Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

Medical and Health Genomics
Medical and Health Genomics
Medical and Health Genomics
Ebook1,246 pages11 hours

Medical and Health Genomics

Rating: 0 out of 5 stars

()

Read preview

About this ebook

Medical and Health Genomics provides concise and evidence-based technical and practical information on the applied and translational aspects of genome sciences and the technologies related to non-clinical medicine and public health. Coverage is based on evolving paradigms of genomic medicine—in particular, the relation to public and population health genomics now being rapidly incorporated in health management and administration, with further implications for clinical population and disease management.

  • Provides extensive coverage of the emergent field of health genomics and its huge relevance to healthcare management
  • Presents user-friendly language accompanied by explanatory diagrams, figures, and many references for further study
  • Covers the applied, but non-clinical, sciences across disease discovery, genetic analysis, genetic screening, and prevention and management
  • Details the impact of clinical genomics across a diverse array of public and community health issues, and within a variety of global healthcare systems
LanguageEnglish
Release dateJun 4, 2016
ISBN9780127999227
Medical and Health Genomics

Related to Medical and Health Genomics

Related ebooks

Medical For You

View More

Related articles

Related categories

Reviews for Medical and Health Genomics

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    Medical and Health Genomics - Dhavendra Kumar

    Medical and Health Genomics

    Editors

    Dhavendra Kumar

    Institute of Cancer & Genetics University Hospital of Wales, Cardiff University School of Medicine Cardiff, UK

    Genomic Policy Unit Faculty of Life Sciences and Education, University of South Wales Pontypridd, UK

    Stylianos Antonarakis

    Department of Medical Genetics, University Hospitals of Geneva, Switzerland

    Institute of Genetics and Genomics of Geneva, Geneva, University of Switzerland

    Table of Contents

    Cover image

    Title page

    Medical and Health Genomics

    Dedication

    Copyright

    List of Contributors

    Foreword

    Preface

    Chapter 1. The Human Genome

    Introduction

    Hereditary Factors, Genes, Genetics, and Genomics

    Structure and Organization of Nucleic Acids

    Human Genome Variation and Human Disease

    The Mitochondrial Genome

    Functional Genomics, Transcriptomics, and Proteomics

    Translational Human Genomics

    Human Genomics for Socioeconomic Development

    Conclusions

    Chapter 2. Genomic Technologies in Medicine and Health: Past, Present, and Future

    Introduction

    Sequencing Technologies

    Computational and Information Technologies

    Applications of Genomic Technologies

    The Microbiome and Human Health

    The 1000 Genomes Project and Structural Variations in the Human Genomes

    Noninvasive Prenatal Testing by Sequencing of Cell-Free Fetal DNA in the Maternal Blood

    Prenatal Diagnosis by Whole Genome Sequencing of Jumping Libraries

    Postnatal Diagnosis

    Genome Sequencing in Newborn Healthcare

    Genome Sequencing in Other Research Areas

    The Cancer Genome Atlas and Cancer Genomics

    Cancer Diagnosis

    Precision Medicine

    Policy and Regulatory Issues

    Genomic Education

    Chapter 3. Genomic Databases, Access Review, and Data Access Committees

    Introduction

    Underlying Principles, Policies, and Guidelines

    Examples of Controlled-Access Databases and Pertinent Data Access Committees

    Ethical and Legal Challenges

    Conclusion

    Chapter 4. Diagnostic Genomics and Clinical Bioinformatics

    Introduction

    The UK100K Genomes Project: Large-Scale Implementation of Whole Genome Sequencing

    Conclusion

    Chapter 5. Epigenetics and Epigenomics in Human Health and Disease

    Introduction

    Epigenotype and Regulation of Gene Expression

    Epigenotypes and Human Disease

    Epigenetic Mechanisms in Cancer

    Conclusion

    Chapter 6. Mitochondrial Genomics: Emerging Paradigms and Challenges

    Introduction

    Nongenetic Diagnostics for Mitochondrial Dysfunction

    Genome Data Deluge

    DNA Testing, Next Generation Sequencing, and Mitochondrial Disease Diagnosis

    Next Generation Sequencing

    Next Generation Sequencing–Based Diagnostic Assay Design for Mitochondrial Disorders

    Translational Advancements

    Semantic Data Standards for Community Collaboration

    The Road Ahead

    Conclusions

    Chapter 7. The Significance of Metabolomics in Human Health

    Introduction

    Metabolomics in Human Diseases

    Biomarker Detection

    Group Discrimination (Diagnosis)

    Inborn Metabolic Disorders

    Cancer Metabolomics

    Biomarkers for Psychiatric Diseases

    Metabolomics in Environmental and Public Health

    Human Nutrition

    Intestinal Metabolome

    Conclusions

    Chapter 8. Microbial Genomics: Diagnosis, Prevention, and Treatment

    Background

    Control and Prevention

    Treatment and Diagnostics

    Chapter 9. A Metagenomic Insight Into the Human Microbiome: Its Implications in Health and Disease

    Outline of the Chapter

    Introduction

    Human-Associated Microflora

    Alterations in the Healthy Human Microbiome: Association With Diseases

    The Oral Microbiome

    The Skin Microbiome

    The Gut Microbiome

    Novel Therapeutic Strategies Based on the Human-Associated Microbiome

    Conclusion

    Glossary

    List of Acronyms and Abbreviations

    Chapter 10. Pharmacogenetics and Pharmacogenomics

    Introduction

    Pharmacogenomic Information in Drug Labeling

    Pharmacogenetic Study Design

    Conclusions

    Chapter 11. Medical and Health Aspects of Genetics and Genomics

    Introduction

    Chromosomal Disorders

    Mendelian (Single-Gene) Disorders

    Polygenic or Multifactorial Disorders

    Mitochondrial Genetic Disorders

    Genomic Disorders

    Disease Spectrum, Biological Pathways, and Genotypes

    Summary

    Chapter 12. Content and Variation of the Human Genome

    Introduction: The Genome Anatomy

    Special Genomic Structures Containing Selected Repeats

    Chapter 13. Spectrum of Genetic Diseases and Management

    Introduction

    Molecular Approach to Genetic Disease Nosology

    From Marfan Syndrome to Fibrillinopathies

    The Emergence of RASopathies

    Making a Diagnosis of a Genetic Disorder in the Era of Molecular Pathways

    Conclusion

    Chapter 14. Genomic Analysis in Clinical Practice: What Are the Challenges?

