 30% at 0.62microM, but killed 50% of the embryos at higher concentrations. Cangoronine was much weaker, inhibiting vessel formation by 20% at 2.5microM. These three components all showed stronger antiangiogenic activities than 2-methoxyestradiol, a putative compound currently under clinical trials as an antiangiogenic agent for cancer treatment, as the latter inhibited angiogenesis in zebrafish embryos by 34% at 10microM. The loss of vessel formation in the embryos treated with triptolide was further confirmed using endogenous alkaline phosphatase staining. Semi-quantitative RT-PCR analysis revealed that triptolide dose- and time-dependently reduced the mRNA expression of angiopoietin (angpt2) and tie2 in zebrafish, indicating the involvement of angpt2/tie2 signaling pathway in the antiangiogenic action of triptolide. / This research revealed that zebrafish model is a promising antiangiogenic model for both the screening of antiangiogenic agents from Chinese herbal medicine and the subsequent discovery for the drug targets. Triptolide, an anti-inflammatory component from T. wilfordii, is a potent angiogenic inhibitor through targeting VEGFR-2 and Tie2 pathways in mammal models whereas targeting ang2-tie2 pathway in zebrafish model. The anti-tumor action of triptolide was demonstrated to be partly through inhibition of tumor angiogenesis. Moreover, the potent antiangiogenic action exerted by triptolide at nanomolar dosage level gives proof that it is a promising lead compound for the development of antiangiogenic drug for cancer treatment. (Abstract shortened by UMI.) / He, Mingfang. / Adviser: Paul Pui-Hey Bot. / Source: Dissertation Abstracts International, Volume: 71-01, Section: B, page: 0247. / Thesis (Ph.D.)--Chinese University of Hong Kong, 2009. / Includes bibliographical references (leaves 84-106). / Electronic reproduction. Hong Kong : Chinese University of Hong Kong, [2012] System requirements: Adobe Acrobat Reader. Available via World Wide Web. / Electronic reproduction. Ann Arbor, MI : ProQuest Information and Learning Company, [200-] System requirements: Adobe Acrobat Reader. Available via World Wide Web. / Abstracts in English and Chinese.
25

Investigating the chemopreventive effect of hesperetin, luteolin and cyclooxygenase inhibitors in a mouse model of breast cancer.

January 2012 (has links)
乳腺癌是女性最常見的腫瘤之一,多發生在女性絶經後,並具有雌激素依賴性。芳香化酶(CYP19)是雌激素生物合成過程中的關鍵酶,而芳香化酶抑製劑(AI)則被用於替代治療雌激素依賴性的乳腺癌。然而,AI在降低雌激素水平的同時能夠引起骨質酥鬆。此項研究的目的是找尋AI替代物。 / 黃酮類化合物是一種多酚化合物,廣泛分佈于植物中。我們先前的研究發現二氢黄酮陈皮素能夠抑制芳香化酶的生物活性,并且抑制芳香化酶高表達的乳腺癌生長。在本研究中,我們發現陳皮素在抑制腫瘤生長的同時能夠降低来曲唑引起的骨質流失。木犀草素是另外一種黄酮类化合物,它同樣能夠抑制芳香化酶的活性并減少骨流失。而與陳皮素不同的是,它能夠抑制芳香化酶的表達。在芳香化酶高表達的乳腺癌細胞(MCF-7 aro)中,木犀草素抑制芳香化酶活性的IC50是3 μM。在MCF-7 細胞中,5 μM的木犀草素能夠抑制CYP19 mRNA 的表達,螢光素酶報告實驗顯示木犀草素是通過作用于啟動子I.3和II來抑制CYP19的表達。蛋白印跡實驗表明木犀草素抑制CYP19表達的分子機制可能通過調節JNK信號通路進而減少AP-1的活性來實現。動物實驗結果顯示木犀草素能夠抑制MCF-7aro腫瘤的生長并改善來曲唑引起的骨流失。 / 環氧化酶(COX)是花生四烯酸轉化為前列腺素途徑中的一種關鍵酶。研究發現COX-2在乳腺癌組織中廣泛表達。本實驗研究了COX抑製劑在裸鼠動物模型中對乳腺癌腫瘤的作用機制。研究結果表明塞來昔布和阿司匹林在不影響血液中雌激素水平的情況下抑制乳腺癌腫瘤的生長。蛋白印迹實驗顯示這兩種藥物能夠降低腫瘤中COX-2,Cyclin A和Bcl-xL的表達。miR-98, miR-222和miR-145也能夠被塞來昔布和阿司匹林影響。 / 本研究表明陳皮素,木犀草素及COX抑制劑有潛力成為替代AI的化學治療藥物或共同治療藥物。 / Breast cancer is one of the most prevalent cancers affecting women. The majority of breast tumor growth occurred in the post-menopausal period are estrogen dependent. Aromatase (CYP19) catalyzes the rate-limiting step in the synthetic reaction of estrogen and aromatase inhibitors (AIs) are contemporary treatment for estrogen-positive breast cancer. However, estrogen-lowering drugs may promote osteoporosis. Our objective of this study further identified some alternatives for AIs. / Flavonoids are polyphenolic compounds that are ubiquitously distributed in plants. We have previously found that the flavanone hesperetin can inhibit the activity of aromatase and suppress aromatase-expressing breast tumor growth. In this project, we investigated the potential interaction between hesperetin and the AI letrozole in a mouse model. Our results showed that hesperetin could inhibit the tumor growth and reduce bone loss induced by letrozole. Similarly, another flavonoid luteolin also inhibited aromatase and prevented bone deterioration as observed in this project. In cells stably transfected with CYP19 (MCF-7aro), luteolin inhibited the aromatase activity with an IC50 value of 3μM. In addition, 5μM luteolin significantly reduced CYP19 mRNA expression in MCF-7 cells. Luciferase reporter assay revealed