Interactions of Bartonella Henselae with Myeloid Angiogenic Cells and Consequences for Pathological Angiogenesis

Total Page:16

File Type:pdf, Size:1020Kb

Interactions of Bartonella Henselae with Myeloid Angiogenic Cells and Consequences for Pathological Angiogenesis Interactions of Bartonella henselae with Myeloid Angiogenic Cells and Consequences for Pathological Angiogenesis Dissertation zur Erlangung des Doktorgrades der Naturwissenschaften vorgelegt beim Fachbereich Biowissenschaften der Johann Wolfgang Goethe-Universität in Frankfurt am Main von Fiona OʼRourke aus Calgary (Kanada) Frankfurt am Main 2015 vom Fachbereich Biowissenschaften der Johann Wolfgang Goethe-Universität als Dissertation angenommen. Dekanin: Prof. Dr. Meike Piepenbring 1. Gutachter: Prof. Dr. Volker Müller 2. Gutachter: Prof. Dr. Volkhard A. J. Kempf Datum der Disputation: 02.12.2015 I Table of Contents 1. Introduction .................................................................................................... 1 1.1 Bartonella ................................................................................................................................1 1.2 Bartonella henselae ..................................................................................................................2 1.3 Infection-associated pathological angiogenesis ..........................................................................2 1.4 B. henselae pathogenicity strategy ............................................................................................4 1.4.1 Mitogenic stimulus ....................................................................................................................... 6 1.4.2 Inhibtion of apoptosis .................................................................................................................. 7 1.4.3 HIF/ VEGF ..................................................................................................................................... 7 1.4.4 Myeloid cells and the inflammatory response............................................................................. 8 1.4.5 Immune evasion ........................................................................................................................... 9 1.5 B. henselae pathogenicity factors ............................................................................................ 10 1.5.1 Bartonella Adhesin A (BadA) ...................................................................................................... 10 1.5.2 VirB/D4 Type IV Secretion System (T4SS) .................................................................................. 11 1.5.3 Interaction of BadA and T4SS ..................................................................................................... 12 1.6 Angiogenesis .......................................................................................................................... 13 1.6.1 Development of the vascular network ...................................................................................... 13 1.6.2 The angiogenic process .............................................................................................................. 13 1.6.3 Angiogenesis in health and disease ........................................................................................... 16 1.7 Tumor angiogenesis ................................................................................................................ 17 1.8 Angiogenic progenitor cells ..................................................................................................... 18 1.8.1 Endothelial Progenitor Cells (EPCs) ............................................................................................ 18 1.8.2 Myeloid Angiogenic Cells (MACs) .............................................................................................. 19 1.9 Monocytes and macrophages .................................................................................................. 22 1.9.1 Macrophage functions ............................................................................................................... 22 1.9.2 Macrophage polarization ........................................................................................................... 23 1.10 Macrophages and angiogenesis ............................................................................................. 25 1.10.1 Macrophages and pathological angiogenesis .......................................................................... 27 1.11 Tumor Associated Macrophages (TAM)s ................................................................................ 27 1.12 Interaction of bacteria with stem and progenitor cells ........................................................... 30 1.13 Interaction of B. henselae with MACs .................................................................................... 32 2. Objectives ..................................................................................................... 33 3. Materials and Methods ................................................................................. 34 3.1 Laboratory equipment and analytical instruments ................................................................... 34 3.2 Laboratory consumables ......................................................................................................... 35 3.3 Chemicals and reagents .......................................................................................................... 36 3.4 Kits ......................................................................................................................................... 37 3.5 Bacteria .................................................................................................................................. 