    Introduction

    Consent for Genome Testing in Clinical Practice

    Incidental Findings

    The Data Interpretation Problem

    Conclusions

    Chapter 15. Genomic Perspective of Genetic Counseling

    Introduction

    Ethos and Principles of Genetic Counseling

    Elements of Genetic Counseling

    Counseling Issues

    Case Study

    Practicalities of Counseling for Genomic Tests

    Conclusion

    Chapter 16. Genetics and Genomics of Reproductive Medicine and Health

    Introduction

    Female Infertility

    Male Factor Infertility

    Genetic Factors in In Vitro Fertilization

    Recent Advances in Prenatal Diagnosis

    Summary

    Chapter 17. Stratified and Precision Medicine

    Introduction

    Molecular, Genetic, and Genomic Revolutions in Medicine

    Personalized Medicine

    Stratified Medicine

    Summary

    Chapter 18. Teaching and Training Medicine in Genomic Era

    Introduction

    Integration of Genomics into Medical Practice

    Genomics and the Medical Education Landscape

    Approaches to Medical Genomics Education

    Conclusions and Final Comments

    Chapter 19. Genomics, New Drug Development, and Precision Medicines

    Introduction

    Applying Genomics to Drug Discovery

    Applying Pharmacogenetics to Drug Development

    Predicting Safety

    Summary

    Chapter 20. Cancer Genetics and Genomics

    Introduction

    Inherited Cancers: Germ Line and Somatic

    Genetic Testing in Cancer Predisposition Syndromes

    Inherited Cancer Genes

    Genetic Testing and Mainstreaming

    Conclusion

    Chapter 21. The Provision of Medical and Health Genetics and Genomics in the Developing World

    Introduction

    Public Health Programs in Genetics and Genomics

    Medical Services Incorporating Genetics and Genomics

    Summary

    Chapter 22. Genomic Applications in Forensic Medicine

    Introduction

    The Basics of Next Generation Sequencing

    Single Molecule Sequencing

    Next Generation Sequencing Solutions in Forensic Genetics

    Concluding Remarks

    Chapter 23. Public and Population Health Genomics

    Introduction

    Breast Cancer

    Colorectal Cancer

    Crohn Disease

    Alzheimer Dementia

    Cystic Fibrosis

    Cross-Cutting Issues of Population Genomics

    Summary

    Glossary: Medical and Health Genomics

    Index

    Medical and Health Genomics

    "Physician who fails to enter the body of a patient with the lamp of knowledge and understanding can never treat diseases"

    - Charaka, a noted Ayurveda practitioner, wrote the famous treatise Charak Samhita on Ayurvedic medicine in Ancient India c.1000 BC. The ancient Indian text refers to genetic factors determining the sex of the child and the origin of congenital blindness in the sperm or ovum rather than the mother or the father.

    …εἰ γὰρ ἐκ φλεγματώδεος φλεγματώδης, καὶ ἐκ χολώδεος χολώδης γίνεται, καὶ ἐκ φθινώδεος φθινώδης, καὶ ἐκ σπληνώδεος σπληνώδης, τί κωλύει ὅτῳ πατὴρ καὶ μήτηρ εἴχετο, τούτῳ τῷ νοσήματι καὶ τῶν ἐκγόνων ἔχεσθαί τινα; ὡς ὁ γόνος ἔρχεται πάντοθεν τοῦ σώματος, ἀπό τε τῶν ὑγιηρῶν ὑγιηρὸς, ἀπό τε τῶν νοσερῶν νοσερός…

    Περὶ ἱερῆς νούσου

    …For if a phlegmatic person be born of a phlegmatic, and a bilious of a bilious, and a phthisical of a phthisical, and one having spleen disease, of another having disease of the spleen, what is to hinder it from happening that where the father and mother were subject to this disease, certain of their offspring should be so affected also? As the semen comes from all parts of the body, healthy particles will come from healthy parts, and unhealthy from unhealthy parts…

    On the Sacred Disease Hippocrates of Kos (Ἱπποκράτης; c.460–c.370 BC)

    Dedication

    To,

    The Late Shri Anand Swarup Kumar, Our Father and Shrimati Hardevi Kumar, Our Mother.

    Dhavendra, Anju, Ashish, Jaime, Jaya, Nikita and Mayank

    To,

    Grigoria, Emmanuel, Gregory, Alexander, Christina and their spouses

    Stylianos, Athena, Stylianos, Anne-Grigoria, Sophia, Raphael, Elisabeth

    Copyright

    Academic Press is an imprint of Elsevier

    125 London Wall, London EC2Y 5AS, UK

    525 B Street, Suite 1800, San Diego, CA 92101-4495, USA

    50 Hampshire Street, 5th Floor, Cambridge, MA 02139, USA

    The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, UK

    Copyright © 2016 Elsevier Inc. All rights reserved.

    No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

    This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein).

    Notices

    Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary.

    Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.

    To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein.

    British Library Cataloguing-in-Publication Data

    A catalogue record for this book is available from the British Library

    Library of Congress Cataloging-in-Publication Data

    A catalog record for this book is available from the Library of Congress

    ISBN: 978-0-12-420196-5

    For information on all Academic Press publications visit our website at https://www.elsevier.com/

    Publisher: Mica Haley

    Acquisition Editor: Peter Linsley

    Editorial Project Manager: Lisa Eppich

    Production Project Manager: Edward Taylor

    Designer: Matthew Limbert

    Typeset by TNQ Books and Journals

    www.tnq.co.in

    List of Contributors

    A. Alfirevic,     University of Liverpool, Liverpool, United Kingdom

    S.E. Antonarakis

    University of Geneva Medical School, Geneva, Switzerland

    University Hospitals of Geneva, Geneva, Switzerland

    iGE3 Institute of Genetics and Genomics of Geneva, Geneva, Switzerland

    HUGO (Human Genome Organization), Geneva, Switzerland

    A. Bhardwaj,     Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh, India

    P. Borry,     University of Leuven, Leuven, Belgium

    C. Børsting,     Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

    M.V. Busi,     Universidad Nacional de Rosario, Rosario, Argentina

    V.H.W. Dissanayake,     University of Colombo, Colombo, Sri Lanka

    R. Festenstein,     Imperial College, London, United Kingdom

    C.L. Gaff

    Melbourne Genomics Health Alliance, Melbourne, Australia

    The University of Melbourne, Melbourne, Australia

    D.F. Gomez-Casati,     Universidad Nacional de Rosario, Rosario, Argentina

    M. Grisolía,     Universidad Nacional de Rosario, Rosario, Argentina

    P. Gupta

    Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh, India

    Bhaskaracharya College of Applied Sciences, University of Delhi, New Delhi, India

    A. Haworth,     Congenica Ltd., Hinxton, United Kingdom

    B. Kerr,     Manchester Academic Health Sciences Centre (MAHSC), Manchester, United Kingdom

    B.M. Knoppers,     McGill University, Montreal, QC, Canada

    B. Korf,     University of Alabama at Birmingham, Birmingham, AL, United States

    D. Kumar

    The University of South Wales, Pontypridd, Wales, United Kingdom

    Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom

    N. Lench,     Congenica Ltd., Hinxton, United Kingdom

    A. Lucassen

    University of Southampton Medical Centre, Southampton, United Kingdom

    University Medical Centre Groningen and Rijksuniversiteit Groningen, Groningen, The Netherlands

    I. Macciocca,     Victorian Clinical Genetics Service, Melbourne, Australia

    E. Maher,     University of Cambridge, Cambridge, United Kingdom

    T.A. Manolio,     National Institutes of Health (NIH), Bethesda, MD, United States

