Molecular Diagnostics George Patrinos

Molecular Diagnostics George Patrinos PDF

By George Patrinos, Wilhelm Ansorge and Phillip B. Danielson

Molecular Diagnostics 3rd Edition PDF focuses on the technologies and applications that professionals need to work in, develop, and manage a clinical diagnostic laboratory. Each chapter contains an expert introduction to each subject that is next to technical details and many applications for molecular genetic testing that can be found in comprehensive reference lists at the end of each chapter. Contents are divided into three parts, technologies, application of those technologies, and related issues. The first part is dedicated to the battery of the most widely used molecular pathology techniques. New chapters have been added, including the various new technologies involved in next-generation sequencing (mutation detection, gene expression, etc.), mass spectrometry, and protein-specific methodologies. All revised chapters have been completely updated, to include not only technology innovations, but also novel diagnostic applications. As with previous editions, each of the chapters in this section includes a brief description of the technique followed by examples from the area of expertise from the selected contributor. The second part of the book attempts to integrate previously analyzed technologies into the different aspects of molecular diagnostics, such as identification of genetically modified organisms, stem cells, pharmacogenomics, modern forensic science, molecular microbiology, and genetic diagnosis. Part three focuses on various everyday issues in a diagnostic laboratory, from genetic counseling and related ethical and psychological issues, to safety and quality management.

  • Presents a comprehensive account of all new technologies and applications used in clinical diagnostic laboratories
  • Explores a wide range of molecular-based tests that are available to assess DNA variation and changes in gene expression
  • Offers clear translational presentations by the top molecular pathologists, clinical chemists, and molecular geneticists in the field
Table of Contents
  • List of Contributors
  • Preface, Third Edition
  • Chapter 1. Molecular Diagnostics: Past, Present, and Future
    • 1.1. Introduction
    • 1.2. History of Molecular Diagnostics: Inventing the Wheel
    • 1.3. The Post-Polymerase Chain Reaction Revolution
    • 1.4. Molecular Diagnostics in the Post-Genomic Era
    • 1.5. Future Perspectives: What Lies Beyond
    • 1.6. Conclusions
  • Chapter 2. Describing DNA Variants (Mutation Nomenclature)
    • 2.1. Introduction
    • 2.2. History
    • 2.3. Authorization
    • 2.4. Definitions
    • 2.5. Variant Descriptions
    • 2.6. Mutalyzer
    • 2.7. Concluding Remarks
  • Chapter 3. Low- and Medium-Throughput Variant Detection Methods: A Historical Perspective
    • 3.1. Introduction
    • 3.2. Genetic Screening Methods
    • 3.3. Genetic Scanning Methods
    • 3.4. Conclusions
  • Chapter 4. Quantitative Polymerase Chain Reaction
    • 4.1. History of the Polymerase Chain Reaction
    • 4.2. Principle of Real-Time Polymerase Chain Reaction
    • 4.3. Real-Time Thermal Cyclers
    • 4.4. How Data Are Obtained
    • 4.5. How Data Are Quantified
    • 4.6. Multiplex Quantitative Polymerase Chain Reaction
    • 4.7. Applications of Quantitative Polymerase Chain Reaction and Reverse Transcriptase-Quantitative Polymerase Chain Reaction
    • 4.8. Criteria for Optimizing Quantitative Polymerase Chain Reaction Assays
    • 4.9. Conclusions
  • Chapter 5. Integrated Polymerase Chain Reaction Technologies (Sample-to-Answer Technologies)
    • 5.1. Introduction
