Review Article

Genomic Medicine

Alan E. Guttmacher, M.D., Editor, Francis S. Collins, M.D., Ph.D., Editor

Genomic Medicine — A Primer

Alan E. Guttmacher, M.D., and Francis S. Collins, M.D., Ph.D.

N Engl J Med 2002; 347:1512-1520November 7, 2002DOI: 10.1056/NEJMra012240

Article

Humans have known for millennia that heredity affects health.1 However, Mendel's seminal contribution to the elucidation of the mechanisms by which heredity affects phenotype occurred less than 150 years ago, and Garrod began applying this knowledge to human health only at the start of the past century. For most of the 20th century, many medical practitioners viewed genetics as an esoteric academic specialty; that view is now dangerously outdated.

The Advent of Genomic Medicine

The recent completion of the draft sequence of the human genome2,3 and related developments have increased interest in genetics, but confusion remains among health professionals and the public about the role of genetic information in medical practice. Inaccurate beliefs about genetics persist, including the view that in the past it had no effect on the practice of medicine and that its influence today is pervasive. In fact, for decades knowledge of genetics has had a large role in the health care of a few patients and a small role in the health care of many. We have recently entered a transition period in which specific genetic knowledge is becoming critical to the delivery of effective health care for everyone.

If genetics has been misunderstood, genomics is even more mysterious — what, exactly, is the difference? Genetics is the study of single genes and their effects. “Genomics,”4 a term coined only 15 years ago, is the study not just of single genes, but of the functions and interactions of all the genes in the genome. Genomics has a broader and more ambitious reach than does genetics. The science of genomics rests on direct experimental access to the entire genome and applies to common conditions, such as breast cancer5 and colorectal cancer,6 human immunodeficiency virus (HIV) infection,7 tuberculosis,8 Parkinson's disease,9 and Alzheimer's disease.10 These common disorders are also all due to the interactions of multiple genes and environmental factors. They are thus known as multifactorial disorders. Genetic variations in these disorders may have a protective or a pathologic role in the expression of diseases.

The role of genomics in health care is in part highlighted by the decreasing effect of certain environmental factors, such as infectious agents, on the burden of disease. Genomics also contributes to the understanding of such important infectious diseases as the acquired immunodeficiency syndrome (AIDS)7 and tuberculosis.8

The following two case vignettes illustrate how knowledge of genomics may lead to better management of common medical conditions.

Thirty-four-year-old Kathleen becomes pregnant and sees a new physician for her first prenatal visit. Her medical history is remarkable for an episode of deep venous thrombosis five years earlier while she was taking oral contraceptives; her mother had had deep venous thrombosis when pregnant with Kathleen. Her physician suspects that Kathleen has a hereditary thrombophilia and obtains blood tests to screen for a genetic predisposition to thrombosis. Kathleen proves to be among the approximately 4 percent of Americans who are heterozygous for a mutation in factor V known as factor V Leiden that increases the risk of thrombotic events. On the basis of this knowledge and her history of possibly estrogen-related thromboembolism, she is treated with prophylactic subcutaneous heparin for the balance of her pregnancy. She remains asymptomatic and delivers a healthy, term infant.

Four-year-old John has acute lymphoblastic leukemia and tolerates induction and consolidation chemotherapy well, with minimal side effects. As a key part of his maintenance-treatment protocol, he begins to receive oral mercaptopurine daily, but because a genetic test shows that John is homozygous for a mutation in the gene that encodes thiopurine S-methyltransferase, an enzyme that inactivates mercaptopurine, he receives a greatly reduced dose. Only a few years ago, about 1 in 300 patients had serious, sometimes lethal, hematopoietic adverse effects during mercaptopurine therapy. Although John is in this at-risk minority, a simple genetic test, which is now routine for patients beginning mercaptopurine therapy, alerts his physicians to this genetic predisposition. They reduce his dose of mercaptopurine and carefully monitor his blood levels, ensuring that the drug levels remain therapeutic, rather than toxic. John subsequently has an uneventful several-year maintenance period and achieves complete remission.

The Human Genome

These are two examples of genomic medicine, the application of our rapidly expanding knowledge of the human genome (Figure 1Figure 1A Depiction of Chromosome 16 Based on the Determination of Its Actual Sequence by the Human Genome Project.) to medical practice. Much is known, but much remains mysterious. We know that less than 2 percent of the human genome codes for proteins, while over 50 percent represents repeat sequences of several types, whose function is less well understood. These stretches of repetitive sequences, sometimes wrongly dismissed as “junk DNA,” constitute an informative historical record of evolutionary biology, provide a rich source of information for population genetics and medical genetics, and by introducing changes into coding regions, are active agents for change within the genome.2

A draft sequence of the human genome was announced in 2000, and by September 2002 over 90 percent was in final form — that is, there were no gaps and the data were greater than 99.99 percent accurate. The entire 3.1 gigabases of the DNA sequence should essentially be complete (except for the centromeres and rare unclonable segments) by the spring of 2003. Even with this knowledge in hand, however, we still do not know precisely how many genes the genome contains. Current data indicate that the human genome includes approximately 30,000 to 35,000 genes2 — a number that is substantially smaller than was previously thought. Only about half these genes have recognizable motifs, or DNA-sequence patterns, that suggest possible functions. Mutations known to cause disease have been identified in approximately 1000 genes. However, it is likely that nearly all human genes are capable of causing disease if they are altered substantially. Whereas it was once dogma that one gene makes one protein, it now appears that, through the mechanism of alternative splicing12 (Figure 2Figure 2Alternative Splicing.), more than 100,000 proteins can be derived from these 30,000 to 35,000 genes.2

In addition to alternative splicing, a number of “epigenetic” phenomena, such as methylation and histone modification, can alter the effect of a gene. Furthermore, a complex array of molecular signals allows specific genes to be “turned on” (expressed) or “turned off” in specific tissues and at specific times.

Genes are distributed unevenly across the human genome (Figure 1). Certain chromosomes, particularly 17, 19, and 22, are relatively gene dense as compared with others, such as 4, 8, 13, 18, and Y.3 Moreover, gene density varies within each chromosome, being highest in areas rich in the bases cytosine and guanine, rather than adenine and thymine.2,3 Chromosomes 13, 18, and 21, the three autosomes with the fewest genes, are also the three for which the occurrence of trisomy (i.e., three copies of a chromosome) is compatible with viability.

Not all genes reside on nuclear chromosomes; several dozen involved with energy metabolism are on the mitochondrial chromosome.13 Since ova are rich in mitochondria and sperm are not, mitochondrial DNA is usually inherited from the mother. Therefore, mitochondrial genes — and diseases due to DNA-sequence variants in them — are transmitted in a matrilineal pattern that is distinctly different from the pattern of inheritance of nuclear genes.