    D. McHale,     UCB, Braine L’Alleud, Belgium

    N. Morling,     Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

    A. Mutreja

    MSD-Wellcome Trust Hilleman Laboratories, New Delhi, India

    Wellcome Trust Sanger Institute, Cambridge, United Kingdom

    M. Penny,     Biogen, Cambridge, MA, United States

    M. Pirmohamed,     University of Liverpool, Liverpool, United Kingdom

    N.K. Rajput,     Institute of Microbial Technology, Council of Scientific and Industrial Research, Chandigarh, India

    Y.-H. Rogers,     The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States

    H. Savage,     Congenica Ltd., Hinxton, United Kingdom

    R. Saxena,     Indian Institute of Science Education and Research Bhopal, Madhya Pradesh, India

    M. Shabani,     University of Leuven, Leuven, Belgium

    V.K. Sharma,     Indian Institute of Science Education and Research Bhopal, Madhya Pradesh, India

    V. Singh,     Amity University, Noida, India

    N. Sirisena,     University of Colombo, Colombo, Sri Lanka

    D. Sumathipala,     University of Colombo, Colombo, Sri Lanka

    I. van Langen,     University Medical Centre Groningen and Rijksuniversiteit Groningen, Groningen, The Netherlands

    K. Wettasinghe,     University of Colombo, Colombo, Sri Lanka

    J. Whitworth,     University of Cambridge, Cambridge, United Kingdom

    A.L. Wise,     National Institutes of Health (NIH), Bethesda, MD, United States

    C. Zhang,     The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States

    Foreword

    Although inherited diseases in man have been known since biblical times, and the first clear scientific descriptions of genetic disorders by Garrod and others date back to the early years of the 20th century, genetics was only introduced into regular medical practice with the advent of genetic counseling clinics in several countries, about 40 or 50  years ago. The discovery, in 1959, that Down syndrome was caused by a chromosome abnormality was an enormous step toward recognition that laboratory-based genetics had a role to play in medicine. In the early 1970s, few members of the public (or of the health professions) had heard of genetics and it was considered pretty irrelevant to healthcare practice.

    The change since then has been astonishing. Genetics and genomics are now fully on the radar of most practicing physicians, researchers, and health managers, as well as journalists (who enjoy exaggerating the expected rate of progress) and politicians (who are commonly enthusiastic and sometimes surprisingly well informed). There can no longer be any doubt that the inherited components of disease are firmly embedded in medical practice and popular culture. When I say at parties that I am a medical geneticist, people more often look interested than mystified and bored.

    Although it is true that much of this change has been driven by advancing technology, that is an oversimplification. The single biggest event was the sequencing of the human genome. That achievement and the decision of those involved to make their data freely available to all others, in academia and in industry, at an early stage of the research process, were transformative. Its echoes have completely changed not only medical research, but also anthropology, agriculture, and even historical research—witness the use of mitochondrial sequencing to confirm the identity of King Richard III several 100  years after his burial.

    But the technology that we now enjoy was not there when the Human Genome Project started, and arguably would not be there today without that initiative. At the time, the genome project was an act of scientific imagination; a few people with extraordinary vision were able to see, long before most others and against a significant amount of scientific and public opposition, the immense value such sequence information would be. The technology was not confined to sequencing methodologies. The volume of data generated could not have been handled without commensurate improvements in computer hardware and software. Today it is arguable that the real block to understanding genetics and applying it clinically has more to do with our inability to understand and manage the vast amounts of data being generated, than to a need for better and cheaper sequencing.

    Despite this meteoric progress, there is still actually rather little that modern genetics can do in the clinic, outside of defining the molecular basis of rare inherited disorders. But, particularly in cancer studies, that is changing rapidly. It is now very likely indeed that, despite all the previous overoptimistic statements that failed to materialize, genetics will pay big health dividends within a modest period of time. We can see it happening around us, eg, in new targeted cancer therapies. It is very exciting indeed.

    Since the completion of the human genome sequence and rapid advances in genomic diagnostic methods, many avenues for diagnostic, therapeutic, and preventative intervention have emerged with promising medical and health applications. Those working in public and population health are considering many aspects of genomics for improving population health, particularly for the benefit of less developed or developing nations. Many national and international genetic and genomic communities and organizations are now actively engaged in furthering these objectives.

    Against that background, this new book could not be better timed. It reviews many aspects of the field, and tries to help both professionals and interested observers to understand some of the core principles and complexities. Contributions from a distinguished group of authors, led by experienced and professional medical geneticists, provide an excellent resource of information. I am confident it will be of great value.

    Martin Bobrow

    Cambridge, England, August 2015

    Preface

    Genomic Medicine and Healthcare

    In any sociocultural and geographic setting the provision of medical and healthcare are dependent upon several factors, including societal, financial, and political denominators. However, in the background of all these factors, the fact remains that peoples for whom any medical and healthcare system is aimed for are fundamentally different. The physical and psychological variation of the majority and minority population groups in any society or country determine the outcomes of medical and healthcare provision, however basic or sophisticated. Assessing human variation solely for the purposes of assessing the outcomes of medicine and healthcare practices would require many different approaches. Many such approaches include conventional anthropological and sociocultural variables. Since the discovery of the ABO, Rhesus, and other blood group systems in the early 20th century, the human variation has been assessed using a number of different biomarkers. However, despite many years’ work and enormous data, a meaningful and scientific plausible correlation has not been possible. Following the Human Genome Project and the subsequent sequencing of thousands of individual human genomes, and the discovery of the extensive genomic variation and individuality, a major paradigm shift is taking place with considerable impact on the nature and pattern of medicine and healthcare. The people and the society at large have high expectations from the genomic-led contemporary and future medical and healthcare practices. There are huge expectations and hype surrounding the idea of genomic medicine (also known as personalized/precision medicine), which is based on the individual genomic variation. We are probably not fully prepared for this; there is a lot more to be learned and accomplished before the society and medical/health professions could offer genomic healthcare with robust scientific confidence while making this efficient and cost-effective.

    Genetic and genomic variation among peoples and population groups are one of the many variables that influence the outcomes of any medical and healthcare practice. This needs to be linked with the specific gene–molecule systems that operate at the cell and tissue levels. Following the successes of clinical genetics, along with related genetic laboratory techniques, a number of specific genetic diseases (chromosomal, single gene, and rare genetic syndromes) are now causally linked to pathogenic changes in many genes and molecules that operate in conjunction with many other biological systems. Some of these rare conditions result from specific mutations or pathogenic sequence variation within one particular gene or loss of function of other genes that belong to a multigene family encoding many peptides with overlapping structural similarity and physiological functions. Examples include transcription factors, nuclear envelope genes, RAS-MAPK genes, TBX genes, genes for many inherited metabolic disorders, and a large number of gene–molecule families for sophisticated neuronal functions. Equipped with the knowledge from genetic and molecular advances in uncommon and extremely rare diseases, researchers and clinicians are now looking into solving the molecular complexities of common medical diseases with a considerable heritability. The scientific information from studying specific system and multisystem rare genetic disorders has given us insight to many fundamental molecular biological processes that are not only important for understanding the pathogenesis but also govern the outcomes of specific therapeutic interventions. Thus successful outcomes of genetic and genomic applications in medicine and healthcare practices would be dependent upon understanding the molecular biology of sequential diseases processes and their overall clinical impact governed by individual genomic variation. This is in essence the basis of medical and health genomics, rapidly emerging along with the practice of specific genetic and genomic medicine.