    • 5.2. Commercial Sample-to-Answer Assay Systems
    • 5.3. Clinical Applications: Performance for Infectious Pathogen Diagnostics
    • 5.4. Forensic Applications: Performance for Human Identity Testing
    • 5.5. Continuing Evolution of Sample-to-Answer Technologies
  • Chapter 6. High-Resolution Melting Curve Analysis for Molecular Diagnostics
    • 6.1. Introduction to Melting Analysis
    • 6.2. Genotyping of Known Variants by High-Resolution Melting
    • 6.3. Variant Scanning by High-Resolution Melting
    • 6.4. Specific Examples of High-Resolution Melting in Clinical Diagnostics
    • 6.5. Other Applications of High-Resolution Melting in Molecular Diagnostics
    • 6.6. Melting Curve Prediction and Assay Design Tools
    • 6.7. Conclusions
  • Chapter 7. Molecular Techniques for DNA Methylation Studies
    • 7.1. Introduction
    • 7.2. Clinical Applications of DNA Methylation Analysis
    • 7.3. Methods for DNA Methylation Analysis
    • 7.4. Single-Cell DNA Methylation Analysis
    • 7.5. Conclusions
  • Chapter 8. Perspectives for Future DNA Sequencing Techniques and Applications
    • 8.1. Introduction
    • 8.2. Commercially Available Analysis Platforms
    • 8.3. Techniques/Systems in Development
    • 8.4. Potential Future Techniques/Systems/Analysis Platforms
    • 8.5. Perspectives for Future Applications and Diagnostics Techniques
    • 8.6. Conclusions
  • Chapter 9. Advanced Personal Genome Sequencing as the Ultimate Diagnostic Test
    • 9.1. Introduction
    • 9.2. Advanced Whole-Genome Sequencing
    • 9.3. What Is Needed to Implement This Vision of Genomic Precision Health Care Fully?
    • 9.4. Conclusion
  • Chapter 10. Application of Padlock and Selector Probes in Molecular Medicine
    • 10.1. Introduction
    • 10.2. Padlock and Selector Probes
    • 10.3. Application of Padlock and Molecular Inversion Probes for Genotyping
    • 10.4. Biosensor Approaches Based on Rolling Circle–Amplified Padlock Probes
    • 10.5. Application of Padlock Probes for Infectious Disease Diagnostics
    • 10.6. Targeted Multiplex Copy Number Variation Analysis Using Selector Probes
    • 10.7. High-Throughput Targeted Sequencing Using Selectors and Gap-Fill Padlock Probes
    • 10.8. In Situ Nucleic Acid Detection Using Padlock Probes
    • 10.9. Automation and Miniaturization of Padlock Probe/Rolling Circle Amplification Assays
    • 10.10. Conclusions
  • Chapter 11. Advances in Microfluidics and Lab-on-a-Chip Technologies
    • 11.1. Overview
    • 11.2. Microfluidics for DNA Amplification and Analysis
    • 11.3. Microfluidics for High-Resolution Melting Analysis
    • 11.4. Microfluidics in Cytogenetics
    • 11.5. Microfluidics for Protein Detection and Analysis
    • 11.6. Microfluidic Sample Preparation
    • 11.7. Microfluidics in Cell Sorting
    • 11.8. Future of Microfluidics for Medical Diagnostics
  • Chapter 12. Protein Diagnostics by Proximity Ligation: Combining Multiple Recognition and DNA Amplification for Improved Protein Analyses
    • 12.1. Introduction
    • 12.2. Binding the Proteome
    • 12.3. Current Affinity-Based Protein Detection Assays
    • 12.4. Proximity-Dependent Nucleic Acid–Based Assays
    • 12.5. Conclusion and Future Perspectives
  • Chapter 13. Application of Proteomics to Medical Diagnostics
    • 13.1. Introduction
    • 13.2. Clinical Impact and “Proteomics” Potential
    • 13.3. Strategies for Mass Spectrometry–Based Proteomics: Discovery and Verification
    • 13.4. Bioinformatics
    • 13.5. Examples of Discovery and Verification Proteomics
    • 13.6. Examples of Protein-Based Diagnostics Assays
    • 13.7. Challenges in Clinical Proteomics
    • 13.8. Future Advances and Concluding Remarks