Monogenic Conditions

Over the course of the 20th century, a combination of theoretical insights, basic-science research, and clinical observation elucidated the inheritance of single-gene, or monogenic, disorders (also known as mendelian disorders, since they are transmitted in a manner consonant with Mendel's laws of inheritance). Modes of inheritance have been established for thousands of conditions caused by mutations in single genes; these have been catalogued in a textbook11 and, more recently, in an online compendium14 known as Mendelian Inheritance in Man (OMIM). For nearly 100 years, autosomal dominant, autosomal recessive, and X-linked modes of inheritance have been understood and known to cause human disease. In the past few decades, other mechanisms of monogenic inheritance have been described. These include mitochondrial inheritance,13 imprinting (a mechanism by which the effects of certain genes depend on whether they are inherited through the mother or through the father),15 uniparental disomy (the occasional situation in which both members of 1 pair of a person's 23 pairs of chromosomes derive from one parent),16 and expanding trinucleotide repeats (a phenomenon in which a sequence of three base pairs that is normally repeated a number of times in a row in the genome becomes repeated by more than the normal number of times, sometimes causing disease).17

Most single-gene conditions are uncommon. Even the commonest, such as hereditary hemochromatosis (approximate incidence, 1 in 300 persons), cystic fibrosis (approximate incidence, 1 in 3000), alpha1-antitrypsin deficiency (approximate incidence, 1 in 1700), or neurofibromatosis (approximate incidence, 1 in 3000), affect no more than 1 in several hundred people in the United States. However, the total effect of monogenic conditions is substantial, from both the individual patient's and public health perspectives, and increased understanding of genetics has already begun to improve the health of some patients with such conditions. The delineation of the mechanisms by which genetic factors cause monogenic disorders has provided important information about basic pathophysiological processes that underlie related disorders that occur with far greater frequency than do these genetic disorders. For instance, insights regarding familial hypercholesterolemia, a genetic disorder that affects only 1 of every 500 people in the United States, were instrumental to understanding the pathophysiology of atherosclerosis, which affects a large fraction of the population, and the development of the statin drugs, which are among the most frequently prescribed medications.18

Types of Mutation

There are a number of ways to categorize mutations. One is according to the causative mechanism, whereas another is according to their functional effect. When classified according to the mechanism, point mutations — that is, a change in a single DNA base in the sequence — are the most common. There are many types of point mutations. One type is a missense mutation (Figure 3Figure 3Examples of Point Mutations.), a substitution that leads to an alternative amino acid, because of the way in which it changes the three-base sequence, or codon, that codes for an amino acid. Nonsense mutations (Figure 3) are a more dramatically deleterious type of point mutation that change the codon to a “stop” codon, a codon that causes the termination of the protein instead of producing an amino acid. Another type of mutation is the frame-shift mutation (Figure 3), which changes the reading frame of the gene downstream from it, often leading to a premature stop codon.

In terms of functional effect, rather than mechanism, many variants in the human-genome sequence have no phenotypic effect. Among these are silent mutations (Figure 3), which replace one base with another, so that the resultant codon still codes for the same amino acid. Also, mutations may not change the phenotype if the altered codon substitutes one amino acid for another that produces little change in the function of the protein or proteins that the gene encodes. These are referred to as “conservative mutations” (Figure 3). Nonconservative mutations (Figure 3) replace an amino acid with a very different one and are more likely to affect the phenotype.

Although mutations can cause disease by a variety of means, the most common is loss of function. Loss-of-function mutations alter the phenotype by decreasing the quantity or the functional activity of a protein. For instance, mutations in the glucose-6-phosphate dehydrogenase (G6PD) gene on the X chromosome decrease the functional activity of this enzyme, leading to acute hemolytic anemia if a male (who would, of course, have only one copy of the X chromosome) with the mutation is exposed to certain drugs, including sulfonamides, primaquine, and nitrofurantoins. Since genes do not exist just to handle pharmacologic agents, variants that cause a more severe deficiency of glucose-6-phosphate dehydrogenase also lead to hemolytic anemia when affected males ingest fava beans (favism), since this enzyme is also important in the degradation of a component of the beans.19

Some mutations cause disease through a gain of function, whereby the protein takes on some new, toxic function. Expanding exonic CAG trinucleotide repeats that cause disorders including Huntington's disease and spinocerebellar ataxia appear to lead to neuropathologic abnormalities by producing proteins that function abnormally because of expanded polyglutamine tracts (CAG codes for the amino acid glutamine).20 Such gain-of-function mutations are often dominantly inherited, since a single copy of the mutant gene can alter function.

One might assume that mutations in the approximately 98.5 percent of the genome that does not code for proteins do not affect the phenotype. Indeed, most do not. But others are regulatory mutations that may ultimately prove as important in the etiologic process of common diseases as the coding region variants. Regulatory mutations act by altering the expression of a gene. For instance, a regulatory mutation might lead to the loss of expression of a gene, to unexpected expression in a tissue in which it is usually silent, or to a change in the time at which it is expressed. Examples of regulatory mutations associated with disease are those in the flanking region of the FMR1 gene (causing fragile X syndrome),21 the insulin gene flanking region (increasing the risk of type 1 diabetes mellitus),22 a regulatory site of the type I collagen gene (increasing the risk of osteoporosis),23 and an intronic regulatory site of the calpain-10 gene (increasing the risk of type 2 diabetes mellitus).24

Mutations can also decrease the risk of a disease. One example of this is a 32-bp deletion (a frame shift) in a chemokine receptor gene, CCR5. Persons who are homozygous for this deletion prove almost completely resistant to infection with HIV type 1, and those who are heterozygous for the deletion have slower progression from infection to AIDS. These effects arise because CCR5 is an important part of the mechanism by which HIV enters the cell.25

Genes in Common Disease

The study of genomics will most likely make its greatest contribution to health by revealing mechanisms of common, complex diseases, such as hypertension, diabetes, and asthma. So far, most genes involved in common diseases have been identified by virtue of their high penetrance — that is, the mutations lead to disease in a fairly large proportion of people who have them. Examples include mutations in BRCA1 and BRCA2 (increasing the risk of breast and ovarian cancer),26 HNPCC (increasing the risk of hereditary nonpolyposis colorectal cancer),6 MODY 1, MODY 2, and MODY 3 (increasing the risk of diabetes),27 and the gene for α-synuclein (causing Parkinson's disease).9 One can think of these as near-mendelian subgroups of disease within a larger group of affected persons. If a person has such a mutation, the likelihood of disease is great. However, each of these highly penetrant mutations associated with common disease has a prevalence in the general population of only one in several hundred to several thousand people.

From a public health perspective, genes with mutations that are less highly penetrant but much more prevalent have a greater effect on the population than genes that are highly penetrant but uncommon. Such mutations have been reported in genes such as APC (which increases the risk of colorectal cancer)28 and factor V Leiden (which increases the risk of thrombosis).29

An example of the relative contributions of rare, highly penetrant mutations as opposed to common, less penetrant ones is seen in Alzheimer's disease. Rare mutations in presenilin 1, presenilin 2, or the β-amyloid precursor protein gene are highly penetrant causes of early-onset Alzheimer's disease; indeed, Alzheimer's disease develops by the age of 60 years in most people who are heterozygous for a mutation in one of these genes.30 However, because so few people carry a mutation in any of these genes, these mutations play a part in fewer than 1 percent of cases of Alzheimer's disease.31 In contrast, the apolipoprotein E ε4 allele also increases the risk of late-onset Alzheimer's disease (and atherosclerosis32), but more subtly. One representative Finnish study found that Alzheimer's disease develops during the mid-70s in approximately 8 percent of persons who are heterozygous for the ε4 allele and 21 percent of those who are homozygous for it, as compared with 3 percent of those with no ε4 allele.33 Nonetheless, because approximately 26 percent of the U.S. population is heterozygous and 2 percent is homozygous34 for the apolipoprotein E ε4 allele, this genetic factor has a role in many more cases of Alzheimer's disease than do the mutations in the genes for presenilin 1, presenilin 2, and β-amyloid precursor protein combined.