that luteolin could suppress CYP19 transcription at promoter regions I.3 and II. Western analysis illustrated that JNK signaling pathway was involved and deactivation of AP-1 could be the underlying molecular mechanism. Subsequently, we examined the effect in vivo. Our results showed that luteolin could inhibit the MCF-7aro tumor growth and improved bone loss induced by letrozole. / Cyclooxygenase (COX) is an enzyme responsible for the conversion of arachidonic acid into prostaglandins. It is over-expressed in breast cancer tissue and an increased expression of COX-2 was also observed in the xenograft model employed in this project. In the last study we evaluated the importance of COX-2 in breast tumor growth in this model. Our data showed that celecoxib and aspirin could significantly suppress the tumor growth without changing the plasma estrogen level. Western analysis illustrated that COX-2, Cyclin A, Bcl-xL and ER were reduced in celecoxib- and aspirin- treated tumor samples and miR-98, miR-222 and miR-145 were altered by celecoxib or aspirin. / After all, this project demonstrated that hesperetin, luteolin and COX-inhibitors could be potential chemopreventive or co-therapeutic agents. / Detailed summary in vernacular field only. / Detailed summary in vernacular field only. / Detailed summary in vernacular field only. / Detailed summary in vernacular field only. / Li, Fengjuan. / Thesis (Ph.D.)--Chinese University of Hong Kong, 2012. / Includes bibliographical references (leaves 131-148). / Abstract also in Chinese. / ACKNOWLEDGEMENTS --- p.I / ABSTRACT --- p.II / 摘要 --- p.IV / LIST OF ABBREVIATIONS --- p.V / TABLE OF CONTENTS --- p.VII / CHAPTER 1 --- p.1 / GENERAL INTRODUCTION --- p.1 / Chapter 1.1 --- Types of Breast Cancer --- p.3 / Chapter 1.2 --- Nuclear Receptor Signaling Pathways in Breast Cancer --- p.5 / Chapter 1.3 --- Estrogen and Breast Cancer --- p.7 / Chapter 1.4 --- Estrogen and Bone Health --- p.8 / Chapter 1.5 --- Estrogen Biosynthesis and Aromatase --- p.10 / Chapter 1.6 --- Tissue Specific Promoter for Aromatase Expression --- p.13 / Chapter 1.7 --- Nuclear Receptors and Aromatase Promoter Regulation --- p.15 / Chapter 1.8 --- Signaling Pathway and Aromatase Expression --- p.17 / Chapter 1.9 --- Cell Cycle in Breast Cancer --- p.20 / Chapter 1.10 --- Cell Apoptosis --- p.23 / Chapter 1.11 --- Treatment of breast cancer --- p.25 / Chapter 1.12 --- Phytoestrogens --- p.29 / Chapter 1.13 --- Aim of My Study --- p.32 / CHAPTER 2 --- p.33 / MATERIALS AND METHODS --- p.33 / Chapter 2.1 --- Chemicals and Materials --- p.33 / Chapter 2.1.1 --- Chemicals --- p.33 / Chapter 2.1.2 --- Plasmids --- p.33 / Chapter 2.2 --- Cell Culture --- p.33 / Chapter 2.3 --- Aromatase Activity Assay --- p.34 / Chapter 2.4 --- Quantitative Real Time PCR --- p.36 / Chapter 2.4.1 --- RNA Isolation and cDNA Synthesis --- p.36 / Chapter 2.4.2 --- Quantitative Real Time PCR Assay --- p.37 / Chapter 2.4.3 --- MiRNA Quantitative Real Time PCR Assay --- p.38 / Chapter 2.5 --- Western Blot --- p.39 / Chapter 2.6 --- Measurement of Promoter Activity --- p.41 / Chapter 2.6.1 --- Plasmid Preparation --- p.41 / Chapter 2.6.2 --- Transient Transfection and Dual-Luciferase Assay --- p.42 / Chapter 2.7 --- Electrophoretic Mobility Shift Assay (EMSA) --- p.43 / Chapter 2.7.1 --- Nuclear protein extraction --- p.43 / Chapter 2.7.2 --- Electrophorectic Mobility Shift Assay --- p.44 / Chapter 2.8 --- Animal Experiment Design --- p.45 / Chapter 2.8.1 --- Animal Model for Hesperetin Study --- p.45 / Chapter 2.8.2 --- Animal Model for Luteolin Study --- p.46 / Chapter 2.8.3 --- Animal Model for Cycooxygenase Inhibitors Study --- p.48 / Chapter 2.8.4 --- Serum Estradiol Determination --- p.49 / Chapter 2.8.5 --- Analysis of serum lipoproteins --- p.49 / Chapter 2.8.6 --- Bone Image Acquisition and Region of Interest Selection --- p.50 / Chapter 2.9 --- Statistical Analysis --- p.50 / CHAPTER 3 --- p.51 / The citrus flavonone hesperetin prevents letrozole- induced bone loss in a mouse model of breast cancer --- p.51 / Chapter 3.1 --- Introduction --- p.51 / Chapter 3.2 --- Results --- p.54 / Chapter 3.2.1 --- Murine Body Weight and Liver Weight --- p.54 / Chapter 3.2.2 --- Effect of Hesperetin and Letrozole on Xenograft Growth in Ovariectomized Mice --- p.55 / Chapter 3.2.3 --- Hesperetin Reduced Plasma Estradiol Concentration --- p.58 / Chapter 3.2.4 --- PS2 mRNA Expression in Tumor --- p.59 / Chapter 