    In keeping with the many dilemmas and predicaments surrounding the genetic and genomic applications in medicine and healthcare, this new book sets out to collate basic facts and information that could form the core of genomic (personalized/precision) medicine and genomic healthcare. A number of scientists, clinicians and healthcare professionals have contributed to this high profile work under the broad meaning title of Medical and Health Genomics.

    The core concepts of human genomics are presented with emphasis on new emerging genomic technologies, burgeoning genomic databases with enormous amount classified and unclassified data, genomic applications, and translations in clinical medicine and public health, particularly the diagnostic genomics and clinical bioinformatics contributing to deciphering many complex phenotypes, citing the specific example of the model of Victor McKusick’s Online Mendelian Inheritance in Man (OMIM).

    Some additional areas of particular importance are highlighted, including epigenetics modifications in human health and disease; metabolomics with its many applications; introduction to metagenomics and ecogenomics; wide-ranging applications of microbial genomics in diagnosis, treatment, and prevention of microbial diseases; and the personalized pharmacotherapy based on the genetic variation of each individual (pharmacogenomics).

    Medical and health aspects of genetics and genomics are discussed in detail with evidence on the relevance of genome variation in human health and disease. This is further supported by critical information on multidisciplinary medical management using examples of systemic rare genetic diseases, genetic and genomic testing and screening, and genomic perspectives of genetic counseling. The section on personalized and stratified medicine includes information on novel genomics-led drug discovery and development and discussion on novel genomics-led therapeutic approaches. The organization, availability, and harmonization of genetic and genomic healthcare are reviewed with emphasis on medical and health burden of rare genetic diseases, multidisciplinary genetics and genomics-led reproductive healthcare, and common and complex genetic cancer. This section includes a separate chapter on teaching and training genetics/genomics for medical and healthcare professionals. The detailed glossary lists a number of key phrases, definitions, abbreviations, and acronyms that the reader might find useful.

    Editors and contributors hope that the book will convey the core concepts of medical and health genomics, highlighting the specialist genomic fields, emphasis on delivery of genetic/genomic medicine and healthcare, drawing attention to specific issues and requirements of human genomics in developing countries and emerging perspectives of human and medical genomics in the context of public and population health.

    The society and many people have high hopes and expectations from many recent new developments and progress in human (medical) genomics in the diagnosis and management of both rare and common medical and health problems. These sentiments were echoed by Mr. Anthony Charles Lynton (Tony) Blair, the British Prime Minister, on the occasion of the White House announcement of the completion of the First Survey of the Entire Human Genome Project, broadcast (jointly with the United States President Bill Clinton) on the day of the publication of the first draft of the human genome, Ever so often in the history of human endeavor, there comes a breakthrough that takes humankind across a frontier into a new era. ... today’s announcement is such a breakthrough, a breakthrough that opens the way for massive advancement in the treatment of cancer and hereditary diseases. And that is only the beginning.

    We are delighted to present this book at a critical historical phase of genome science, with the new global wave to incorporate strengths and unlimited potential of genomics for much needed advances in healthcare. This is reflected in President Obama’s State of the Union address on 30 January, 2015, launching the new United States Precision Medicine initiative, "To enable a new era of medicine through research, technology, and policies that empower patients, researchers, and providers to work together toward development of individualized treatments."

    Dhavendra Kumar, Cardiff, Wales, UK

    Stylianos Antonarakis, Geneva, Switzerland

    Editors

    December 2015

    Chapter 1

    The Human Genome

    D. Kumar¹,²     ¹The University of South Wales, Pontypridd, Wales, United Kingdom     ²Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom

    Abstract

    New discoveries and innovations in biological and life sciences during the five decades before the 21st century have centered on genetics and genomics. It took just over 50  years after the unraveling of the structure of the molecule of nucleic acids, the key unit of the biological life, for scientists to embark on sequencing the major entire genetic constitution or genome of many single-cell and large mammalian creatures including man. The word genome includes gene and -ome, implying complete knowledge of all genes and related elements in any single organism. Inevitably, this led to enthusiastic expansion of the whole science and thence to the emergence of genomics. The suffix -omic, derived from the ancient Greek, refers to in-depth knowledge. Not surprisingly, genomics was followed by a plethora of related -omics; for example, proteomics, metabolomics, transcriptomics, and so on. Currently, we have over 30 such disciplines with the -omics suffix. Developments and advances in genetics have led to a better understanding of genomic variation, the principles governing heredity and the familial transmission of physical characteristics and diseases, in-depth understanding of the pathophysiology of diseases, the development of new methods of clinical and laboratory diagnosis, and innovative approaches to making early diagnoses (eg, prenatal diagnoses and newborn screening) and offering reproductive choices, including preimplantation genetic diagnoses. All these developments are now accepted within the broad fields of human genetics, medical genetics, clinical genetics, genetic medicine, and the new emerging field of genomic medicine. Not surprisingly, the field remains wide open, encompassing the massive field of human genomics, broadly focusing on medical and health genomics. This chapter leads the book, providing the basic factual information for grasping the concepts of human heredity, genes, genetics, and genomics. It is expected that the reader will proceed to subsequent chapters better equipped with the introduction to genetic/genome sciences as applied to humans, specifically genetic diseases, genetics, and genomics in medicine; public health; and specific issues related to society, ethics, and law.

    Keywords

    Bioinformatics; DNA; Gene; Genome; Genomic variation; Genomics; Heredity; Mitochondrial genome; Nuclear genome; Nucleic acids; Proteomics; RNA; Transcriptomics

    Chapter Outline

    Introduction 1

    Hereditary Factors, Genes, Genetics, and Genomics 1

    Structure and Organization of Nucleic Acids 2

    Human Genome Variation and Human Disease 5

    Measuring Genetic and Genomic Variation 6

    Genome Variation and Human Disease 7

    The Mitochondrial Genome 8

    Functional Genomics, Transcriptomics, and Proteomics 9

    Translational Human Genomics 10

    Human Genomics for Socioeconomic Development 11

    Conclusions 12

    References 12

    Introduction

    Toward the end of the last millennium, tremendous growth in the sophistication of the biological sciences was harnessed in medicine, the food industry, and related bioindustries. New discoveries and innovations in biological sciences during the five decades leading up to the 21st century have centered on genetics and genomics. It took just over 50  years after the unraveling of the structure of the molecule of nucleic acids, the key unit of the biological life, for scientists to embark on sequencing of major organisms’ entire genetic constitution or genome. The word genome includes gene and -ome, implying complete knowledge of all genes and related elements in any single organism. Inevitably, this led to enthusiastic expansion of the whole science and thence to the emergence of genomics [1]. The suffix -omic, derived from the ancient Greek, refers to in-depth knowledge. Not surprisingly, genomics was followed by a plethora of related -omics; for example, proteomics, metabolomics, transcriptomics, and so on [2]. Currently, we have over 30 such disciplines with the -omics suffix.