Variation in the Human Genome

One characteristic of the human genome with medical and social relevance is that, on average, two unrelated persons share over 99.9 percent of their DNA sequences.3 However, given the more than 3 billion base pairs that constitute the human genome, this also means that the DNA sequences of two unrelated humans vary at millions of bases. Since a person's genotype represents the blending of parental genotypes, we are each thus heterozygous at about 3 million bases. Many efforts are currently under way, in both the academic and commercial sectors, to catalogue these variants, commonly referred to as “single-nucleotide polymorphisms” (SNPs), and to correlate these specific genotypic variations with specific phenotypic variations relevant to health.

Some SNP–phenotype correlations occur as a direct result of the influence of the SNP on health. More commonly, however, the SNP is merely a marker of biologic diversity that happens to correlate with health because of its proximity to the genetic factor that is actually the cause. In this sense, the term “proximity” is only a rough measure of physical closeness; instead, it connotes that, as genetic material has passed through 5000 generations from our common African ancestral pool, recombination between the SNP and the actual genetic factor has occurred only rarely. In genetic terms, the SNP and the actual genetic factor are said to be in linkage disequilibrium (Figure 4Figure 4Example of Linkage Disequilibrium.).

An extension of the current efforts to catalogue SNPs and correlate them to phenotype are efforts to map and use haplotypes. Whereas a SNP represents a single-nucleotide variant, a haplotype represents a considerably longer sequence of nucleotides (averaging about 25,000), as well as any variants, that tend to be inherited together. SNPs and haplotypes will be the key to the association studies (i.e., studies of affected persons and control subjects) necessary to identify the genetic factors in complex, common diseases, just as family studies have been important to the identification of the genes involved in monogenic conditions.35,36 Also, at least until whole-genome sequencing of individual patients becomes feasible clinically, the identification of SNPs and haplotypes will prove instrumental in efforts to use genomic medicine to individualize health care.

Conclusions

Except for monozygotic twins, each person's genome is unique. All physicians will soon need to understand the concept of genetic variability, its interactions with the environment, and its implications for patient care. With the sequencing of the human genome only months from its finish, the practice of medicine has now entered an era in which the individual patient's genome will help determine the optimal approach to care, whether it is preventive, diagnostic, or therapeutic. Genomics, which has quickly emerged as the central basic science of biomedical research, is poised to take center stage in clinical medicine as well.

Source Information

From the National Human Genome Research Institute, National Institutes of Health, Bethesda, Md.

Address reprint requests to Dr. Guttmacher at Bldg. 31, Rm. 4B09, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-2152, or at .

References

References

  1. 1

    Adams FL. The genuine works of Hippocrates. Vol. 2. New York: William Wood, 1886:338.

  2. 2

    Lander ES, Linton LM, Birren B, et al. Initial sequencing and analysis of the human genome. Nature 2001;409:860-921[Erratum, Nature 2001;411:720, 412:565.]
    CrossRef | Web of Science | Medline

  3. 3

    Venter JC, Adams MD, Myers EW, et al. The sequence of the human genome. Science 2001;291:1304-1351[Erratum, Science 2001;292:1838.]
    CrossRef | Web of Science | Medline

  4. 4

    McKusick VA, Ruddle FH. A new discipline, a new name, a new journal. Genomics 1987;1:1-2
    CrossRef

  5. 5

    Armstrong K, Eisen A, Weber B. Assessing the risk of breast cancer. N Engl J Med 2000;342:564-571
    Full Text | Web of Science | Medline

  6. 6

    Lynch HT. Hereditary nonpolyposis colorectal cancer (HNPCC). Cytogenet Cell Genet 1999;86:130-135
    CrossRef | Medline

  7. 7

    Michael NL. Host genetic influences on HIV-1 pathogenesis. Curr Opin Immunol 1999;11:466-474
    CrossRef | Web of Science | Medline

  8. 8

    Small PM, Fujiwara PI. Management of tuberculosis in the United States. N Engl J Med 2001;345:189-200
    Full Text | Web of Science | Medline

  9. 9

    Mouradian MM. Recent advances in the genetics and pathogenesis of Parkinson disease. Neurology 2002;58:179-185
    CrossRef | Web of Science | Medline

  10. 10

    St George-Hyslop PH. Molecular genetics of Alzheimer disease. Semin Neurol 1999;19:371-383
    CrossRef | Web of Science | Medline

  11. 11

    McKusick VA. Mendelian inheritance in man: catalogs of human genes and genetic disorders. 12th ed. Baltimore: Johns Hopkins University Press, 1998.

  12. 12

    Graveley BR. Alternative splicing: increased diversity in the proteomic world. Trends Genet 2001;17:100-107
    CrossRef | Web of Science | Medline

  13. 13

    Wallace DC. Mitochondrial diseases in man and mouse. Science 1999;283:1482-1488
    CrossRef | Web of Science | Medline

  14. 14

    Online Mendelian Inheritance in Man, OMIM (TM). Baltimore: McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University, 2000. (Accessed October 15, 2002, at http://www.ncbi.nlm.nih.gov/omim/.)

  15. 15

    Reik W, Walter J. Genomic imprinting: parental influence on the genome. Nat Rev Genet 2001;2:21-32
    CrossRef | Web of Science | Medline

  16. 16

    Robinson WP. Mechanisms leading to uniparental disomy and their clinical consequences. Bioessays 2000;22:452-459
    CrossRef | Web of Science | Medline

  17. 17

    Sinden RR. Biological implications of the DNA structures associated with disease-causing triplet repeats. Am J Hum Genet 1999;64:346-353
    CrossRef | Web of Science | Medline

  18. 18

    Goldstein JL, Hobbs HH, Brown MS. Familial hypercholesterolemia. In: Scriver CR, Beaudet AL, Sly WS, Valle D, eds. The metabolic & molecular bases of inherited disease. 8th ed. Vol. 2. New York: McGraw-Hill, 2001:2863-913.

  19. 19

    Luzzatto L, Mehta A, Vulliamy T. Glucose 6-phosphate dehydrogenase deficiency. In: Scriver CR, Beaudet AL, Sly WS, Valle D, eds. The metabolic & molecular bases of inherited disease. 8th ed. Vol. 3. New York: McGraw-Hill, 2001:4517-53.