  • Chapter 14. Molecular Cytogenetics in Molecular Diagnostics
    • 14.1. Introduction
    • 14.2. From Conventional to Molecular Cytogenetics
    • 14.3. Fluorescence In Situ Hybridization
    • 14.4. Basic Technical Elements and Materials
    • 14.5. Types of Fluorescence In Situ Hybridization Probes and Fluorescence In Situ Hybridization Approaches for Metaphase and Interphase Fluorescence In situ Hybridization
    • 14.6. Multicolor Fluorescence In Situ Hybridization Screening Assays
    • 14.7. Multicolor Whole-Metaphase Scanning Techniques
    • 14.8. Multicolor Chromosome Banding Techniques
    • 14.9. Whole-Genome Scanning and Comparative Genomic Hybridization
    • 14.10. Array-Based Techniques (Microarray)
    • 14.11. Conclusions and Future Perspectives
    • Glossary
  • Chapter 15. Cytogenomics of Solid Tumors by Next-Generation Sequencing: A Clinical Perspective
    • 15.1. A Tumor Presents
    • 15.2. From Microscopic Examination to Molecular Cytogenomics
    • 15.3. The Promise of Liquid Biopsy for Cancer Diagnostics
    • 15.4. Cytogenomic Applications
    • 15.5. Implementation of Cytogenomics in the Clinic
    • 15.6. Bioinformatics and Data Analysis
    • 15.7. Concluding Remarks
  • Chapter 16. Pharmacogenomics in Clinical Care and Drug Discovery
    • 16.1. Introduction
    • 16.2. Pharmacogenetics Versus Pharmacogenomics
    • 16.3. History of Pharmacogenomics
    • 16.4. Analytical Methods in Pharmacogenomics
    • 16.5. Pharmacogenomics in Clinical Settings
    • 16.6. Population Differences in Pharmacogenomics
    • 16.7. Complex Phenotypes
    • 16.8. Pharmacogenomics and Regulatory Agencies
    • 16.9. Pharmacogenomics in Drug Development
    • 16.10. Useful Resources in Pharmacogenomics
    • 16.11. New Trends in Pharmacogenomics
    • 16.12. Ethical Implications
    • 16.13. Public Health Pharmacogenomics
    • 16.14. Conclusions and Future Perspectives
  • Chapter 17. Nutrigenomics: Integrating Genomic Approaches Into Nutrition Research
    • 17.1. Introduction
    • 17.2. Nature of Genetic Variation
    • 17.3. Nutritional Epidemiology
    • 17.4. Experimental Models
    • 17.5. Defining the Phenotype
    • 17.6. Integrating Complex Data Sets: Data Management, Bioinformatics, and Statistics
    • 17.7. Conclusions
  • Chapter 18. DNA Microarrays and Genetic Testing
    • 18.1. Introduction
    • 18.2. DNA Microarrays
    • 18.3. New Developments in DNA Microarrays and Genetic Testing
  • Chapter 19. Bioinformatics Tools for Data Analysis
    • 19.1. Introduction
    • 19.2. Next-Generation Sequencing Pipelines
    • 19.3. Molecular Pathway Analysis: Why and What?
    • 19.4. Conclusions
  • Chapter 20. Genomic Databases: Emerging Tools for Molecular Diagnostics
    • 20.1. Introduction
    • 20.2. Historical Overview of Genetic Databases
    • 20.3. Models for Database Management
    • 20.4. Mutation Database Types
    • 20.5. Locus-Specific Databases in Molecular Genetic Testing
    • 20.6. National/Ethnic Mutation Databases: Archiving the Genomic Basis of Human Disorders on a Population Basis
    • 20.7. Database Management Systems for Locus-Specific Databases and National/Ethnic Mutation Databases
    • 20.8. Incentivizing Data Sharing: The Microattribution Approach
    • 20.9. Future Challenges
    • 20.10. Conclusions
  • Chapter 21. Molecular Diagnostic Applications in Forensic Science
    • 21.1. Introduction
    • 21.2. Genetic Markers Commonly Used for Forensic Analysis
    • 21.3. DNA Extraction Methodologies
    • 21.4. DNA Quantitation
    • 21.5. Capillary Electrophoresis and Data Interpretation
    • 21.6. Statistical Calculations
    • 21.7. Next Generation of Forensic DNA Technologies
    • 21.8. Conclusions