    The ultimate goal of any scientific discipline is its translation for the benefit of all humans, crossing all possible barriers and boundaries. Major advances in medicine and health were only possible through understanding basic principles and mechanisms underlying disease processes. This was facilitated by rapid applications of physical and chemical sciences in medicine and health; for example, radiographical diagnosis, ultrasound diagnosis, microbiology diagnosis, immunohistochemical diagnosis, and finally, molecular diagnosis. Developments and advances in genetics have led to a better understanding of the principles governing heredity and the familial transmission of physical characteristics and diseases, better understanding of the pathophysiology of diseases, the development of new methods of clinical and laboratory diagnosis, and innovative approaches to making early diagnoses (eg, prenatal diagnoses and newborn screening) and offering reproductive choices, including preimplantation genetic diagnoses. All these developments are now accepted within the broad fields of human genetics, medical genetics, clinical genetics, genetic medicine, and the new emerging field of genomic medicine. Not surprisingly, the field remains wide open, encompassing the massive field of human genomics, broadly focusing on medical and health genomics [3].

    This chapter leads the book, providing the basic factual information for grasping the concepts of heredity, genes, genetics, and genomics. It is expected that the reader will proceed to subsequent chapters better equipped with the introduction to genetic/genome sciences, genetic diseases, genetics and genomics in medicine, applications in public health, and specific issues related to society, ethics, and law [4].

    Hereditary Factors, Genes, Genetics, and Genomics

    The concepts of heredity and hereditary factors date back several hundred and probably even thousands of years. The popular darwinian theory of natural selection rests on the core concept of the transmission of hereditary factors[5]. For several thousand years, various descriptions and explanations have been put forward to define the physical shape and functional nature of hereditary factors. In the historical context, the concept of the gene was introduced only recently as the most acceptable answer to explain one of the hereditary factors. However, it remains unclear when and by whom this term was first introduced. It does not matter, as the term gene (from the Greek genos, race) is now universally accepted and used in the context of understanding heredity and hereditary factors, and is probably the single most important biological factor regulating biological life, ranging from single-cell organisms to multicellular mammals. Rapid and extraordinary scientific progress made during the 19th and 20th centuries has led to the development of genetics, the science of heredity. This has now been transformed into the broader field of genomics that includes all genes with all possible heritable biologically active or inactive regulatory and evolutionary genetic elements, whether recent or extending back through several thousand years of life on our planet.

    In biological terms, genes, genetics, and genomics are keys to procreation, development, growth, function, and survival. The health of any living organism is judged by its physical and functional existence. Thus genes, genetics, and genomics are central to all forms of biological health, including that of humans. Human health depends not only on its own genetic or genomic constitution, but on that of other organisms whose well-being is also essential to human health—for example, food (plants, fish, and animals), shelter (homes made of wood from trees), the environment (water, trees, and plants), protection (clothes from cotton and animal skin), and transportation (animals and vehicles made of wood from trees). From a medical perspective, the science of genetics or genomics offers deep insight into and evidence for a number of human diseases, including infectious diseases resulting from either lack of protection and/or failure in controlling the spread of microbial infections or parasitic infestations. This chapter introduces the reader to some of the basic facts about genes, genetics, and genomics, and discusses how these impact human health and that of the plants, crops, and animals necessary for human health and survival. This is obviously more relevant to millions of people in the developing and less-developed countries, where limited resources and lack of infrastructure limit the optimal use of the science of genetics and genomics in applications to eradicate poverty and ensure optimal health. The reader will find cross-references to separate chapters in the book containing detailed information and further discussion of each subject.

    A detailed description of the basic principles of genetics and human genetic diseases is beyond the scope of this chapter. Some of these facts are explained in subsequent chapters and various other information resources on basic genetics and medical genetics. However, some basic principles and relevant information are outlined in this section to assist the reader with limited understanding of basic genetics.

    Structure and Organization of Nucleic Acids

    Living organisms are divided into two large classes—the eukaryotes and prokaryotes. The cells of the eukaryotes have a complex compartmentalized internal structure, the nucleus; these include algae, fungi, plants, and animals. Prokaryotes, on the other hand, are single-celled microorganisms without any specific part harboring the genetic material or genome; examples include bacteria and other related microorganisms. The other types of living organisms are viruses, which are intracellular obligate parasites living in both eukaryotes and prokaryotes, and are composed of short dispersed nucleic acid [deoxyribonucleic acid (DNA) or ribonucleic acid (RNA)] sequences.

    Genetic information is transferred from one generation to the next by small sections of the nucleic acid, DNA, which is tightly packaged into subcellular structures called chromosomes. Prokaryotes usually have a single circular chromosome, while most eukaryotes have more than two, and in some cases up to several hundred. In humans, there are 46 chromosomes arranged in 23 pairs, with one of each pair inherited from each parent (Fig. 1.1A and B). Twenty-two pairs are called autosomes, and one pair is called sex chromosomes, designated as X and Y; females have two X chromosomes (46, XX) and males have an X and a Y (46, XY).

    A chromosome consists of a tightly coiled length of DNA and the proteins (eg, chromatins) that help define its structure and level of activity. DNA consists of two long strands of nucleotide bases wrapped round each other along a central spine made up of phosphate and sugar (Fig. 1.2). There are four bases: adenine (A), guanine (G), cytosine (C), and thymine (T). Pairing of these bases follows strict rules: A always pairs with T, and C with G. Two strands are therefore complementary to each other.

    Genes are made up of specific lengths of DNA that encode the information to make a protein, or RNA product. RNA differs from DNA in that the base thymine (T) is replaced by uracil (U), and the sugar is ribose. It acts as a template to take the coded information across to ribosomes for final assembly of amino acids into the protein peptide chain (Fig. 1.3). The bases are arranged in sets of three, referred to as codons. Each codon codes for a specific amino acid; hence the term genetic code. Codons are located in exons, which contain the coding sequences. A gene may consist of several such coding DNA segments. Exons are separated from each other by noncoding sequences of DNA, called introns. Although they are not yet known to be associated with any specific function, it is likely that some of these introns might be of evolutionary significance or associated with other fundamental biological functions. During the transcription of DNA, the introns are spliced out, and the exons then attach to messenger RNA (mRNA) to start the process of protein synthesis.

    Figure 1.1  Human chromosomes. (A) Diploid set in a male (46, XY). (B) Complete set of human chromosomes map.

    Figure 1.2  The Watson-Crick model of the double helix structure of the nucleic acid molecule Adopted with permission from Turnpenny P, Ellar S, editors. Emery’s elements of medical genetics. 14th ed. Edinburgh: Elsevier Churchill Livingstone; 2012.