38 3.5.1 Bacterial strains and growth conditions .................................................................................... 38 3.5.2 Preparation of Bartonella henselae bacterial stocks ................................................................. 38 II 3.6 Cell culture ............................................................................................................................. 39 3.6.1 Ethics statement ........................................................................................................................ 39 3.6.2 Human umbilical vein endothelial cells (HUVECs) ..................................................................... 40 3.6.3 Isolation and cultivation of Myeloid Angiogenic Cells (MACs) .................................................. 40 3.7 Infection experiments ............................................................................................................. 42 3.8 Electron microscopy ................................................................................................................ 42 3.9 Nicoletti apoptosis assay ......................................................................................................... 43 3.10 HIF-1 Western Blot ................................................................................................................ 43 3.10.1 Cell stimulation and preparation of protein extracts .............................................................. 44 3.10.2 Sodium dodecyl sulfate- polyacrylamice gel electrophoresis (SDS-PAGE) .............................. 44 3.10.3 Western Blot ............................................................................................................................ 44 3.11 Spheroid assay of sprouting angiogenesis .............................................................................. 47 3.12 Matrigel capillary formation assay ......................................................................................... 48 3.13 16S rDNA PCR ....................................................................................................................... 49 3.14 Flow cytometry ..................................................................................................................... 51 3.15 Immunohistochemistry ......................................................................................................... 53 3.16 Quantitative real-time PCR (qRT-PCR) .................................................................................... 56 3.16.1 Isolation of total RNA ............................................................................................................... 56 3.16.2 Reverse Transcription PCR (RT-PCR) ........................................................................................ 56 3.16.3 Quantitative RT-PCR ................................................................................................................. 57 3.17 Microarray gene expression profiling ..................................................................................... 58 3.18 Secretome analysis ............................................................................................................... 60 3.19 Enzyme linked immunosorbent assays (ELISAs) ...................................................................... 60 3.20 Statistical analysis ................................................................................................................. 61 4. Results .......................................................................................................... 62 4.1 B. henselae invades MACs and resides in intracellular vacuoles .......................................... 62 4.2 B. henselae infected MACs maintain high viability ............................................................. 62 4.3 B. henselae infection of MACs induces BadA dependent HIF-1 activation ..........................
Recommended publications
  • An Investigation of ADAM-Like Decysin 1 in Macrophage-Mediated Inflammation and Crohn’S Disease
    An Investigation of ADAM-like Decysin 1 in Macrophage-mediated Inflammation and Crohn’s Disease By Nuala Roisin O’Shea A thesis submitted to UCL for the degree of Doctor of Philosophy Division of Medicine Declaration I, Nuala Roisin O’Shea, confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. 2 Abstract Crohn’s disease (CD) is now recognised as a defective host response to bacteria in genetically susceptible individuals. The role of innate immunity and impaired bacterial clearance are widely accepted. In this thesis the role of ADAM-like, Decysin-1 (ADAMDEC1) in macrophage-mediated inflammation and gut mucosal immunity is explored. Using transcriptomic analysis of monocyte derived macrophages (MDM) ADAMDEC1 was identified as grossly under expressed in a subset of patients with CD. ADAMDEC1 was found to be highly selective to the intestine, peripheral blood monocyte-derived and lamina propria macrophages. It was shown to be an inflammatory response gene, upregulated in response to bacterial antigens and inflammation. ADAMDEC1 was expressed in prenatal and germ free mice, demonstrating exposure to a bacterial antigen is not a prerequisite for expression. Adamdec1 knock out mice were used to investigate the role of ADAMDEC1 in vivo. Adamdec1-/- mice displayed an increased susceptibility to dextran sodium sulphate (DSS), Citrobacter rodentium and Salmonella typhimurium induced colitis. In Adamdec1-/- mice, bacterial translocation and systemic infection were increased in bacterial models of colitis. These results suggest that individuals with grossly attenuated expression of ADAMDEC1 may be at an increased risk of developing intestinal inflammation as a consequence of an impaired ability to handle enteric bacterial pathogens.