  20. 20

    Goldberg YP, Nicholson DW, Rasper DM, et al. Cleavage of huntingtin by apopain, a proapoptotic cysteine protease, is modulated by the polyglutamine tract. Nat Genet 1996;13:442-449
    CrossRef | Web of Science | Medline

  21. 21

    Jin P, Warren ST. Understanding the molecular basis of fragile X syndrome. Hum Mol Genet 2000;9:901-908
    CrossRef | Web of Science | Medline

  22. 22

    Todd JA. From genome to aetiology in a multifactorial disease, type 1 diabetes. Bioessays 1999;21:164-174
    CrossRef | Web of Science | Medline

  23. 23

    Grant SFA, Reid DM, Blake G, Herd R, Fogelman I, Ralston SH. Reduced bone density and osteoporosis associated with a polymorphic Sp1 binding site in the collagen type I α 1 gene. Nat Genet 1996;14:203-205
    CrossRef | Web of Science | Medline

  24. 24

    Horikawa Y, Oda N, Cox NJ, et al. Genetic variation in the gene encoding calpain-10 is associated with type 2 diabetes mellitus. Nat Genet 2000;26:163-175[Erratum, Nat Genet 2000;26:502.]
    CrossRef | Web of Science | Medline

  25. 25

    Dean M, Carrington M, Winkler C, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Science 1996;273:1856-1862[Erratum, 1996;274:1069.]
    CrossRef | Web of Science | Medline

  26. 26

    Nathanson KN, Wooster R, Weber BL. Breast cancer genetics: what we know and what we need. Nat Med 2001;7:552-556[Erratum, Nat Med 2001;7:749.]
    CrossRef | Web of Science | Medline

  27. 27

    Froguel P, Velho G. Molecular genetics of maturity-onset diabetes of the young. Trends Endocrinol Metab 1999;10:142-146
    CrossRef | Web of Science | Medline

  28. 28

    Fearnhead NS, Britton MP, Bodmer WF. The ABC of APC. Hum Mol Genet 2001;10:721-733
    CrossRef | Web of Science | Medline

  29. 29

    Major DA, Sane DC, Herrington DM. Cardiovascular implications of the factor V Leiden mutation. Am Heart J 2000;140:189-195
    CrossRef | Web of Science | Medline

  30. 30

    Campion D, Dumanchin C, Hannequin D, et al. Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum. Am J Hum Genet 1999;65:664-670
    CrossRef | Web of Science | Medline

  31. 31

    Roses AD. Alzheimer diseases: a model of gene mutations and susceptibility polymorphisms for complex psychiatric diseases. Am J Med Genet 1998;81:49-57
    CrossRef | Web of Science | Medline

  32. 32

    Curtiss LK, Boisvert WA. Apolipoprotein E and atherosclerosis. Curr Opin Lipidol 2000;11:243-251
    CrossRef | Web of Science | Medline

  33. 33

    Kuusisto J, Koivisto K, Kervinen K, et al. Association of apolipoprotein E phenotypes with late onset Alzheimer's disease: population based study. BMJ 1994;309:636-638
    CrossRef | Web of Science | Medline

  34. 34

    Roses AD. Apolipoprotein E alleles as risk factors in Alzheimer's disease. Annu Rev Med 1996;47:387-400
    CrossRef | Web of Science | Medline

  35. 35

    Reich DE, Cargill M, Bolk S, et al. Linkage disequilibrium in the human genome. Nature 2001;411:199-204
    CrossRef | Web of Science | Medline

  36. 36

    Goldstein DB, Weale ME. Population genetics: linkage disequilibrium holds the key. Curr Biol 2001;11:R576-R579
    CrossRef | Web of Science | Medline

Citing Articles (207)

Citing Articles

  1. 1

    A. van de Winkel, R. Massl, E. J. Kuipers, L. J. W. van der Laan, M. P. Peppelenbosch. (2013) Digestive Disease Week 2011: highlights of clinical and preclinical research on Barrett's esophagus and associated esophageal adenocarcinoma. Diseases of the Esophagus 26:2, 130-140
    CrossRef

  2. 2

    Teri A. Manolio, Rex L. Chisholm, Brad Ozenberger, Dan M. Roden, Marc S. Williams, Richard Wilson, David Bick, Erwin P. Bottinger, Murray H. Brilliant, Charis Eng, Kelly A. Frazer, Bruce Korf, David H. Ledbetter, James R. Lupski, Clay Marsh, David Mrazek, Michael F. Murray, Peter H. O’Donnell, Daniel J. Rader, Mary V. Relling, Alan R. Shuldiner, David Valle, Richard Weinshilboum, Eric D. Green, Geoffrey S. Ginsburg. (2013) Implementing genomic medicine in the clinic: the future is here. Genetics in Medicine
    CrossRef

  3. 3

    Barry S. Coller. A Brief History of Ideas About Platelets in Health and Disease. In: Platelets. Elsevier, 2013:xix-xliv.
    CrossRef

  4. 4

    Samir B. Damani, Eric J. Topol. Coronary Artery Disease and Myocardial Infarction. In: Genomic and Personalized Medicine. Elsevier, 2013:516-529.
    CrossRef

  5. 5

    Katherine Chadwell. (2013) Clinical Practice on the Horizon. Clinical Nurse Specialist 27:1, 36-43
    CrossRef

  6. 6

    Claude Lenfant. (2013) Prospects of personalized medicine in cardiovascular diseases. Metabolism 62, S6-S10
    CrossRef

  7. 7

    Siobhan M Dolan, Inge Christiaens. (2013) Genome-wide association studies in preterm birth: implications for the practicing obstetrician-gynaecologist. BMC Pregnancy and Childbirth 13:Suppl 1, S4
    CrossRef

  8. 8

    John B. Korman, David E. Fisher. (2013) Developing melanoma therapeutics: overview and update. Wiley Interdisciplinary Reviews: Systems Biology and Medicinen/a-n/a
    CrossRef

  9. 9

    Christoph I. Lee, Lawrence W. Bassett, Mei Leng, Sally L. Maliski, Bryan B. Pezeshki, Colin J. Wells, Carol M. Mangione, Arash Naeim. (2012) Patients’ willingness to participate in a breast cancer biobank at screening mammogram. Breast Cancer Research and Treatment 136:3, 899-906
    CrossRef

  10. 10

    P. Cardinal-Fernández, A. Ferruelo, A. Martín-Pellicer, N. Nin, A. Esteban, J.A. Lorente. (2012) Genetic determinants of acute renal damage risk and prognosis: A systematic review. Medicina Intensiva (English Edition) 36:9, 626-633
    CrossRef

  11. 11

    S. Giampaoli, G. Chillemi, F. Valeriani, D. Lazzaro, M. Borro, G. Gentile, M. Simmaco, G. Zanni, A. Berti, V. Romano Spica. (2012) The SNPs in the human genetic blueprint era. New Biotechnology
    CrossRef

  12. 12

    Haiying Wang, John Jiang, Bianca Mostert, Anieta Sieuwerts, John W.M. Martens, Stefan Sleijfer, John A. Foekens, Yixin Wang. (2012) Allele-Specific, Non-extendable Primer Blocker PCR (AS-NEPB-PCR) for DNA Mutation Detection in Cancer. The Journal of Molecular Diagnostics
    CrossRef

  13. 13

    , Carol A. Christianson, Karen Potter Powell, Susan Estabrooks Hahn, Susan H. Blanton, Jessica Bogacik, Vincent C. Henrich. (2012) The Use of a Family History Risk Assessment Tool within a Community Health Care System: Views of Primary Care Providers. Journal of Genetic Counseling 21:5, 652-661
    CrossRef

  14. 14

    Jessica T. DeFrank, Lisa A. Carey, Noel T. Brewer. (2012) Understanding how breast cancer patients use risk information from genomic tests. Journal of Behavioral Medicine
    CrossRef