3.2.5 --- Uterine Wet Weight --- p.60 / Chapter 3.2.6 --- Hesperetin Prevent Bone Deterioration Induced by Letrozole --- p.61 / Chapter 3.3 --- DISCUSSION --- p.63 / CHAPTER 4 --- p.66 / dIETARY FLAVONOID LUTEOLIN ON cyp19 transcription in the breast cancer cells mcf-7 --- p.66 / Chapter 4.1 --- Introduction --- p.66 / Chapter 4.2 --- Results --- p.68 / Chapter 4.2.1 --- Inhibitory Effect of Luteolin on Aromatase Activity --- p.68 / Chapter 4.2.2 --- Luteolin Reduced Aromatase mRNA Expression in MCF-7 Cells --- p.70 / Chapter 4.2.3 --- Effect of Luteolin on Promoter I.3/II Activity of CYP19 in MCF-7 Cells --- p.71 / Chapter 4.2.4 --- The Effect of Luteolin on Truncation CYP19 Gene Reporter Assay --- p.72 / Chapter 4.2.5 --- Luteolin Reduced AP-1 Binding in Promoter I.3/II DNA Fragment --- p.74 / Chapter 4.2.6 --- Inhibitory Effect of Luteolin on Protein Kinase Signaling --- p.76 / Chapter 4.3 --- Discussion --- p.78 / CHAPTER 5 --- p.83 / interaction OF LUTEOLIN and letrozole in a postmenopausal breast cancer model --- p.83 / Chapter 5.1 --- Introduction --- p.83 / Chapter 5.2 --- Results --- p.86 / Chapter 5.2.1 --- Luteolin and letrozole treatment had no effect on mouse body weight and liver weight --- p.86 / Chapter 5.2.2 --- Effect of luteolin and Letrozole on Xenograft Growth in Ovariectomized Mice --- p.88 / Chapter 5.2.3 --- Luteolin reduced plasma estradiol concentration --- p.91 / Chapter 5.2.4 --- Luteolin Counteracted Uterine Weight Reduction under Letrozole Treatment --- p.92 / Chapter 5.2.5 --- Luteolin Prevented Bone Deterioration Induced by Letrozole --- p.93 / Chapter 5.2.6 --- The Effect of Luteolin on Plasma TC and TG --- p.95 / Chapter 5.2.7 --- Luteolin Increased HDL Level and Reduced the Ratio of LDL/HDL --- p.97 / Chapter 5.2.8 --- Effect of Luteolin on Cell Cycle and Apoptotic Protein Expression --- p.99 / Chapter 5.3 --- DISCUSSION --- p.104 / CHAPTER 6 --- p.107 / cyclooxygenase inhibitors suppresse breast tumor growth in NUDE MICE --- p.107 / Chapter 6.1 --- Introduction --- p.107 / Chapter 6.2 --- Results --- p.109 / Chapter 6.2.1 --- Celecoxib and aspirin treatment had no effect on mouse body weight and liver weight --- p.109 / Chapter 6.2.2 --- Effect of celecoxib and aspirin on Xenograft Growth in Ovariectomized Mice --- p.111 / Chapter 6.2.3 --- Celecoxib and aspirin had no effect on plasma estradiol concentration --- p.113 / Chapter 6.2.4 --- Celecoxib and Aspirin Had no Effect on Uterine Weight --- p.114 / Chapter 6.2.5 --- Protein expression of COX-2, Cell cycle-related and cell Apoptotic Genes --- p.115 / Chapter 6.2.6 --- Detection of Related miRNA Expression Level in Tumors --- p.118 / Chapter 6.2.7 --- c-Myc mRNA Expression Level were Regulated in Tumors --- p.121 / Chapter 6.3 --- DISCUSSION --- p.124 / CHAPTER 7 --- p.127 / SUMMARY --- p.127 / REFERENCE --- p.131
26

Cyanidin protects HK-2 proximal tubular cells against cisplatin-induced apoptosis through modulating AKT and ERK pathways.

January 2010 (has links)
Gao, Si. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2010. / Includes bibliographical references (leaves 77-85). / Abstracts in English and Chinese. / Acknowledgement --- p.i / Abstract --- p.ii / Abstract (in Chinese) --- p.iv / List of Abbreviations --- p.v / List of Figures --- p.vii / Table of Contents --- p.ix / Chapter Chapter One: --- Introduction --- p.1 / Chapter 1.1. --- Cancer --- p.1 / Chapter 1.2. --- Chemotherapy --- p.2 / Chapter 1.3. --- Cisplatin --- p.3 / Chapter 1.4. --- Cisplatin-induced nephrotoxicity --- p.4 / Chapter 1.5. --- Mechanisms of cisplatin-induced nephrotoxicity --- p.5 / Chapter 1.5.1. --- Apoptosis in cisplatin-induced nephrotoxicity --- p.5 / Chapter 1.5.2. --- MAPK activation in cisplatin-induced nephrotoxicity --- p.7 / Chapter 1.5.3. --- Oxidative stress in cisplatin-induced nephrotoxicity --- p.8 / Chapter 1.6. --- Polyphenols --- p.10 / Chapter 1.7. --- Anthocyanins --- p.10 / Chapter 1.8. --- Rose --- p.11 / Chapter 1.9. --- Cyanidin --- p.12 / Chapter 1.10. --- Objectives of this project --- p.13 / Chapter Chapter Two: --- Materials and Methods --- p.15 / Chapter 2.1. --- Materials --- p.15 / Chapter 2.2. --- Cell culture --- p.15 / Chapter 2.3. --- Drug treatment --- p.16 / Chapter 2.4. --- MTT assay --- p.16 / Chapter 2.5. --- Lactate dehydrogenase (LDH) assay --- p.16 / Chapter 2.6. --- TUNEL assay and DAPI staining --- p.17 / Chapter 2.7. --- Flow cytometric analysis --- p.17 / Chapter 2.8. --- Determination of caspase-3 activity --- p.18 / Chapter 2.9. --- Measurement of ROS generation --- p.18 / Chapter 2.10. --- Evaluation of mitochondrial membrane potential --- p.19 / Chapter 2.11. --- Single Cell Gel Electrophoresis (Comet Assay) --- p.19 / Chapter 2.12. --- Western blot analysis --- p.20 / Chapter 2.13. --- Statistical analysis --- p.21 / Chapter Chapter Three: --- Results --- p.22 / Chapter 3.1. --- Cyanidin attenuates cisplatin-induced cytotoxicity in HK-2 cells --- p.22 / Chapter 3.1.1. --- Cytotoxicity induces by cisplatin in HK-2 cells --- p.22 / Chapter 3.1.2. --- Rose extract attenuates cisplatin-induced cytotoxicity and LDH leakage --- p.26 / Chapter 3.1.3. --- Cyanidin attenuates cisplatin-induced cytotoxicity and LDH leakage --- p.26 / Chapter 3.1.4. --- Cyanidin did not affect cisplatin-induced cytotoxicity in Hela cell --- p.30 / Chapter 3.2. --- Cyanidin rescues HK-2 cells from cisplatin-induced apoptosis --- p.31 / Chapter 3.2.1. --- Cisplatin induces cell apoptosis in HK-2 cells --- p.31 / Chapter 3.2.2. --- Rose extract rescues HK-2 cells from cisplatin-induced apoptosis --- p.31 / Chapter 3.2.3. --- Cyanidin rescues HK-2 cells from cisplatin-induced apoptosis --- p.32 / Chapter 3.3. --- Cyanidin suppresses cisplatin-induced activation of caspase and cleavage of PARP --- p.38 / Chapter 3.3.1. --- Cisplatin induces activation of caspase-3 --- p.38 / Chapter 3.3.2. --- Rose extract suppresses cisplatin-induced activation of caspase-3 --- p.38 / Chapter 3.3.3. --- Cyanidin suppresses cisplatin-induced activation of caspase-3 --- p.38 / Chapter 3.3.4. --- Rose extract suppresses cisplatin-induced caspase activation and PARP cleavage --- p.41 / Chapter 3.3.5. --- Cyanidin suppresses cisplatin-induced caspase activation and PARP cleavage --- p.43 / Chapter 3.4. --- Cyanidin rescues HK-2 cells from cisplatin-induced mitochondrial dysfuntion by regulating the expression of Bcl-2 family proteins --- p.43 / Chapter 3.4.1. --- Cyanidin prevents cisplatin-induced loss of mitochondrial membrane potential (A^m) --- p.43 / Chapter 3.4.2. --- Cyanidin regulates the expression of Bcl-2 family proteins to prevent cisplatin-induced mitochondrial dysfunction --- p.44 / Chapter 3.5. --- Cyanidin reduces cisplatin-induced apoptosis by suppressing the activation of p53 --- p.46 / Chapter 3.6. --- Cyanidin inhibits ROS-mediated DNA damage in HK-2 cells --- p.48 / Chapter 3.6.1. --- Cyanidin prevents cisplatin-induced DNA damage --- p.48 / Chapter 3.6.2. --- Cyanidin inhibits cisplatin-induced accumulation of ROS --- p.48 / Chapter 3.7. --- "Cyanidin suppresses cisplatin-induced apoptosis by activation of AKT, JNK and ERK" --- p.52 / Chapter 3.7.1. --- Cisplatin activates ERK and AKT pathways --- p.52 / Chapter 3.7.2. --- Cyanidin suppresses cisplatin-induced activation of MAPKs and AKT pathways --- p.52 / Chapter 3.7.3. --- AKT and ERK Inhibitors attenuates cisplatin-induced apoptosis in HK-2 cells --- p.53 / Chapter Chapter Four: --- Discussion --- p.60 / Chapter 4.1. --- Cell model and cisplatin treatment --- p.60 / Chapter 4.2. --- Cisplatin nephrotoxicity and its renoprevention --- p.61 / Chapter 4.3. --- Rose extract prevents cisplatin-induced apoptosis in HK-2 cells --- p.62 / Chapter 4.3.1. --- Rose extract prevents cisplatin-induced apoptosis in HK-2 cells --- p.63 / Chapter 4.3.2. --- Rose extract inhibits cisplatin-induced caspase activation and PARP cleavage --- p.64 / Chapter 4.4. --- Cyanidin prevents cisplatin-induced apoptosis in HK-2 cells --- p.66 / Chapter 4.4.1. --- Cyanidin will not affect cisplatin-induced cell death in HeLa cells --- p.66 / Chapter 4.4.2. --- Cyanidin prevents cisplatin-induced apoptosis by inhibiting caspase activation and PARP cleavage in HK-2 cells --- p.66 / Chapter 4.4.3. --- Cyanidin prevents the cisplatin-induced loss of mitochondrial membrane potential by regulating Bcl-2 proteins in HK-2 cells --- p.67 / Chapter 4.4.4. --- Cyanidin suppresses cisplatin-induced total and phosphorylated p53 activation --- p.68 / Chapter 4.4.5. --- Cyanidin prevents the cisplatin-induced overproduction of intracellular ROS and subsequent DNA damage in HK-2 cells --- p.69 / Chapter 4.4.6. --- Cyanidin suppresses the cisplatin-induced activation of MAPKs and AKT pathways in HK-2 cells --- p.71 / Chapter Chapter Five: --- Conclusion --- p.74 / References --- p.77
27

In vitro and in vivo study of effects of sinigrin on liver.