    [Show full text]
  • Proteomic Analysis of Human Osteoarthritis Synovial Fluid
    Balakrishnan et al. Clinical Proteomics 2014, 11:6 http://www.clinicalproteomicsjournal.com/content/11/1/6 CLINICAL PROTEOMICS RESEARCH Open Access Proteomic analysis of human osteoarthritis synovial fluid Lavanya Balakrishnan1,2, Raja Sekhar Nirujogi1,3, Sartaj Ahmad1,4, Mitali Bhattacharjee1,5, Srikanth S Manda1,3, Santosh Renuse1,5, Dhanashree S Kelkar1,5, Yashwanth Subbannayya1,6, Rajesh Raju1, Renu Goel1,2, Joji Kurian Thomas1,5, Navjyot Kaur7, Mukesh Dhillon7, Shantal Gupta Tankala7, Ramesh Jois8, Vivek Vasdev9, YL Ramachandra2, Nandini A Sahasrabuddhe1, TS Keshava Prasad1,3,4, Sujatha Mohan10, Harsha Gowda1, Subramanian Shankar7* and Akhilesh Pandey11,12,13,14* Abstract Background: Osteoarthritis is a chronic musculoskeletal disorder characterized mainly by progressive degradation of the hyaline cartilage. Patients with osteoarthritis often postpone seeking medical help, which results in the diagnosis being made at an advanced stage of cartilage destruction. Sustained efforts are needed to identify specific markers that might help in early diagnosis, monitoring disease progression and in improving therapeutic outcomes. We employed a multipronged proteomic approach, which included multiple fractionation strategies followed by high resolution mass spectrometry analysis to explore the proteome of synovial fluid obtained from osteoarthritis patients. In addition to the total proteome, we also enriched glycoproteins from synovial fluid using lectin affinity chromatography. Results: We identified 677 proteins from synovial fluid of patients
    [Show full text]
  • The Conserved DNMT1-Dependent Methylation Regions in Human Cells
    Freeman et al. Epigenetics & Chromatin (2020) 13:17 https://doi.org/10.1186/s13072-020-00338-8 Epigenetics & Chromatin RESEARCH Open Access The conserved DNMT1-dependent methylation regions in human cells are vulnerable to neurotoxicant rotenone exposure Dana M. Freeman1 , Dan Lou1, Yanqiang Li1, Suzanne N. Martos1 and Zhibin Wang1,2,3* Abstract Background: Allele-specifc DNA methylation (ASM) describes genomic loci that maintain CpG methylation at only one inherited allele rather than having coordinated methylation across both alleles. The most prominent of these regions are germline ASMs (gASMs) that control the expression of imprinted genes in a parent of origin-dependent manner and are associated with disease. However, our recent report reveals numerous ASMs at non-imprinted genes. These non-germline ASMs are dependent on DNA methyltransferase 1 (DNMT1) and strikingly show the feature of random, switchable monoallelic methylation patterns in the mouse genome. The signifcance of these ASMs to human health has not been explored. Due to their shared allelicity with gASMs, herein, we propose that non-tradi- tional ASMs are sensitive to exposures in association with human disease. Results: We frst explore their conservancy in the human genome. Our data show that our putative non-germline ASMs were in conserved regions of the human genome and located adjacent to genes vital for neuronal develop- ment and maturation. We next tested the hypothesized vulnerability of these regions by exposing human embryonic kidney cell HEK293 with the neurotoxicant rotenone for 24 h. Indeed,14 genes adjacent to our identifed regions were diferentially expressed from RNA-sequencing. We analyzed the base-resolution methylation patterns of the predicted non-germline ASMs at two neurological genes, HCN2 and NEFM, with potential to increase the risk of neurodegenera- tion.