    Figure 1.3  The synthesis of the peptide chain from the coding sequences in the exon. mRNA , messenger RNA; CAP , adenyl cyclase associated protein; IVS , intervening sequence (Turnpenny and Ellard, 2011).

    Proteins are one of the major constituents of the body’s chemistry. These are remarkably variable in their structure, ranging from tough collagen that forms connective tissue and bone, through the fluid hemoglobin that transports oxygen, to thousands of enzymes, hormones, and other biological effectors and their receptors that are essential for the structures and functions of the body. Each protein is made up of one or more peptide chains consisting of series of amino acids, of which only 20 occur in living organisms. The different structures and functions of proteins depend on the order of amino acids as determined by the genetic code.

    DNA has the remarkable property of self-replication. The two strands of a DNA molecule separate as chromosomes divide during cell division. There are two types of cell division; mitosis in all body cells, and meiosis, which is specifically confined to the gonads in making sperm and eggs (Fig. 1.4). During mitosis, no reduction of the number of chromosomes takes place (diploid, or 2n), while meiosis results in half the number of chromosomes (haploid, or 1n). The new pairs of DNA are identical to those from which they were synthesized. However, sometimes mistakes or mutations occur. These usually result from substitution of a different base, or are caused by extensive structural changes to genes. In other words, any spelling mistake in the letters A–T or C–G could result in either absence of coded information (nonsense mutation) or a different message (missense mutation). However, not all mutations or spelling mistakes have an adverse effect (neutral mutations). Conversely, some changes in the genes might result in a favorable property; for example, resistance to disease or other environmental hazard. This is the basis for the gradual changes in species over millions of years of evolution. On the other hand, mutations may result in defective gene functions, leading to a disease or susceptibility to a disease as a result of qualitative or quantitative changes in the gene product, the peptide chain. However, these changes may also result from epigenetic mechanisms, abnormal RNA molecules, and posttranslational modifications (see Glossary). A brief introduction to these molecular processes is provided elsewhere in this chapter; interested readers are advised to consult dedicated texts on cell and molecular biology.

    Studies on human genomic variations in different population groups and the resemblance of several genome sequences to other genomes (comparative genomics) have offered wide-ranging evidence to support the followers of Charles Darwin. Apart from reproduction, genes, gene-sequence variation, genomic variation, and epigenetic factors are important in growth, development, aging, and senescence. Some of these may be evolutionarily conserved across species, but relevant to human health. Mutations and alterations in several of these genomic elements are linked to a broad range of medical conditions.

    Figure 1.4  Steps in mitosis and meiosis during a eukaryotic cell division; note (bottom) exchange of the genetic material (recombination) through homologous pairing (Turnpenny and Ellard, 2011).

    Human Genome Variation and Human Disease

    The advent of recombinant DNA technology in the 1970s revolutionized our ability to characterize and capitalize on the molecular basis of human genetic disease. This laid the foundation of eventually mapping and deciphering the DNA sequence of all the structural and functional genes of the human genome. The Human Genome Project (HGP) was therefore a natural progression from all previous developments in the field of human genetics. Such a mammoth task could not have been accomplished without the international collective efforts supported by generous funding from governmental and nongovernmental sources [6].

    The project (HGP) has helped map and provide nucleotide sequences of around 23,000 nuclear genes, which, along with a number of other sequence variations, compose the whole human genome. Although a large number of the nuclear genes have been assigned with a structural or functional link, the precise roles of other parts of the genome are not yet fully understood. However, HGP provides the basis for functional genomics to explore further the genome’s functional role and understand the complex mechanisms through which genes and their products interact to affect biological function and influence disease processes. The development of new therapeutic agents is now possible on the basis of genomic arrangement and its designated functional role. This approach also helps characterize the genomes of various pathogens and other organisms, an invaluable tool in realizing the full potential of this field to improve human health [7].

    Table 1.1

    DNA and Gene Content of the Human Reference Genome (GRCh37, February 2009)

    The overall totals are derived from a slightly different analysis from the individual chromosome totals, so the figures do not exactly add up.

    Data from Ensembl Release 66, March 20, 2012.

    Measuring Genetic and Genomic Variation

    Humans have two genomes: nuclear and mitochondrial. Normal diploid cells contain two copies of the nuclear genome and a much larger but variable number of copies of the mitochondrial genome. The nuclear genome is approximately 2  ×  10⁵ times larger than the mitochondrial genome (3  ×  10⁹ vs 16,569  bp), and contains more than 1500 times the number of protein-coding genes (approximately 21,000 vs 13), including many required for mitochondrial functions. Genetic and genomic variation is abundant in both genomes. However, in general this implies to nuclear genome.

    The finished human genome sequence was published in 2004 [7]. Table 1.1 shows the current best estimates of the size and gene content of each chromosome. These figures are for the human reference genome. There are striking differences between the nuclear and mitochondrial DNA (Table 1.2). They do not correspond precisely to the genome of any actual individual, because the genomes of healthy normal individuals vary somewhat in chromosome sizes and numbers of genes, as described below. Nor is the reference genome in any sense an ideal human genome. It is simply an arbitrary and reasonably typical reference point for comparing human genome sequences. Uncertainties in the figures relate primarily to the highly repetitive DNA of centromeres and telomeres and to the number of RNA genes, which are difficult to identify from sequence data.

    Table 1.2

    Comparison of the Human Nuclear and Mitochondrial Genomes

    The most direct way to measure genetic differences, or genetic variation, is to estimate how often two individuals differ at a specific site in their DNA sequences (that is, whether they have a different nucleotide base pair at a specific location in their DNA). First, DNA sequences are obtained from a sample of individuals. The sequences of all possible pairs of individuals are then compared to see how often each nucleotide differs. When this is done for a sample of humans, the result is that individuals differ, on average, at only about one in 1300 DNA base pairs. In other words, any two humans are about 99.9% identical in terms of their DNA sequences.

    During the past several years, a new type of genetic variation has been studied extensively in humans: copy-number variants (CNVs), comprising of DNA sequences of 1000 base pairs or larger, fairly distributed across the genome [8]. In some instances, CNVs could be deleted, duplicated, or inverted in some individuals with mild phenotypic effects. Several thousand CNVs have been discovered in humans, indicating that at least 4 million nucleotides of the human genome (and perhaps several times more) vary in copy number among individuals. CNVs thus are another important class of genetic variation and contribute to at least an additional 0.1% difference, on average, between individuals. Despite significant progress, the medical and health implications of CNVs are not entirely clear [9].

    Comparisons of DNA sequences can be done for pairs of individuals from the same population or for pairs of individuals from different populations. Populations can be defined in various ways; one common way is to group individuals into populations according to the continent of origin. Using this definition, individuals from different populations have roughly 10–15% more sequence differences than do individuals from the same population [this estimate is approximately the same for both single nucleotide polymorphisms (SNPs) and CNVs]. In other words, people from different populations are slightly more different at the DNA level than are people from the same population. The slightness of this difference supports the conclusion that all humans are genetically quite similar to one another, irrespective of their geographic ancestry [10].