January 2006 (has links)
Meng Jie. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2006. / Includes bibliographical references. / Abstracts in English and Chinese. / Acknowledgements --- p.i / Abstract --- p.ii / 論文摘要 --- p.iv / Table of Contents --- p.vi / Abbreviation --- p.x / List of Figures --- p.xi / List of Tables --- p.xiii / Chapter Chapter 1: --- Introduction --- p.1 / Chapter 1.1 --- Black Mustard and Sinigrin --- p.2 / Chapter 1.2 --- Hepatocellular Carcinoma --- p.5 / Chapter 1.2.1 --- Different Stages of HCC --- p.6 / Chapter 1.2.2 --- Risk Factors --- p.8 / Chapter 1.2.3 --- Treatments of HCC --- p.10 / Chapter 1.3 --- Biomarkers Used to Evaluate Effects of Sinigrin on HCC --- p.12 / Chapter 1.3.1 --- AST & ALT --- p.12 / Chapter 1.3.2 --- Glutathione S Transferase -p (GST-p) --- p.13 / Chapter 1.4 --- Tumor Suppressor Genes and Oncogenes --- p.14 / Chapter 1.4.1 --- "p53, the Tumor Suppressor Gene" --- p.15 / Chapter 1.4.2 --- p53-dependent pathway --- p.15 / Chapter 1.4.2.1 --- Mdm2 --- p.16 / Chapter 1.4.2.2 --- Bax and Bcl-2 --- p.17 / Chapter 1.4.2.3 --- PCNA and p21wAF1/CIP1 --- p.18 / Chapter 1.5 --- Aim of the Project --- p.19 / Chapter Chapter 2: --- Materials and Methods --- p.20 / Chapter 2.1 --- In vitro Studies --- p.21 / Chapter 2.1.1 --- Neutral Red Assay --- p.21 / Chapter 2.1.1.1 --- Chemicals and Reagents --- p.21 / Chapter 2.1.1.2 --- Liver Cells --- p.23 / Chapter 2.1.1.3 --- Neutral Red Assay --- p.24 / Chapter 2.1.2 --- Flow Cytometery --- p.24 / Chapter 2.1.2.1 --- Chemicals and Reagents --- p.25 / Chapter 2.1.2.2 --- Flow Cytometery Analysis --- p.25 / Chapter 2.1.3 --- DNA Fragmentation --- p.26 / Chapter 2.1.3.1 --- Chemicals and Reagents --- p.26 / Chapter 2.1.3.2 --- DNA Extraction --- p.28 / Chapter 2.1.3.3 --- DNA Agarose Gel Electrophoresis --- p.29 / Chapter 2.1.4 --- cDNA Microarray --- p.29 / Chapter 2.1.4.1 --- Chemicals and Reagents --- p.30 / Chapter 2.1.4.2 --- RNA Extraction --- p.33 / Chapter 2.1.4.3 --- RNA Quantity and Quality Control --- p.34 / Chapter 2.1.4.4 --- RT-PCR --- p.35 / Chapter 2.1.4.5 --- cRNA Convention and Purification --- p.36 / Chapter 2.1.4.6 --- Hybridization --- p.37 / Chapter 2.1.4.7 --- Washing and Detection --- p.37 / Chapter 2.1.4.8 --- Data Analysis --- p.38 / Chapter 2.2 --- In vivo Studies --- p.39 / Chapter 2.2.1 --- Animal Treatment --- p.39 / Chapter 2.2.1.1 --- Chemicals and Reagents --- p.39 / Chapter 2.2.1.2 --- Chemical Carcinogens --- p.40 / Chapter 2.2.1.3 --- Promotion Stage --- p.41 / Chapter 2.2.1.4 --- Progression Stage --- p.44 / Chapter 2.2.2 --- Measurement of Serum ALT and AST Activities --- p.46 / Chapter 2.2.2.1 --- Chemicals and Reagents --- p.46 / Chapter 2.2.2.2 --- Activity Assay --- p.46 / Chapter 2.2.3 --- Histological Analysis --- p.47 / Chapter 2.2.3.1 --- Chemicals and Reagents --- p.47 / Chapter 2.2.3.2 --- Preparation of Slides --- p.49 / Chapter 2.2.3.3 --- H&E Staining --- p.49 / Chapter 2.2.3.4 --- GST-p Immuno-staining --- p.50 / Chapter 2.2.4 --- Semi-Quantitative RT-PCR Analysis of mRNA Expression --- p.53 / Chapter 2.2.4.1 --- Chemicals and Reagents --- p.53 / Chapter 2.2.4.2 --- Extraction of total RNA from rat liver --- p.53 / Chapter 2.2.4.3 --- Quantity and Quality Control of RNA --- p.53 / Chapter 2.2.4.4 --- RT-PCR (Reverse Transcription) --- p.54 / Chapter 2.2.4.5 --- PCR --- p.54 / Chapter 2.2.4.6 --- DNA gel electrophoresis --- p.55 / Chapter 2.2.4.7 --- Data Analysis --- p.56 / Chapter 2.2.5 --- Western Blot Analysis for Biomarkers --- p.56 / Chapter 2.2.5.1 --- Chemicals and Reagents --- p.56 / Chapter 2.2.5.2 --- Extraction of the Cytosol Protein --- p.60 / Chapter 2.2.5.3 --- Extraction of the Nuclear protein --- p.61 / Chapter 2.2.5.4 --- SDS Gel Electrophoresis --- p.61 / Chapter 2.2.5.5 --- Western Blot --- p.62 / Chapter 2.2.5.6 --- Interaction with Antibodies --- p.63 / Chapter 2.2.5.7 --- ECL Detection --- p.63 / Chapter 2.2.5.8 --- Data Analysis --- p.64 / Chapter Chapter 3: --- Results --- p.65 / Chapter 3.1 --- In vitro Studies --- p.66 / Chapter 3.1.1 --- Cell Viability test and IC50 --- p.66 / Chapter 3.1.2 --- Cell Cycle Analysis --- p.68 / Chapter 3.1.3 --- DNA Fragmentation --- p.71 / Chapter 3.1.4 --- Effects of Sinigrin on Gene Expression --- p.73 / Chapter 3.2 --- In vivo Studies --- p.77 / Chapter 3.2.1 --- Effects of Sinigrin on HCC Development (Promotion stage) in Rats --- p.77 / Chapter 3.2.1.1 --- Direct Observation --- p.77 / Chapter 3.2.1.2 --- Relative Liver / Body Weight Ratio --- p.79 / Chapter 3.2.1.3 --- AST/ALT Assay --- p.81 / Chapter 3.2.1.4 --- Basic Structure of Hepatocytes --- p.83 / Chapter 3.2.1.5 --- GST-p Foci Area --- p.85 / Chapter 3.2.1.6 --- mRNA Expression of p53 and Mdm2 --- p.88 / Chapter 3.2.1.7 --- Protein Expression of Biomarkers --- p.90 / Chapter 3.2.2 --- Effects of Sinigrin on HCC Development (Progression stage) in Rats --- p.97 / Chapter 3.2.2.1 --- Direct Observation --- p.97 / Chapter 3.2.2.2 --- Relative Liver / Body Weight Ratio --- p.99 / Chapter 3.2.2.3 --- AST/ALT Assay --- p.101 / Chapter 3.2.2.4 --- Basic Structure of Hepatocytes --- p.103 / Chapter 3.2.2.5 --- GST-p Foci Area --- p.105 / Chapter 3.2.2.6 --- mRNA Expression of p53 and Mdm2 --- p.108 / Chapter 3.2.2.7 --- Protein Expression of Biomarkers --- p.110 / Chapter Chapter 4: --- Discussion --- p.116 / Chapter 4.1 --- Protective and Therapeutic Benefits of Sinigrin --- p.117 / Chapter 4.1.1 --- Effects of SIN on Cancer and Normal Cells --- p.117 / Chapter 4.1.2 --- Effective Tumor Induction by DEN-CC14 Treatment --- p.118 / Chapter 4.1.3 --- Protective Effect of SIN in the Promotion Stage of HCC --- p.118 / Chapter 4.1.4 --- Therapeutic Effect of SIN in the Progression Stage of HCC --- p.119 / Chapter 4.2 --- Biological Activities of SIN --- p.121 / Chapter 4.3 --- Summary --- p.134 / References --- p.xiv
28

In vitro and in vivo study of effects of andrographolide on hepatocarcinogenesis.