    [Show full text]
  • Inflammatory Modulation of Hematopoietic Stem Cells by Magnetic Resonance Imaging
    Electronic Supplementary Material (ESI) for RSC Advances. This journal is © The Royal Society of Chemistry 2014 Inflammatory modulation of hematopoietic stem cells by Magnetic Resonance Imaging (MRI)-detectable nanoparticles Sezin Aday1,2*, Jose Paiva1,2*, Susana Sousa2, Renata S.M. Gomes3, Susana Pedreiro4, Po-Wah So5, Carolyn Ann Carr6, Lowri Cochlin7, Ana Catarina Gomes2, Artur Paiva4, Lino Ferreira1,2 1CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal, 2Biocant, Biotechnology Innovation Center, Cantanhede, Portugal, 3King’s BHF Centre of Excellence, Cardiovascular Proteomics, King’s College London, London, UK, 4Centro de Histocompatibilidade do Centro, Coimbra, Portugal, 5Department of Neuroimaging, Institute of Psychiatry, King's College London, London, UK, 6Cardiac Metabolism Research Group, Department of Physiology, Anatomy & Genetics, University of Oxford, UK, 7PulseTeq Limited, Chobham, Surrey, UK. *These authors contributed equally to this work. #Correspondence to Lino Ferreira ([email protected]). Experimental Section Preparation and characterization of NP210-PFCE. PLGA (Resomers 502 H; 50:50 lactic acid: glycolic acid) (Boehringer Ingelheim) was covalently conjugated to fluoresceinamine (Sigma- Aldrich) according to a protocol reported elsewhere1. NPs were prepared by dissolving PLGA (100 mg) in a solution of propylene carbonate (5 mL, Sigma). PLGA solution was mixed with perfluoro- 15-crown-5-ether (PFCE) (178 mg) (Fluorochem, UK) dissolved in trifluoroethanol (1 mL, Sigma). This solution was then added to a PVA solution (10 mL, 1% w/v in water) dropwise and stirred for 3 h. The NPs were then transferred to a dialysis membrane and dialysed (MWCO of 50 kDa, Spectrum Labs) against distilled water before freeze-drying. Then, NPs were coated with protamine sulfate (PS).
    [Show full text]
  • Genomic and Transcriptomic Investigations Into the Feed Efficiency Phenotype of Beef Cattle
    Provided by the author(s) and NUI Galway in accordance with publisher policies. Please cite the published version when available. Title Genomic and transcriptomic investigations into the feed efficiency phenotype of beef cattle Author(s) Higgins, Marc Publication Date 2019-03-06 Publisher NUI Galway Item record http://hdl.handle.net/10379/15008 Downloaded 2021-09-25T18:07:39Z Some rights reserved. For more information, please see the item record link above. Genomic and Transcriptomic Investigations into the Feed Efficiency Phenotype of Beef Cattle Marc Higgins, B.Sc., M.Sc. A thesis submitted for the Degree of Doctor of Philosophy to the Discipline of Biochemistry, School of Natural Sciences, National University of Ireland, Galway. Supervisor: Dr. Derek Morris Discipline of Biochemistry, School of Natural Sciences, National University of Ireland, Galway. Supervisor: Dr. Sinéad Waters Teagasc, Animal and Bioscience Research Department, Animal & Grassland Research and Innovation Centre, Teagasc, Grange. Submitted November 2018 Table of Contents Declaration ................................................................................................................ vii Funding .................................................................................................................... viii Acknowledgements .................................................................................................... ix Abstract ......................................................................................................................