  15. 15

    Kathleen T. Hickey, Robert R. Sciacca, Mary S. McCarthy. (2012) Descriptive survey of Summer Genetics Institute nurse graduates in the USA. Nursing & Health Sciencesno-no
    CrossRef

  16. 16

    Heather Skirton, Leigh Jackson, Lesley Goldsmith, Anita O’Connor. (2012) Genomic medicine: what are the challenges for the National Health Service?. Personalized Medicine 9:5, 539-545
    CrossRef

  17. 17

    P. Boffetta, D. M. Winn, J. P. Ioannidis, D. C. Thomas, J. Little, G. D. Smith, V. J. Cogliano, S. S. Hecht, D. Seminara, P. Vineis, M. J. Khoury. (2012) Recommendations and proposed guidelines for assessing the cumulative evidence on joint effects of genes and environments on cancer occurrence in humans. International Journal of Epidemiology 41:3, 686-704
    CrossRef

  18. 18

    Amilcar Ezequiel Rizzo, Howard I. Maibach. (2012) Personalizing dermatology: The future of genomic expression profiling to individualize dermatologic therapy. Journal of Dermatological Treatment 23:3, 161-167
    CrossRef

  19. 19

    Xavier Delannay, Graham McLaren, Jean-Marcel Ribaut. (2012) Fostering molecular breeding in developing countries. Molecular Breeding 29:4, 857-873
    CrossRef

  20. 20

    Nagla Gasmelseed, Afrah Awad Elsir, Pasquale DeBlasio, Ida Biunno. (2012) Sub-Saharan centralized biorepository for genetic and genomic research. Science of The Total Environment 423, 210-213
    CrossRef

  21. 21

    P. Cardinal-Fernández, A. Ferruelo, A. Martín-Pellicer, N. Nin, A. Esteban, J.A. Lorente. (2012) Determinantes genéticos del riesgo y pronóstico del daño renal agudo: una revisión sistemática. Medicina Intensiva
    CrossRef

  22. 22

    Jeffrey Comer, Aleksei Aksimentiev. (2012) Predicting the DNA Sequence Dependence of Nanopore Ion Current Using Atomic-Resolution Brownian Dynamics. The Journal of Physical Chemistry C 116:5, 3376-3393
    CrossRef

  23. 23

    Jean F. Jenkins, Kathleen A. Calzone. (2012) Are Nursing Faculty Ready to Integrate Genomic Content Into Curricula?. Nurse Educator 37:1, 25-29
    CrossRef

  24. 24

    Mark L. Gonzalgo, Alan Keith Meeker. Molecular Genetics and Cancer Biology. In: Campbell-Walsh Urology. Elsevier, 2012:530-567.e16.
    CrossRef

  25. 25

    H P Rang, M M Dale, J M Ritter, R J Flower, G Henderson. Biopharmaceuticals and gene therapy. In: Rang & Dale's Pharmacology. Elsevier, 2012:715-725.
    CrossRef

  26. 26

    David J. Tester, Michael J. Ackerman. Genomics and Principles of Clinical Genetics. In: Electrophysiological Disorders of the Heart. Elsevier, 2012:73-84.
    CrossRef

  27. 27

    Shweta U. Dhar, Raye L. Alford, Elizabeth A. Nelson, Lorraine Potocki. (2012) Enhancing exposure to genetics and genomics through an innovative medical school curriculum. Genetics in Medicine 14:1, 163-167
    CrossRef

  28. 28

    Moo-Sik Lee, Andreas J. Flammer, Lilach O. Lerman, Amir Lerman. (2012) Personalized Medicine in Cardiovascular Diseases. Korean Circulation Journal 42:9, 583
    CrossRef

  29. 29

    Arkadiusz Z. Dudek, Steven F. Powell. Oncology. In: Pharmaceutical Sciences Encyclopedia. John Wiley & Sons, Inc., 2011.
    CrossRef

  30. 30

    Kathleen A. Calzone, Bonnie Jerome-D’Emilia, Jean Jenkins, Constance Goldgar, Michael Rackover, John Jackson, Ye Chen, John Voss, W. Gregory Feero. (2011) Establishment of the Genetic/Genomic Competency Center for Education. Journal of Nursing Scholarship 43:4, 351-358
    CrossRef

  31. 31

    Alison Harvey. (2011) From lab to lifestyle: translating genomics into healthcare practices. New Genetics and Society 30:4, 309-327
    CrossRef

  32. 32

    Rogan Carr, Jeffrey Comer, Aleksei Aksimentiev. Modeling the Interface between Biological and SyntheticComponents in Hybrid Nanosystems. In: Simulations in Nanobiotechnology. CRC Press, 2011:43-60.
    CrossRef

  33. 33

    Isaac S. Chan, Geoffrey S. Ginsburg. (2011) Personalized Medicine: Progress and Promise. Annual Review of Genomics and Human Genetics 12:1, 217-244
    CrossRef

  34. 34

    Sam Kassegne, Bhuvnesh Arya. Genomics and DNA Microarrays. In: Microfluidics and Nanofluidics Handbook. CRC Press, 2011:1043-1052.
    CrossRef

  35. 35

    Patricia Marino, Carole Siani, François Bertucci, Henri Roche, Anne-Laure Martin, Patrice Viens, Valérie Seror. (2011) Economic issues involved in integrating genomic testing into clinical care: the case of genomic testing to guide decision-making about chemotherapy for breast cancer patients. Breast Cancer Research and Treatment 129:2, 401-409
    CrossRef

  36. 36

    Luke Burchill, Steven Greenway, Candice K. Silversides, Seema Mital. (2011) Genetic Counseling in the Adult with Congenital Heart Disease: What is the Role?. Current Cardiology Reports 13:4, 347-355
    CrossRef

  37. 37

    Gwenyth R. Wallen, Georgie Cusack, Suzan Parada, Claiborne Miller-Davis, Tannia Cartledge, Jan Yates. (2011) Evaluating a hybrid web-based basic genetics course for health professionals. Nurse Education Today 31:6, 638-642
    CrossRef

  38. 38

    Mariano A Giorgi, Christian Caroli, Hernán Cohen Arazi, Guillermo Di Girolamo. (2011) Pharmacogenomics and adverse drug reactions: the case of statins. Expert Opinion on Pharmacotherapy 12:10, 1499-1509
    CrossRef

  39. 39

    Kenneth Offit. (2011) Personalized medicine: new genomics, old lessons. Human Genetics 130:1, 3-14
    CrossRef

  40. 40

    Sanjay Jain, Laura De Petris, Masato Hoshi, Shreeram Akilesh, Rajshekhar Chatterjee, Helen Liapis. (2011) Expression profiles of podocytes exposed to high glucose reveal new insights into early diabetic glomerulopathy. Laboratory Investigation 91:4, 488-498
    CrossRef

  41. 41

    Brent J. Small, Kerri Sharp Rawson, Erin Walsh, Heather S. L. Jim, Tiffany F. Hughes, Lindsay Iser, Michael A. Andrykowski, Paul B. Jacobsen. (2011) Catechol-O-methyltransferase genotype modulates cancer treatment-related cognitive deficits in breast cancer survivors. Cancer 117:7, 1369-1376
    CrossRef