    Because it is still fairly expensive to assess DNA sequences on a large scale, investigators often study genetic variations at specific sites that are known to vary among individuals. Suppose that a specific site in the DNA sequence harbors an A in some individuals’ DNA sequences and a G in others’. This is an SNP, where polymorphism refers to a genetic site that exists in multiple forms. The proportion of individuals who have an A and the proportion with a G give the frequency of each form, or allele, and this frequency can be estimated for a sample of individuals from a population. If the frequencies of A in three different populations are 0.10, 0.20, and 0.50, the genetic distance between the first two populations is smaller than that between the third population and the first two. On the basis of this assessment, the first two populations are genetically more similar than either is to the third. To get a more accurate picture of genetic differences, hundreds or thousands of SNP frequencies would be assessed to yield the average genetic difference among pairs of populations [11].

    Genome Variation and Human Disease

    A number of genes have direct or indirect influence on most human diseases. Because individuals have different variants of genes, it follows that the risk of developing various diseases will also differ among individuals. Consider a simple example: Jim Fixx, a well-known runner and fitness enthusiast, died of a heart attack at the age of 52. Sir Winston Churchill, who was renowned for his abhorrence of exercise and his love of food, drink, and tobacco, lived to the age of 90. It is plausible that genetic differences between Fixx and Churchill were responsible, at least in part, for the paradoxical difference in their life spans. (Indeed, Jim Fixx’s father had a heart attack at the age of 35 and died of a second heart attack at the age of 43.)

    Because genes are passed down from parents to offspring, diseases tend to cluster in families. For example, if an individual has had a heart attack, the risk that his or her close relatives, offspring, or siblings will have a heart attack is two to three times higher than that of the general population. Similar levels of increased risk among family members are seen for colon cancer, breast cancer, prostate cancer, type 2 diabetes mellitus, and many other diseases. This clustering in families is partly the result of shared nongenetic factors (eg, families tend to be similar in terms of their dietary and exercise habits) and partly the result of shared genes. As we have seen, populations differ somewhat in their genetic backgrounds. It is thus possible that genetic differences could be partly responsible for differences in disease prevalence. For many disorders caused by genetic changes in single genes, these differences are readily apparent. Cystic fibrosis, for example, is seen in about one in 2500 Europeans, but only in one in 90,000 Asians. Sickle cell disease is much more common in individuals of African and Mediterranean descent than in others, although it is found at lower rates in many other populations because of migration and intermarriage.

    These differences in prevalence can be attributed to the evolutionary factors that influence genetic variation in general. Mutation is the ultimate source of all genetic variation. In some cases, such as hemochromatosis in Europeans and sickle cell disease in Africans, the responsible mutations have arisen within the last few thousand years, helping to account for a fairly restricted distribution of the disease. Natural selection also plays a role in population differences in some genetic diseases. For sickle cell disease and related diseases known as the thalassemias, heterozygotes (those who carry a single copy of a disease-causing mutation) are relatively resistant to the malaria parasite. Cystic fibrosis heterozygotes are resistant to typhoid fever, and hemochromatosis heterozygotes absorb iron more readily, perhaps protecting them against anemia. Also, the process of genetic drift, which is accentuated in small populations, can raise the incidence of disease-causing mutations quickly just by chance (eg, Ellis-van Creveld syndrome, a reduced-stature disorder, is unusually common among the Old Order Amish of Pennsylvania) [12]. In contrast to the effects of natural selection and genetic drift, which tend to promote population differences in disease prevalence, gene flow (the exchange of DNA among populations) tends to decrease differences among populations. With the enhanced mobility of populations worldwide, gene flow is thought to be increasing steadily.

    These same factors can affect common diseases such as cancer, diabetes mellitus, hypertension, and heart disease, but the picture is more complex, because these diseases are influenced by multiple genetic and nongenetic factors. Common diseases do vary in incidence among populations: hypertension occurs more often in African Americans than European Americans, and type 2 diabetes mellitus is especially common among Hispanic and Native American populations [13]. Although genes clearly play a role in causing common diseases, it is less clear that genetic differences between populations play a significant role in causing differences in prevalence rates among populations. Consider another example: the Pima Native American population in the southwestern United States now has one of the highest known rates of type 2 diabetes mellitus in the world. About half of adult Pimas are affected. Yet this disease was virtually unknown in this population before World War II. Obviously, the Pimas’ genes have not changed much since the middle of the 20th century. Their environment, however, has changed dramatically with the adoption of a Western high-calorie, high-fat diet, and a decrease in physical exercise. In this case, it is almost certain that the rapid increase in type 2 diabetes mellitus prevalence has much more to do with nongenetic than genetic causes [14].

    But why does a Western diet seem to have a greater effect on some populations than others? Perhaps differences in genetic background, interacting with dietary and other lifestyle changes, help account for this variation. As additional genes that influence susceptibility to common diseases are discovered, and as the roles of nongenetic factors are also taken into account, it is likely that this picture will become clearer.

    The Mitochondrial Genome

    The mitochondrial genome is very different from the nuclear genome (Fig. 1.5; Table 1.2). In many respects, it has more in common with bacterial genomes than the eukaryotic nuclear genome. This is consistent with the idea that mitochondria originated as endosymbiotic bacteria within some ancestral eukaryotic cell. If this theory is correct, then over the years, the mitochondria have gradually transferred more and more of their functions to the nucleus. This is evident from the fact that a number of nuclear genes encode the great majority of mitochondrial proteins. Cells contain many mitochondria (typically 100–1000; maybe 100,000 in an oocyte), so mitochondrial DNA (mtDNA) might be formally classified among the repetitive DNA in a cell. Although the mitochondrial genome is very small compared with its nuclear counterpart, because there are many copies, mtDNA often makes up 1% or so of total cellular DNA.

    Figure 1.5  The -omics paradigm, showing four major branches. iRNA , Informational RNA; LC-MS , liquid chromatography–mass spectrometry.

    As in bacteria, the mitochondrial genome is circular and closely packed with genes. There are no introns and little intergenic noncoding DNA. Some genes even overlap. In the nuclear genome, it is not uncommon for genes on opposite strands to overlap. However, in this case, genes on the same strand overlap, using the same template but read in different reading frames. Twenty-four of the 37 genes specify functional RNAs [two ribosomal RNAs and 22 transfer (tRNAs)]; the other 13 genes encode components of the electron transport pathway.

    A short segment of the mitochondrial genome is triple-stranded, on which the displacement loop (D-loop) is noncoding produced by replication forks overlapping as they travel in opposite directions around the circular DNA. The D-loop contains the only significant amount of noncoding DNA in the mitochondrial genome. Perhaps because of this, it is the location of many of the DNA polymorphisms that are such useful tools for anthropologists researching the origins of human populations. Because there is no recombination among mtDNA, complete haplotypes of polymorphisms are transmitted through the generations, modified only by recurrent mutation. This makes mtDNA a highly informative marker of ancestry, at least along the maternal line.