    [Show full text]
  • Original Article Cytochrome P450 Family Proteins As Potential Biomarkers for Ovarian Granulosa Cell Damage in Mice with Premature Ovarian Failure
    Int J Clin Exp Pathol 2018;11(8):4236-4246 www.ijcep.com /ISSN:1936-2625/IJCEP0080020 Original Article Cytochrome P450 family proteins as potential biomarkers for ovarian granulosa cell damage in mice with premature ovarian failure Jiajia Lin1, Jiajia Zheng1, Hu Zhang1, Jiulin Chen1, Zhihua Yu1, Chuan Chen1, Ying Xiong3, Te Liu1,2 1Shanghai Geriatric Institute of Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; 2Department of Pathology, Yale UniversitySchool of Medicine, New Haven, USA; 3Department of Gynaecology and Obestetrics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China Received May 21, 2018; Accepted June 29, 2018; Epub August 1, 2018; Published August 15, 2018 Abstract: Premature ovarian failure (POF) is the pathological aging of ovarian tissue. We have previously established a cyclophosphamide-induced mouse POF model and found that cyclophosphamide caused significant damage and apoptosis of mouse ovarian granulosa cells (mOGCs). To systematically explore the molecular biologic evidence of cyclophosphamide-induced mOGC damage at the gene transcription level, RNA-Seqwas used to analyse the differ- ences in mOGC transcriptomes between POF and control (PBS) mice. The sequencing results showed that there were 18765 differential transcription genes between the two groups, of which 192 were significantly up-regulated (log2 [POF/PBS] > 2.0) and 116 were significantly down-regulated (log2 [POF/PBS] < -4.0). Kyoto Encyclopedia of Genes and Genomes analysis found that the neuroactive ligand-receptor interaction pathway was significantly up-regulated and metabolic pathways were significantly down-regulated in the POF group. Gene Ontology analy- sis showed that the expression of plasma membrane, regulation of transcription and ion binding functions were significantly up-regulated in the POF group, while the expression of cell and cell parts, catalytic activity and single- organism process functions were significantly down-regulated.
    [Show full text]
  • Prediction of Human Disease Genes by Human-Mouse Conserved Coexpression Analysis
    Prediction of Human Disease Genes by Human-Mouse Conserved Coexpression Analysis Ugo Ala1., Rosario Michael Piro1., Elena Grassi1, Christian Damasco1, Lorenzo Silengo1, Martin Oti2, Paolo Provero1*, Ferdinando Di Cunto1* 1 Molecular Biotechnology Center, Department of Genetics, Biology and Biochemistry, University of Turin, Turin, Italy, 2 Department of Human Genetics and Centre for Molecular and Biomolecular Informatics, University Medical Centre Nijmegen, Nijmegen, The Netherlands Abstract Background: Even in the post-genomic era, the identification of candidate genes within loci associated with human genetic diseases is a very demanding task, because the critical region may typically contain hundreds of positional candidates. Since genes implicated in similar phenotypes tend to share very similar expression profiles, high throughput gene expression data may represent a very important resource to identify the best candidates for sequencing. However, so far, gene coexpression has not been used very successfully to prioritize positional candidates. Methodology/Principal Findings: We show that it is possible to reliably identify disease-relevant relationships among genes from massive microarray datasets by concentrating only on genes sharing similar expression profiles in both human and mouse. Moreover, we show systematically that the integration of human-mouse conserved coexpression with a phenotype similarity map allows the efficient identification of disease genes in large genomic regions. Finally, using this approach on 850 OMIM loci characterized by an unknown molecular basis, we propose high-probability candidates for 81 genetic diseases. Conclusion: Our results demonstrate that conserved coexpression, even at the human-mouse phylogenetic distance, represents a very strong criterion to predict disease-relevant relationships among human genes. Citation: Ala U, Piro RM, Grassi E, Damasco C, Silengo L, et al.
    [Show full text]
  • A Genomic Analysis of Rat Proteases and Protease Inhibitors
    A genomic analysis of rat proteases and protease inhibitors Xose S. Puente and Carlos López-Otín Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, 33006-Oviedo, Spain Send correspondence to: Carlos López-Otín Departamento de Bioquímica y Biología Molecular Facultad de Medicina, Universidad de Oviedo 33006 Oviedo-SPAIN Tel. 34-985-104201; Fax: 34-985-103564 E-mail: [email protected] Proteases perform fundamental roles in multiple biological processes and are associated with a growing number of pathological conditions that involve abnormal or deficient functions of these enzymes. The availability of the rat genome sequence has opened the possibility to perform a global analysis of the complete protease repertoire or degradome of this model organism. The rat degradome consists of at least 626 proteases and homologs, which are distributed into five catalytic classes: 24 aspartic, 160 cysteine, 192 metallo, 221 serine, and 29 threonine proteases. Overall, this distribution is similar to that of the mouse degradome, but significatively more complex than that corresponding to the human degradome composed of 561 proteases and homologs. This increased complexity of the rat protease complement mainly derives from the expansion of several gene families including placental cathepsins, testases, kallikreins and hematopoietic serine proteases, involved in reproductive or immunological functions. These protease families have also evolved differently in the rat and mouse genomes and may contribute to explain some functional differences between these two closely related species. Likewise, genomic analysis of rat protease inhibitors has shown some differences with the mouse protease inhibitor complement and the marked expansion of families of cysteine and serine protease inhibitors in rat and mouse with respect to human.