  • Chapter 22. New Perspectives in Mass Disaster Victim Identification Assisted by DNA Typing and Forensic Genomics
    • 22.1. Introduction
    • 22.2. Classification of Mass Fatalities and Diverse Scenarios for Human Remains Retrieval
    • 22.3. Conventional Identification Criteria Routinely Used for Human Identification
    • 22.4. Criteria for the Preservation of Remains
    • 22.5. DNA Polymorphisms Used for Tracing Kinship Between Fragmentary Human Remains and the Relatives Claiming Them
    • 22.6. Challenges Concerning DNA Degradation and Contamination
    • 22.7. Criteria Evolution and Technical Approaches Applied to DNA-Based Victim Identification in Mass Disasters From the Early 1990s to Date
    • 22.8. Description of Analyzed Cases
    • 22.9. From Forensic Genetics to Forensic Genomics: The Change of a Paradigm Driven by Technology
    • 22.10. Future Perspectives
  • Chapter 23. Preimplantation Genetic Diagnosis
    • 23.1. Introduction
    • 23.2. Assisted Reproductive Technology and Biopsy
    • 23.3. Preimplantation Genetic Diagnosis for Monogenic Disorders
    • 23.4. Preimplantation Genetic Diagnosis for Chromosomal Aberrations
    • 23.5. Emerging Technologies
    • 23.6. Clinical Outcome of Preimplantation Genetic Diagnosis
    • 23.7. Accuracy and Quality Control
    • 23.8. Conclusions and Future Perspectives
    • Web Resources
  • Chapter 24. Noninvasive Cell-Free DNA Prenatal Testing for Fetal Aneuploidy in Maternal Blood
    • 24.1. Introduction
    • 24.2. Established Prenatal Screening and Diagnosis Practices
    • 24.3. Historical Background of Noninvasive Prenatal Testing
    • 24.4. Origin of Cell-Free Fetal DNA
    • 24.5. Noninvasive Prenatal Testing Methodologies
    • 24.6. Biological and Technical Factors That Affect Noninvasive Prenatal Testing Results
    • 24.7. Noninvasive Prenatal Testing in Clinical Trials
    • 24.8. Noninvasive Prenatal Testing in the Clinical Setting
    • 24.9. Counseling and Ethical Issues
    • 24.10. Future Applications of Noninvasive Prenatal Testing
    • 24.11. Conclusions
  • Chapter 25. Genetic Testing and Psychology
    • 25.1. Introduction
    • 25.2. Getting to the Test: Awareness, Access, and Advertising
    • 25.3. Individual Factors Influencing the Utilization of Genetic Testing
    • 25.4. Getting the Genetic Test Results: Personal Impact and Professional Communication
    • 25.5. Family Communication
    • 25.6. Future Challenges: Complexity and Diversity
  • Chapter 26. Genomic Medicine in Developing Countries and Resource-Limited Environments
    • 26.1. Introduction
    • 26.2. Conclusions and Future Perspectives
  • Chapter 27. Public Understanding of Genetic Testing and Obstacles to Genetics Literacy
    • 27.1. Genetics Literacy and the Public Understanding of Genetic Testing
    • 27.2. Obstacles to Genetics Literacy and How These Might Be Overcome
    • 27.3. Conclusions and Suggestions
  • Chapter 28. Safety and the Biorepository
    • 28.1. Introduction
    • 28.2. Understanding Regulatory and Other Safety Issues Relevant to Biorepositories
    • 28.3. Individuals Involved in Oversight of a Biorepository
    • 28.4. Safety Training/Employee Education in a Biorepository
    • 28.5. Biorepository Safety Areas
    • 28.6. Conclusions
  • Chapter 29. Quality Assurance in Genetic Laboratories
    • 29.1. Introduction
    • 29.2. International Standards
    • 29.3. Accreditation and Certification
    • 29.4. Elements of a Quality Management System
    • 29.5. Quality Control
    • 29.6. Quality Assessment
    • 29.7. Diagnostic Validation
    • 29.8. Quality Improvement
    • 29.9. Conclusions
  • Index

This Book is For Premium Members Only

Become a Premium Now