January 2006 (has links)
Lau Ven Gie Vengie. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2006. / Includes bibliographical references (leaves 113-121). / Abstracts in English and Chinese. / ACKNOWLEDGEMENTS --- p.i / ABSTRACT --- p.ii / 論文摘要 --- p.iv / TABLE OF CONTENTS --- p.vi / LIST OF FIGURES --- p.ix / LIST OF TABLES --- p.x / LIST OF ABBREVIATIONS --- p.xi / INTRODUCTION --- p.1 / Chapter I --- Hepatocellular Carcinoma --- p.1 / Risk factors --- p.1 / Stages in chemical carcinogenesis --- p.2 / Initiation --- p.2 / Promotion --- p.3 / Progression --- p.5 / Treatment of hepatocarcinoma --- p.6 / Chemotherapy ´ؤ hepatic arterial infusion (HAI) --- p.6 / Trans-arterial chemoembolization (TACE) --- p.7 / Radiofrequency ablation (RFA) --- p.8 / Percutaneous ethanol injection (PEI) --- p.9 / Liver resection --- p.9 / Liver transplantation --- p.10 / Chapter II --- Molecular Mechanisms: Oncogenes and Tumor-suppressor genes --- p.11 / Cell cycle control --- p.12 / p53 mutation in HCC --- p.13 / Normal functions of p53 and its target genes --- p.13 / p21(Wafl/Cipl/Sdil) --- p.13 / PCNA --- p.14 / Bcl-2 and Bax: the Bcl-2 family --- p.14 / Mdm2 --- p.17 / Chapter III --- Evaluation of the effects of hepatocarcinogenesis --- p.19 / GST-Pi --- p.19 / AST & ALT --- p.19 / Chapter IV --- Traditional Chinese Medicine (TCM) --- p.21 / Andrographis Paniculata --- p.21 / Pharmacological properties of andrographolide --- p.23 / Chapter V --- Aim of the project --- p.26 / MATERIALS AND METHODS --- p.27 / Chapter 1 --- Effects of andrographolide on cell viability and cell cycle --- p.27 / Chapter 1.1 --- Materials and solutions --- p.27 / Chapter 1.2 --- Preparation of solutions --- p.28 / Chapter 1.3 --- Procedures --- p.29 / Chapter 1.3.1 --- Seeding cells into culture flask --- p.29 / Chapter 1.3.2 --- Subculturing technique --- p.30 / Chapter 1.3.3 --- Neutral red assay --- p.30 / Chapter 1.3.4 --- DNA purification of HepG2 cells --- p.31 / Chapter 1.3.5 --- DNA gel electrophoresis --- p.32 / Chapter 1.3.6 --- Flow cytometry --- p.32 / Chapter 2 --- Effects of andrographolide on gene expressions --- p.33 / Chapter 2.1 --- Materials and solutions --- p.33 / Chapter 2.2 --- Preparation of solutions --- p.34 / Chapter 2.3 --- Procedures --- p.35 / Chapter 2.3.1 --- Cell treatments --- p.35 / Chapter 2.3.2 --- mRNA extraction from cell --- p.35 / Chapter 2.3.3 --- Determination of total RNA yield and quality yield --- p.36 / Chapter 2.3.4 --- RNA formaldehyde agarose gel electrophoresis --- p.36 / Chapter 2.3.5 --- cDNA synthesis --- p.37 / Chapter 2.3.6 --- "cRNA synthesis, labeling and amplification" --- p.39 / Chapter 2.3.7 --- cRNA purification --- p.40 / Chapter 2.3.8 --- Oligo GEArray hybridization --- p.41 / Chapter 2.3.9 --- Chemiluminescent detection --- p.43 / Chapter 2.3.10 --- Data analysis --- p.44 / Chapter 3 --- Effects of andrographolide on hepatocarcinogenesis in rats --- p.45 / Chapter 3.1 --- Materials and solutions --- p.45 / Chapter 3.2 --- Preparation of solutions --- p.46 / Chapter 3.3 --- Procedures --- p.47 / Chapter 3.3.1 --- Animal treatment --- p.47 / Chapter 3.3.2 --- Promotion (Experiment 1) --- p.48 / Chapter 3.3.3 --- Progression (Experiment 2) --- p.49 / Chapter 3.3.4 --- Extraction of blood serum --- p.52 / Chapter 3.3.5 --- Measurement of absorbance --- p.52 / Chapter 3.3.6 --- Tissue processing --- p.53 / Chapter 3.3.7 --- Hematoxylin and Eosin (H&E) Staining --- p.53 / Chapter 3.3.8 --- Immunohistochemical staining of GST-P --- p.54 / Chapter 3.3.9 --- Examination of liver sections --- p.55 / Chapter 4 --- "Effects of andrographolide on the expressions of Mdm2, p53, PCNA, Bax, Bcl-2 & p21" --- p.56 / Chapter 4.1 --- Materials and solutions --- p.56 / Chapter 4.2 --- Preparation of solutions --- p.57 / Chapter 4.3 --- Procedures --- p.59 / Chapter 4.3.1 --- Total mRNA extraction from liver --- p.59 / Chapter 4.3.2 --- Reverse transcription of mRNA to cDNA --- p.59 / Chapter 4.3.3 --- Protocol for polymerase chain reaction (PCR) --- p.60 / Chapter 4.3.4 --- DNA gel electrophoresis --- p.61 / Chapter 4.3.5 --- Nuclear protein extraction --- p.61 / Chapter 4.3.6 --- Cytosolic