  42. 42

    W. Gregory Feero, Philip Zazove, Nancy G. Stevens. Clinical Genetics (Genomics). In: Textbook of Family Medicine. Elsevier, 2011:1013-1021.
    CrossRef

  43. 43

    Durjoy Majumder, Abhik Mukherjee. (2011) A passage through systems biology to systems medicine: adoption of middle-out rational approaches towards the understanding of therapeutic outcomes in cancer. The Analyst 136:4, 663
    CrossRef

  44. 44

    Carolina Aponte Urdaneta, John L. Beyer. Genetics of Affective Disorders. In: Principles and Practice of Geriatric Psychiatry. John Wiley & Sons, Ltd, 2010:460-463.
    CrossRef

  45. 45

    Medha Bhagwat. Searching NCBI's dbSNP Database. In: Current Protocols in Bioinformatics. John Wiley & Sons, Inc., 2010.
    CrossRef

  46. 46

    Gustavo de los Campos, Daniel Gianola, David B. Allison. (2010) Predicting genetic predisposition in humans: the promise of whole-genome markers. Nature Reviews Genetics 11:12, 880-886
    CrossRef

  47. 47

    György Kosztolányi. (2010) Az első posztgenom évtized az orvostudományban. Orvosi Hetilap 151:51, 2099-2104
    CrossRef

  48. 48

    Balaji S. Srinivasan, Eric A. Evans, Jason Flannick, A. Scott Patterson, Christopher C. Chang, Tuan Pham, Sharon Young, Amit Kaushal, James Lee, Jessica L. Jacobson, Pasquale Patrizio. (2010) A universal carrier test for the long tail of Mendelian disease. Reproductive BioMedicine Online 21:4, 537-551
    CrossRef

  49. 49

    Chienming Chen, Chihchia Chen, Tsanhuang Shih, Tunwen Pai, Chinhua Hu, Wenshyong Tzou. (2010) Efficient algorithms for identifying orthologous simple sequence repeats of disease genes. Journal of Systems Science and Complexity 23:5, 906-916
    CrossRef

  50. 50

    György Kosztolányi, Jean-Jacques Cassiman. (2010) The medical geneticist as expert in the transgenerational and developmental aspects of diseases. European Journal of Human Genetics 18:10, 1075-1076
    CrossRef

  51. 51

    K. C. Koenen, M. Uddin, A. B. Amstadter, S. Galea. (2010) Incorporating the social environment in genotype environment interaction studies of mental disorders. International Journal of Clinical Practice 64:11, 1489-1492
    CrossRef

  52. 52

    Elizabeth L. Pestka, Della J. Derscheid, Susan M. Ellenbecker, Phyllis J. Schmid, Marybeth L. O'Neil, Rita J. Ray-Mihm, Debra L. Cox. (2010) Use of Genomic Assessments and Interventions in Psychiatric Nursing Practice. Issues in Mental Health Nursing 31:10, 623-630
    CrossRef

  53. 53

    J. Ignacio Valle-Mansilla, Miguel Ruiz-Canela, Daniel P. Sulmasy. (2010) Patients’ Attitudes to Informed Consent for Genomic Research With Donated Samples. Cancer Investigation 28:7, 726-734
    CrossRef

  54. 54

    Fang Li, Richard M. Weinshilboum. Thiopurine S-Methyltransferase Pharmacogenomics: Protein Misfolding, Aggregation, and Degradation. In: Protein Misfolding Diseases. John Wiley & Sons, Inc., 2010:453-468.
    CrossRef

  55. 55

    Kirsty Challen, Hilary Harris, Ulf Kristoffersson, Irmgard Nippert, Joerg Schmidtke, Leo P. Kate, Caroline Benjamin, Elizabeth Anionwu, Anne-Marie Plass, Claire Julian-Reynier, Rodney Harris. (2010) General practitioner management of genetic aspects of a cardiac disease: a scenario-based study to anticipate providers’ practices. Journal of Community Genetics 1:2, 83-90
    CrossRef

  56. 56

    Feero, W. Gregory, Guttmacher, Alan E., , Feero, W. Gregory, Guttmacher, Alan E., Collins, Francis S., . (2010) Genomic Medicine — An Updated Primer. New England Journal of Medicine 362:21, 2001-2011
    Free Full Text

  57. 57

    Martin Schneider, Jürgen Weitz, Markus W. Büchler. (2010) The focus of Langenbeck’s Archives of Surgery in the 21st century. Langenbeck's Archives of Surgery 395:S1, 75-79
    CrossRef

  58. 58

    Ronald E. Reid. The Pharmacogenomics of Personalized Medicine. In: Pharmaceutical Sciences Encyclopedia. John Wiley & Sons, Inc., 2010.
    CrossRef

  59. 59

    Yijun Zuo, Guolian Kang. (2010) A mixed two-stage method for detecting interactions in genomewide association studies. Journal of Theoretical Biology 262:4, 576-583
    CrossRef

  60. 60

    Paul Roepman. (2010) The future of diagnostic gene-expression microarrays: bridging the gap between bench and bedside. Bioanalysis 2:2, 249-262
    CrossRef

  61. 61

    Alison Stewart, Marta Gwinn, Ron Zimmern, Muin Khoury. Public Health Genomics. In: Essentials of Genomic and Personalized Medicine. Elsevier, 2010:245-255.
    CrossRef

  62. 62

    Stacy E.F. Melanson. Pharmacogenomics. In: Encyclopedia of Analytical Chemistry. John Wiley & Sons, Ltd, 2009.
    CrossRef

  63. 63

    Quanhe Yang, W. Dana Flanders, Ramal Moonesinghe, John P.A. Ioannidis, Idris Guessous, Muin J. Khoury. (2009) Using Lifetime Risk Estimates in Personal Genomic Profiles: Estimation of Uncertainty. The American Journal of Human Genetics 85:6, 786-800
    CrossRef

  64. 64

    Rebekah Hamilton. (2009) Nursing Advocacy in a Postgenomic Age. Nursing Clinics of North America 44:4, 435-446
    CrossRef

  65. 65

    Karen E. Greco, Catherine Salveson. (2009) Identifying Genetics and Genomics Nursing Competencies Common Among Published Recommendations. Journal of Nursing Education 48:10, 557-565
    CrossRef

  66. 66

    M.-N. Kêdoté, É. Racine, H. Doucet. (2009) Perceptions des diabétiques à l’égard du développement de la médecine prédictive. Éthique & Santé 6:3, 127-133
    CrossRef

  67. 67

    Robert A Fowler, Woganee Filate, Michael Hartleib, David W Frost, Chris Lazongas, Michelle Hladunewich. (2009) Sex and critical illness. Current Opinion in Critical Care 15:5, 442-449
    CrossRef

  68. 68

    Noel T. Brewer, Alrick S. Edwards, Suzanne C. O’Neill, Janice P. Tzeng, Lisa A. Carey, Barbara K. Rimer. (2009) When genomic and standard test results diverge: implications for breast cancer patients’ preference for chemotherapy. Breast Cancer Research and Treatment 117:1, 25-29
    CrossRef

  69. 69

    Peter A. Kopp. Genetics and Genomics. In: Brook's Clinical Pediatric Endocrinology. Wiley-Blackwell, 2009:1-23.
    CrossRef