    Mitochondrial DNA replication and transcription use nuclear-encoded polymerases. Transcription proceeds in both directions round the circle. The initial products are two large multicistronic RNAs, which are subsequently cleaved to make the individual mtRNAs. All the protein components of the translation machinery are nuclear-encoded, but mitochondria exclusively encode the tRNAs, and these use a coding scheme slightly different from the otherwise universal code. There are four stop codons: UAG, UAA, AGG, and AGA. UGA encodes tryptophan, and AUA specifies isoleucine, rather than arginine as normally. Presumably, with only 13 protein-coding genes, the mitochondrial system could tolerate mutations that modified the coding scheme in a way the main genome could not.

    Mutations in mtDNA are important causes of disease, and perhaps also of aging [15]. Phenotypes caused by variation in mtDNA are transmitted exclusively down the maternal line (matrilineal inheritance), but most genetic diseases where there is mitochondrial dysfunction are caused by mutations in nuclear-encoded genes, and so follow normal mendelian patterns. As cells contain many copies of the mitochondrial genome, they can be heteroplasmic, containing a mix of different sequences. Unlike mosaicism for nuclear variants, heteroplasmy can be transmitted by a mother to her children.

    Functional Genomics, Transcriptomics, and Proteomics

    Functional genomics, specifically transcriptomics, is a systematic effort to understand the function of genes and gene products by high-throughput analysis of gene transcripts in a biological system (cell, tissue, or organism) with the use of automated procedures that allow scale-up of experiments classically performed with single genes [15]. Functional genomics can be conceptually divided into gene-driven and phenotype-driven approaches. Gene-driven approaches rely on genomic information to identify, clone, and express genes, as well as to characterize them at the molecular level. Phenotype-driven approaches rely on phenotypes, either identified from random mutation screens or associated with naturally occurring gene variants, such as those responsible for mouse mutants or human diseases, to identify and clone the responsible genes without prior knowledge of the underlying molecular mechanisms [16]. The tools of functional genomics have enabled the development of systematic approaches to obtaining basic information for most genes in a genome, including when and where a gene is expressed and what phenotype results if it is mutated, as well as the identification of the gene product and the identity of other proteins with which it interacts. Functional genomics aspires to answer such questions systematically for all genes in a genome, in contrast to conventional approaches that address one gene at a time.

    Within the context of functional genomics, an important part of functional arrangement of all genomes consists of areas that are external to any coding gene sequences. These play a crucial role in gene expression. Collectively this is known as epigenome. Studies in epigenetics and epigenomics have established core principles that are responsible for modulating development and differentiation and respond to external changes. Patterns of cell- and tissue-specific gene expression are established and maintained by the patterns of epigenetic marks on the genome. These consist of DNA methylation and a variety of specific covalent modifications of histones. The epigenetic marks or signatures are established by a large series of writers: DNA methyltransferases, histone methyltransferases and demethylases, histone acetyltransferases and deacetylases, histone kinases and phosphatases, and so on. In some cases, small RNA molecules help ensure sequence specificity. The effects on gene expression are mediated by epigenetic readers, including methylated DNA-binding proteins, chromodomain and bromodomain proteins that bind methylated and acetylated histones respectively, and a large number of other proteins [17].

    As a result of epigenetic modifications, chromatin exists in a variety of epigenetic flavors. The basic distinction is between heterochromatin (inactive, repressed) and euchromatin (potentially active), but subtypes define transcriptional activity and regulatory elements such as promoters, enhancers, and insulators. The flavor depends on a combination of types and relative quantities of marks rather than a simple histone code.

    Central to functional genomics is the complex organization of RNA molecules that occupies bulk of the intergenic parts of the genome. Apart from well characterized coding RNA (cRNAs), messenger RNA (mRNA), ribosomal RNA (rRNA), and transfer RNA (tRNA), there are many other noncoding RNAs (ncRNAs) [18]. These can be divided into classical ncRNAs and long intergenic ncRNAs (lincRNAs). The classical ncRNAs are small molecules, typically 16–30  nt, derived by processing much longer precursors. There has been an explosion in our knowledge of the numbers and classes of these molecules, and this is still a very active research area. The main well-established classes are:

    • Small nuclear RNAs (snRNAs) form part of the spliceosomal machinery.

    • Small nucleolar RNAs (snoRNAs) act as sequence-specific guides for enzymes that chemically modify specific bases in ribosomal and other RNAs.

    • MicroRNAs (miRNAs) control translation of many mRNAs by binding to sequences in the 3′ untranslated region.

    • Piwi-associated RNAs (piRNAs) act in gametes to ensure stability of the genome. There appear to be many thousands of piRNA genes, grouped in around 100 clusters.

    Table 1.3 lists the numbers of genes encoding these molecules, but these are subject to major revision because it is very difficult to identify functional ncRNAs and distinguish them from the large number of nonfunctional variants present in the genome [19].

    Analysis and application of the rapid accumulation of highly sophisticated genome and proteome data necessitated development of powerful computational programs and relevant hardware tools. Storage, retrieval, and assimilation of enormous amounts of data require fast and accurate computational skills. Bioinformatics deals with these requirements within the broad biomedical and biotechnology sectors. There are several literature and online resources with detailed descriptions of the role and scope of bioinformatics [20].

    Table 1.3

    RNA Genes in the Human Genome

    miRNAblog.com, piRNAbank.ilab.ac.in.

    A number of biomedical and biotechnology disciplines have emerged during the last two decades, all ending with the suffix -omics. -Omics is derived from ome (Greek, omoyous), which refers to complete knowledge. The ancient language Sanskrit has a similar word, ohm, with similar meaning and expression. A number of these -omics have direct or indirect links to the fundamentals of genome science and technology. A number of biological models have been developed and tested using genomic, transcriptomic, proteomic, and metabolomic approaches (Fig. 1.5). Systems biology refers to developing and testing biological models based on -omic sciences [21]. The central dogma of the systems biology is the computational analysis of complex and enormous data at all biological levels: gene, molecule, cell, tissue, organ, and whole body.

    Translational Human Genomics

    The potential of application of genome science and technology in medicine and health has led to the emergence of genomic medicine, a natural outcome of the tremendous progress made in medical genetics and genomics [22]. However, final endpoints in genomic medicine will largely depend upon judicious and efficacious application and utilization of the diagnostic and therapeutic potential of genome-based technologies; for example, clinical applications of microarray technology. This process requires multifaceted systematic and analytical research efforts to translate the basic scientific information into practical and pragmatic applications following the principles of good medical practice (Fig. 1.6). There is no disagreement that this translational genome research is vital for the successful and efficient delivery of promises made by researchers and physicians behind the genomic medicine movement.

    The process for translational genome research includes the participation of several researchers drawn from different disciplines. The multidisciplinary model for translational genome research is widely accepted, and includes several key elements. Informatics and computational networks remain the core element for translational genomics research and systems biology [23]. A framework for the continuum of multidisciplinary translation research is recommended to utilize previous research outcomes in genomics and related areas of health and prevention [24].

    Enjoying the preview?
    Page 1 of 1