    [Show full text]
  • [ADAMDEC1] for Intestinal Immunity and Inflammation Nuala R
    Journal of Crohn's and Colitis, 2016, 1417–1427 doi:10.1093/ecco-jcc/jjw111 Advance Access publication May 25, 2016 Original Article Original Article Critical Role of the Disintegrin Metalloprotease ADAM-like Decysin-1 [ADAMDEC1] for Intestinal Immunity and Inflammation Nuala R. O’Shea,a Thean S. Chew,a Jenny Dunne,a Rebecca Marnane,a Bahman Nedjat-Shokouhi,a Philip J. Smith,a Stuart L. Bloom,b Andrew M. Smith,a,c,* Anthony W. Segala,* aDivision of Medicine, University College London, London, UK bDepartment of Gastroenterology, University College London Hospital, UK cMicrobial Diseases, Eastman Dental Institute, University College London, London, UK *These authors contributed equally to this work. Corresponding author: Andrew M. Smith, PhD, Microbial Diseases, Eastman Dental Institute, University College London, London WC1X 8LD, UK. E-mail: [email protected] Conference presentations: ECCO Barcelona, DDW San Diego, BSG Nottingham 2012; ECCO Vienna, DDW Chicago, BSG Glasgow 2013; Keystone Sante Fe, 2014 Abstract Background and Aims: ADAM [A Disintegrin And Metalloproteinase] is a family of peptidase proteins which have diverse roles in tissue homeostasis and immunity. Here, we study ADAM- like DECysin-1 [ADAMDEC1] a unique member of the ADAM family. ADAMDEC1 expression is restricted to the macrophage/dendritic cell populations of the gastrointestinal tract and secondary lymphoid tissue. The biological function of ADAMDEC1 is unknown but it has been hypothesised to play a role in immunity. The identification of reduced ADAMDEC1 expression in Crohn’s disease patients has provided evidence of a potential role in bowel inflammation. Methods: Adamdec1-/- mice were exposed to dextran sodium sulphate or infected orally with Citrobacter rodentium or Salmonella typhimurium.
    [Show full text]
  • The Conserved DNMT1 Dependent Methylation Regions in Human Cells Are Vulnerable to Environmental Rotenone
    bioRxiv preprint doi: https://doi.org/10.1101/798587; this version posted October 9, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The conserved DNMT1 dependent methylation regions in human cells are vulnerable to environmental rotenone. Dana M. Freemana, Dan Loua, Yanqiang Lia, Suzanne N. Martosa, Zhibin Wanga* aLaboratory of Environmental Epigenomes, Department of Environmental Health & Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD *To whom correspondence should be addressed: Zhibin Wang, Ph.D., Associate Professor, Laboratory of Environmental Epigenomes, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD. Phone: (410) 955-7840; Email: [email protected] Abstract Allele-specific DNA methylation (ASM) describes genomic loci that maintain CpG methylation at only one inherited allele rather than having coordinated methylation across both alleles. The most prominent of these regions are germline ASMs (gASMs) that control the expression of imprinted genes in a parent of origin- dependent manner and are associated with disease. However, our recent report reveals numerous ASMs at non-imprinted genes. These non-germline ASMs are dependent on DNA methyltransferase 1 (DNMT1) and strikingly show the feature of random, switchable monoallelic methylation patterns in the mouse genome. The significance of these ASMs to human health has not been explored. Due to their shared allelicity with gASMs, herein, we propose that non-traditional ASMs are sensitive to exposures in association with human disease. We first explore their conservancy in the human genome.