  70. 70

    B. Wilfond, L. F. Ross. (2009) From Genetics to Genomics: Ethics, Policy, and Parental Decision-making. Journal of Pediatric Psychology 34:6, 639-647
    CrossRef

  71. 71

    Alison Harvey. (2009) From genetic risk to post-genomic uncertainties: nutrigenomics and the birth of the “genetic entrepreneur”. New Genetics and Society 28:2, 119-137
    CrossRef

  72. 72

    Anna F Dominiczak, Christian Delles, Sandosh Padmanabhan. (2009) Are isolated populations better for studying genes that predispose to hypertension?. Journal of Hypertension 27:5, 939-940
    CrossRef

  73. 73

    Kenneth B. Margulies, Daniel P. Bednarik, Daniel L. Dries. (2009) Genomics, Transcriptional Profiling, and Heart Failure. Journal of the American College of Cardiology 53:19, 1752-1759
    CrossRef

  74. 74

    Alex R. Kemper, Donald B. Bailey. (2009) Pediatricians’ Knowledge of and Attitudes Toward Fragile X Syndrome Screening. Academic Pediatrics 9:2, 114-117
    CrossRef

  75. 75

    Xiaoming Wang, Lili Liu, James Fackenthal, Shelly Cummings, Oluwatobi I. Olopade, Kisha Hope, Jonathan C. Silverstein, Olufunmilayo I. Olopade. (2009) Translational integrity and continuity: Personalized biomedical data integration. Journal of Biomedical Informatics 42:1, 100-112
    CrossRef

  76. 76

    Ilka Warshawsky. Molecular Biology Basics for the Pathologist. In: Cell and Tissue Based Molecular Pathology. Elsevier, 2009:3-9.
    CrossRef

  77. 77

    Matthew Lanktree, Jisun, Robert A. Hegele. (2008) Genetic testing for atherosclerosis risk: Inevitability or pipe dream?. Canadian Journal of Cardiology 24:11, 851-854
    CrossRef

  78. 78

    Arpana Agrawal, Michael T. Lynskey. (2008) Are there genetic influences on addiction: evidence from family, adoption and twin studies. Addiction 103:7, 1069-1081
    CrossRef

  79. 79

    José Antonio Iribarren Loyarte. (2008) Aplicabilidad de los estudios farmacogenéticos en la práctica clínica. Enfermedades Infecciosas y Microbiología Clínica 26, 45-54
    CrossRef

  80. 80

    Elizabeth Penziner, Janet K. Williams, Cheryl Erwin, Yvonne Bombard, Anne Wallis, Leigh J. Beglinger, Michael R. Hayden, Jane S. Paulsen. (2008) Perceptions of discrimination among persons who have undergone predictive testing for Huntington's disease. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics 147B:3, 320-325
    CrossRef

  81. 81

    John W. Holloway, Ian A. Yang, Stephen T. Holgate. (2008) Interpatient variability in rates of asthma progression: Can genetics provide an answer?. Journal of Allergy and Clinical Immunology 121:3, 573-579
    CrossRef

  82. 82

    Kalpesh K. Patel, Mark W. Babyatsky. (2008) Medical Education: A Key Partner in Realizing Personalized Medicine in Gastroenterology. Gastroenterology 134:3, 656-661
    CrossRef

  83. 83

    Claudia Wild. (2008) Polymorphism-screening: genetic testing for predisposition—guidance for technology assessment. Poiesis & Praxis 5:1, 1-14
    CrossRef

  84. 84

    Gwen Anderson, Alison Metcalfe. (2008) Calling for international collaborative research in nursing, genetics and genomics: A discussion paper. International Journal of Nursing Studies 45:2, 323-328
    CrossRef

  85. 85

    Alfredo Fucito, Chiara Lucchetti, Antonio Giordano, Gaetano Romano. (2008) Genetic and epigenetic alterations in breast cancer: What are the perspectives for clinical practice?. The International Journal of Biochemistry & Cell Biology 40:4, 565-575
    CrossRef

  86. 86

    Caroline Benjamin, Maria Flynn, Christine Hallett, Ian Ellis, Katie Booth. (2008) The use of the life course paradigm and life course charts to explore referral for family history of breast cancer. International Journal of Nursing Studies 45:1, 95-109
    CrossRef

  87. 87

    Peter N. Bennett, Morris J. Brown. General pharmacology. In: Clinical Pharmacology. Elsevier, 2008:75-114.
    CrossRef

  88. 88

    Lisa DiMartino, Kelli D. Allen, Edward Kasarskis, Jennifer H. Lindquist, Cynthia J. Coffman, Eugene Z. Oddone. (2007) Characteristics associated with participation in DNA banking: The National Registry of Veterans with ALS. Contemporary Clinical Trials 28:5, 572-582
    CrossRef

  89. 89

    Dhavendra Kumar. (2007) Genomic medicine: a new frontier of medicine in the twenty first century. Genomic Medicine 1:1-2, 3-7
    CrossRef

  90. 90

    Carl Hanson, Lelinneth Novilla, Michael Barnes, Natalie De La Cruz, Aaron Meacham. (2007) Using Family Health History for Chronic Disease Prevention in the Age of Genomics. American Journal of Health Education 38:4, 219-229
    CrossRef

  91. 91

    Jeffrey L. Anderson, John F. Carlquist, Benjamin D. Horne, Paul N. Hopkins. (2007) Progress in unraveling the genetics of coronary artery disease and myocardial infarction. Current Atherosclerosis Reports 9:3, 179-186
    CrossRef

  92. 92

    Siobhan Dolan, Janis Biermann, Karla Damus. (2007) Genomics for Health in Preconception and Prenatal Periods. Journal of Nursing Scholarship 39:1, 4-9
    CrossRef

  93. 93

    Jean Jenkins, Kathleen A. Calzone. (2007) Establishing the Essential Nursing Competencies for Genetics and Genomics. Journal of Nursing Scholarship 39:1, 10-16
    CrossRef

  94. 94

    Brenton Louie, Peter Mork, Fernando Martin-Sanchez, Alon Halevy, Peter Tarczy-Hornoch. (2007) Data integration and genomic medicine. Journal of Biomedical Informatics 40:1, 5-16
    CrossRef

  95. 95

    Wolf Rogowski. (2007) Current impact of gene technology on healthcare. Health Policy 80:2, 340-357
    CrossRef

  96. 96

    Peter Tarczy-Hornoch, Mia K. Markey, John A. Smith, Tadaaki Hiruki. (2007) Bio*Medical informatics and genomic medicine: Research and training. Journal of Biomedical Informatics 40:1, 1-4
    CrossRef

  97. 97

    D Ghosh, M A Skinner, W A Laing. (2007) Pharmacogenomics and nutrigenomics: synergies and differences. European Journal of Clinical Nutrition
    CrossRef

  98. 98

    Minhaz Uddin Ahmed, Koutarou Idegami, Miyuki Chikae, Kagan Kerman, Piyasak Chaumpluk, Shohei Yamamura, Eiichi Tamiya. (2007) Electrochemical DNA biosensor using a disposable electrochemical printed (DEP) chip for the detection of SNPs from unpurified PCR amplicons. The Analyst 132:5, 431
    CrossRef