protein extraction --- p.62 / Chapter 4.3.7 --- Determination of protein concentration --- p.62 / Chapter 4.3.8 --- Immunoprecipitation of p53 from liver nuclear protein --- p.62 / Chapter 4.3.9 --- Protein gel electrophoresis by SDS-PAGE --- p.63 / Chapter 4.3.10 --- Western blotting --- p.64 / RESULTS --- p.66 / Chapter 1 --- Effects of andrographolide on cell viability and cell cycle --- p.66 / Chapter 2 --- Effects of andrographolide on gene expressions --- p.76 / Chapter 3 --- Effects of andrographolide on hepatocarcinogenesis in rats --- p.79 / Chapter 4 --- "Effects of andrographolide on the expressions of Mdm2, p53, PCNA, Bax, Bcl-2 & p21" --- p.91 / DISCUSSION --- p.102 / CONCLUSION --- p.111 / REFERENCES --- p.113
29

Anti-tumor effect of arsenic trioxide (As₂O₃) on human breast cancer.

January 2007 (has links)
Zhou, Linli. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2007. / Includes bibliographical references (leaves 108-118). / Abstracts in English and Chinese. / Acknowledgements --- p.i / Abstract --- p.ii / 論文摘要 --- p.iv / Abbreviations --- p.v / List of Figures --- p.vii / List of Tables --- p.ix / Table of Contents --- p.x / Chapter Chapter 1 --- Introduction --- p.1 / Chapter 1.1 --- Breast Cancer --- p.1 / Chapter 1.1.1 --- Introduction to Breast Cancer --- p.1 / Chapter 1.1.2 --- Types of Breast Cancer --- p.3 / Chapter 1.1.3 --- Epidemiologic Risk Factors and Etiology --- p.4 / Chapter 1.2 --- Estrogen and Breast Cancer --- p.7 / Chapter 1.3 --- Estrogen Receptor --- p.9 / Chapter 1.4 --- Current Treatment of Breast Cancer --- p.10 / Chapter 1.4.1 --- Chemotherapy --- p.10 / Chapter 1.4.2 --- Hormonal (Anti-Estrogen) Therapy --- p.11 / Chapter 1.4.2.1 --- Tamoxifen and Other Anti-estrogens --- p.12 / Chapter 1.4.2.2 --- Disadvantages of Tamoxifen --- p.13 / Chapter 1.5 --- Arsenic Trioxide --- p.14 / Chapter 1.5.1 --- The Characteristics of Arsenic Trioxide (AS2O3) --- p.14 / Chapter 1.5.2 --- The Medical use of Arsenic Trioxide (As2O3) --- p.16 / Chapter 1.5.3 --- Arsenic Trioxide (As2O3) in treating Acute Promyelocytic Leukemia (APL) --- p.17 / Chapter 1.5.3.1 --- Acute Promyelocytic Leukemia (APL) --- p.17 / Chapter 1.5.3.2 --- All-trans Retinoic Acid (ATRA) Treatment of APL --- p.18 / Chapter 1.5.3.3 --- Clinical Trial of the Arsenic Trioxide on APL --- p.19 / Chapter 1.5.3.4 --- In vitro and in vivo Study of Arsenic Trioxide (As2O3) in treating APL --- p.19 / Chapter 1.5.3.5 --- Common Side Effects of Arsenic Trioxide (As2O3) on APL --- p.21 / Chapter 1.5.4 --- Anti-cancer effect of Arsenic Trioxide on other cancers --- p.23 / Chapter 1.6 --- Aim of Study --- p.24 / Chapter Chapter 2 --- Materials and Methods --- p.26 / Chapter 2.1 --- Materials --- p.27 / Chapter 2.1.1 --- Cell Lines and Culture Medium --- p.27 / Chapter 2.1.1.1 --- Cell Lines --- p.27 / Chapter 2.1.1.2 --- Culture Medium --- p.27 / Chapter 2.1.2 --- Chemicals --- p.28 / Chapter 2.1.3 --- Buffers and Reagents --- p.29 / Chapter 2.1.4 --- Reagents for MTT Assay --- p.30 / Chapter 2.1.5 --- Reagents for DNA Fragmentation --- p.31 / Chapter 2.1.5.1 --- Reagents for DNA Extraction --- p.31 / Chapter 2.1.5.2 --- Reagents for Gel Electrophoresis --- p.31 / Chapter 2.1.6 --- Reagents for Western Blotting --- p.32 / Chapter 2.1.6.1 --- Reagents for Protein Extraction --- p.32 / Chapter 2.1.6.2 --- Reagents for SDS-PAGE --- p.33 / Chapter 2.1.7 --- Reagents for Flow Cytometry --- p.36 / Chapter 2.1.8 --- In Vivo Study --- p.37 / Chapter 2.2 --- Methods --- p.38 / Chapter 2.2.1 --- Cell Treatment --- p.38 / Chapter 2.2.2 --- Trypan Blue Exclusion Assay --- p.38 / Chapter 2.2.3 --- MTT Assay --- p.38 / Chapter 2.2.4 --- Detection of DNA Fragmentation --- p.39 / Chapter 2.2.5 --- Flow Cytometry --- p.40 / Chapter 2.2.5.1 --- Detection of Cell Cycle Pattern with PI --- p.40 / Chapter 2.2.5.2 --- Detection of Apoptosis with Annexin V-PI --- p.40 / Chapter 2.2.6 --- Western Blot Analysis --- p.41 / Chapter 2.2.6.1 --- Protein Extraction --- p.41 / Chapter 2.2.6.2 --- Protein Concentration Determination --- p.41 / Chapter 2.2.6.3 --- Western Blotting --- p.42 / Chapter 2.2.7 --- In Vivo Study --- p.44 / Chapter 2.2.7.1 --- Animal Model --- p.44 / Chapter 2.2.7.2 --- Treatment Schedule --- p.44 / Chapter 2.2.7.3 --- Toxicity of Arsenic Trioxide --- p.45 / Chapter Chapter 3 --- Anti-Proliferation Effect of As2O3 on MDA-MB-231 cells --- p.47 / Chapter 3.1 --- Study the Anti-proliferation Effect of As2O3 on MDA-MB-231 Cells by MTT Assay --- p.48 / Chapter 3.2 --- Comparsion Anti-proliferation Effect of AS2O3 on MDA-MB-231 Cells to that of Tamoxifen --- p.50 / Chapter 3.3 --- "Study Toxicity of AS2O3 on Normal Breast Cells Line, 184B5" --- p.52 / Chapter 3.4 --- Summary --- p.54 / Chapter Chapter 4 --- Mechanism of Growth Inhibition Effect of As2O3 on MDA-MB-231 cells --- p.56 / Chapter 4.1 --- Cell Cycle Analysis of As2O3 Treated MDA-MB-231 Cells --- p.57 / Chapter 4.2 --- Detection of DNA Fragmentation --- p.60 / Chapter 4.3 --- Detection of Apoptosis Induced by AS2O3 on MDA-MB-231 Cells by Flow Cytometry --- p.62 / Chapter 4.4 --- Regulation of Apoptotic Related Protein by As2O3 on MDA-MB-231 Cells --- p.64 / Chapter 4.4.1 --- Expression Level of Bcl-2 and Bax Protein --- p.66 / Chapter 4.4.2 --- Expression Level of Cytochrome C --- p.69 / Chapter 4.4.3 --- Expression Level of Caspase9 --- p.71 / Chapter 4.4.4 --- Expression Level of FasL --- p.73 / Chapter 4.4.5 --- Expression Level of Caspase8 --- p.75 / Chapter 4.4.6 --- Expression Level of Caspase3 --- p.77 / Chapter 4.4.7 --- Expression Level of Poly (ADP-ribose) Polymerase (PARP) --- p.79 / Chapter 4.4.8 --- Expression Level of p53 --- p.81 / Chapter 4.5 --- Regulation of Cell Cycle Related Protein by AS2O3 on MDA-MB-231 Cells --- p.83 / Chapter 4.5.1 --- Expression Level of Cyclin B --- p.84 / Chapter 4.5.2 --- Expression Level of Cyclin E --- p.86 / Chapter 4.6 --- Summary --- p.88 / Chapter Chapter 5 --- In Vivo Study of Anti-tumor Effect of As2O3 --- p.89 / Chapter 5.1 --- Anti-tumor Effect of AS2O3 on Tumor Bearing Nude Mice --- p.90 / Chapter 5.2 --- Toxic Effect of AS2O3 on Normal Tissues --- p.93 / Chapter 5.3 --- Summary --- p.98 / Chapter Chapter 6 --- Discussion --- p.99 / Chapter 6.1 --- Anti-tumor Effect of AS2O3 on Breast Cancer --- p.100 / Chapter 6.2 --- Induction of Apoptosis and Cell Cycle arrest by AS2O3 --- p.101 / Chapter 6.3 --- Side Effect of AS2O3 on Breast Cancer Treatment --- p.103 / Chapter Chapter 7 --- Future Perspectives --- p.105 / Chapter 7.1 --- Future Perspectives --- p.106 / References --- p.108
30

A chemical-biology approach for screening novel inhibitors of focal adhesion signaling in relation to breast cancer /

Cao, Yangxiezi. January 2008 (has links)
Focal adhesion kinase (FAK), a non-receptor kinase, is a key regulator of integrin and focal adhesion signaling required for cancer cell survival, cell migration, and cell invasion. Amplification/Overexpression of FAK occurs in a wide variety of human cancers, supporting a role in carcinogenesis. Moreover, preclinical studies using cancer models where FAK is genetically inhibited indicate that this kinase is a potential therapeutic target to interfere with cancer progression. However, very little progress has been made in the identification of chemical inhibitors for potential therapeutic applications, in contrast to other kinases. Herein, I report optimization of the high-throughput in vitro Glo kinase assay for screening inhibitors of FAK kinase activity. Screening a large library of small molecule chemicals using these assays identified at least twenty FAK inhibitors, including a new FAK inhibitor developed by Pfizer and undergoing human clinical trials, and the non-specific kinase inhibitor staurosporine. Molecular studies of selective FAK inhibitors are undergoing in my host laboratory. In addition to this in vitro assay, I established similar assays to examine FAK kinase and adapter function in intact cells. The latter consists of ErbB-transformed cells deficient in FAK, and their matched cells where wild-type or kinase-dead FAK was restored. Biological characterization of these models revealed that both FAK kinase and adaptor activities cooperate for the regulation of cell migration, cell invasion, and tumor formation.

Page generated in 0.0566 seconds