    [Show full text]
  • Content Based Search in Gene Expression Databases and a Meta-Analysis of Host Responses to Infection
    Content Based Search in Gene Expression Databases and a Meta-analysis of Host Responses to Infection A Thesis Submitted to the Faculty of Drexel University by Francis X. Bell in partial fulfillment of the requirements for the degree of Doctor of Philosophy November 2015 c Copyright 2015 Francis X. Bell. All Rights Reserved. ii Acknowledgments I would like to acknowledge and thank my advisor, Dr. Ahmet Sacan. Without his advice, support, and patience I would not have been able to accomplish all that I have. I would also like to thank my committee members and the Biomed Faculty that have guided me. I would like to give a special thanks for the members of the bioinformatics lab, in particular the members of the Sacan lab: Rehman Qureshi, Daisy Heng Yang, April Chunyu Zhao, and Yiqian Zhou. Thank you for creating a pleasant and friendly environment in the lab. I give the members of my family my sincerest gratitude for all that they have done for me. I cannot begin to repay my parents for their sacrifices. I am eternally grateful for everything they have done. The support of my sisters and their encouragement gave me the strength to persevere to the end. iii Table of Contents LIST OF TABLES.......................................................................... vii LIST OF FIGURES ........................................................................ xiv ABSTRACT ................................................................................ xvii 1. A BRIEF INTRODUCTION TO GENE EXPRESSION............................. 1 1.1 Central Dogma of Molecular Biology........................................... 1 1.1.1 Basic Transfers .......................................................... 1 1.1.2 Uncommon Transfers ................................................... 3 1.2 Gene Expression ................................................................. 4 1.2.1 Estimating Gene Expression ............................................ 4 1.2.2 DNA Microarrays ......................................................
    [Show full text]
  • Global Patterns of Changes in the Gene Expression Associated with Genesis of Cancer a Dissertation Submitted in Partial Fulfillm
    Global Patterns Of Changes In The Gene Expression Associated With Genesis Of Cancer A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy at George Mason University By Ganiraju Manyam Master of Science IIIT-Hyderabad, 2004 Bachelor of Engineering Bharatiar University, 2002 Director: Dr. Ancha Baranova, Associate Professor Department of Molecular & Microbiology Fall Semester 2009 George Mason University Fairfax, VA Copyright: 2009 Ganiraju Manyam All Rights Reserved ii DEDICATION To my parents Pattabhi Ramanna and Veera Venkata Satyavathi who introduced me to the joy of learning. To friends, family and colleagues who have contributed in work, thought, and support to this project. iii ACKNOWLEDGEMENTS I would like to thank my advisor, Dr. Ancha Baranova, whose tolerance, patience, guidance and encouragement helped me throughout the study. This dissertation would not have been possible without her ever ending support. She is very sincere and generous with her knowledge, availability, compassion, wisdom and feedback. I would also like to thank Dr. Vikas Chandhoke for funding my research generously during my doctoral study at George Mason University. Special thanks go to Dr. Patrick Gillevet, Dr. Alessandro Giuliani, Dr. Maria Stepanova who devoted their time to provide me with their valuable contributions and guidance to formulate this project. Thanks to the faculty of Molecular and Micro Biology (MMB) department, Dr. Jim Willett and Dr. Monique Vanhoek in embedding valuable thoughts to this dissertation by being in my dissertation committee. I would also like to thank the present and previous doctoral program directors, Dr. Daniel Cox and Dr. Geraldine Grant, for facilitating, allowing, and encouraging me to work in this project.
    [Show full text]