  99. 99

    Angela M. Brand, Nicole M. Probst-Hensch. (2007) Biobanking for Epidemiological Research and Public Health. Pathobiology 74:4, 227-238
    CrossRef

  100. 100

    HP Rang, MM Dale, JM Ritter, RJ Flower. Biopharmaceuticals and gene therapy. In: Rang & Dale's Pharmacology. Elsevier, 2007:770-780.
    CrossRef

  101. 101

    Robert L. Nussbaum, Roderick R. McInnes, Huntington F. Willard, Ada Hamosh. Introduction. In: Thompson & Thompson Genetics in Medicine. Elsevier, 2007:1-3.
    CrossRef

  102. 102

    Helen N. Lyon, Bruce R. Korf. Human genome project, genomics, and clinical research. In: Principles and Practice of Clinical Research. Elsevier, 2007:405-420.
    CrossRef

  103. 103

    Miriam Kelley Bullard, Mini Swift, Alden Harken. (2007) Genomics and the Cardiac Surgeon. Journal of Cardiac Surgery 22:1, 83-86
    CrossRef

  104. 104

    Kathleen A. Calzone, Dale Halsey Lea, Agnes Masny. (2006) Non-Hodgkin's Lymphoma as an Exemplar of the Effects of Genetics and Genomics. Journal of Nursing Scholarship 38:4, 335-343
    CrossRef

  105. 105

    Matthew R.G. Taylor, Janice G. Edwards, Lisa Ku. (2006) Lost in transition: Challenges in the expanding field of adult genetics. American Journal of Medical Genetics Part C: Seminars in Medical Genetics 142C:4, 294-303
    CrossRef

  106. 106

    Rodney Harris. Genetic Services: Access. In: Encyclopedia of Life Sciences. John Wiley & Sons, Ltd, 2006.
    CrossRef

  107. 107

    Dale Halsey Lea, Janet K. Williams, Judith A. Cooksey, Patricia A. Flanagan, Gaetano Forte, Miriam G. Blitzer. (2006) U.S. Genetics Nurses in Advanced Practice. Journal of Nursing Scholarship 38:3, 213-218
    CrossRef

  108. 108

    David Seo, Geoffrey S. Ginsburg, Pascal J. Goldschmidt-Clermont. (2006) Gene Expression Analysis of Cardiovascular Diseases. Journal of the American College of Cardiology 48:2, 227-235
    CrossRef

  109. 109

    Alexandre Loktionov. Nutrigenomics and Pharmacogenomics of Human Cancer. CRC Press, 2006:261-317.
    CrossRef

  110. 110

    C. Michael Cotten, Geoffrey S. Ginsburg, Ronald N. Goldberg, Marcy C. Speer. (2006) Genomic analyses: A neonatology perspective. The Journal of Pediatrics 148:6, 720-726
    CrossRef

  111. 111

    Mark J. Millan. (2006) Multi-target strategies for the improved treatment of depressive states: Conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacology & Therapeutics 110:2, 135-370
    CrossRef

  112. 112

    &NA;. (2006) NNSDO Update. Journal for Nurses in Staff Development (JNSD) 22:3, 160-161
    CrossRef

  113. 113

    William L. Carroll, Deepa Bhojwani, Dong-Joon Min, Naomi Moskowitz, Elizabeth A. Raetz. (2006) Childhood acute lymphoblastic leukemia in the age of genomics. Pediatric Blood & Cancer 46:5, 570-578
    CrossRef

  114. 114

    Holly C. Gooding, Kurt Organista, Jeffrey Burack, Barbara Bowles Biesecker. (2006) Genetic susceptibility testing from a stress and coping perspective. Social Science & Medicine 62:8, 1880-1890
    CrossRef

  115. 115

    Eric Racine, Isabelle Gareau, Hubert Doucet, Danielle Laudy, Guy Jobin, Pamela Schraedley-Desmond. (2006) Hyped biomedical science or uncritical reporting? Press coverage of genomics (1992–2001) in Québec. Social Science & Medicine 62:5, 1278-1290
    CrossRef

  116. 116

    Terrie E. Moffitt, Avshalom Caspi, Michael Rutter. (2006) Measured Gene-Environment Interactions in Psychopathology. Concepts, Research Strategies, and Implications for Research, Intervention, and Public Understanding of Genetics. Perspectives on Psychological Science 1:1, 5-27
    CrossRef

  117. 117

    Peter Oehr. (2006) ‘Omics’-based imaging in cancer detection and therapy. Personalized Medicine 3:1, 19-32
    CrossRef

  118. 118

    Cathy Fomous, Joyce A. Mitchell, Alexa McCray. (2006) ‘Genetics Home Reference’: Helping Patients Understand the Role of Genetics in Health and Disease. Community Genetics 9:4, 274-278
    CrossRef

  119. 119

    Andrew C. Ahn, Muneesh Tewari, Chi-Sang Poon, Russell S. Phillips. (2006) The Limits of Reductionism in Medicine: Could Systems Biology Offer an Alternative?. PLoS Medicine 3:6, e208
    CrossRef

  120. 120

    Gloria R Grice, Terry L Seaton, Abigail M Woodland, Howard L McLeod. (2006) Defining the opportunity for pharmacogenetic intervention in primary care. Pharmacogenomics 7:1, 61-65
    CrossRef

  121. 121

    Dale H. LEA, Judith A. COOKSEY, Patricia A. FLANAGAN, Janet K. WILLIAMS, Gaetano FORTE. (2005) Innovations in United States genetics nursing: Practice and research. Japan Journal of Nursing Science 2:2, 71-83
    CrossRef

  122. 122

    Ancha Baranova, Karen Schlauch, Shobha Gowder, Rochelle Collantes, Vikas Chandhoke, Zobair M. Younossi. (2005) Microarray technology in the study of obesity and non-alcoholic fatty liver disease. Liver International 25:6, 1091-1096
    CrossRef

  123. 123

    Carol Estwing Ferrans, Julie Johnson Zerwic, Jo Ellen Wilbur, Janet L. Larson. (2005) Conceptual Model of Health-Related Quality of Life. Journal of Nursing Scholarship 37:4, 336-342
    CrossRef

  124. 124

    James C. Thomas, Debra E. Irwin, Erin Shaugnessy Zuiker, Robert C. Millikan. (2005) Genomics and the Public Health Code of Ethics. American Journal of Public Health 95:12, 2139-2143
    CrossRef

  125. 125

    Levy, Harvey L., . (2005) Metabolic Disorders in the Center of Genetic Medicine. New England Journal of Medicine 353:18, 1968-1970
    Full Text

  126. 126

    Konstantinos N. Lazaridis, Brian D. Juran. (2005) American Gastroenterological Association Future Trends Committee Report: The Application of Genomic and Proteomic Technologies to Digestive Disease Diagnosis and Treatment and Their Likely Impact on Gastroenterology Clinical Practice. Gastroenterology 129:5, 1720-1752
    CrossRef

  127. 127

    Denis F. Kinane, Hideki Shiba, Thomas C. Hart. (2005) The genetic basis of periodontitis. Periodontology 2000 39:1, 91-117
    CrossRef

  128. 128

    Ioannis S Vizirianakis. (2005) Improving pharmacotherapy outcomes by pharmacogenomics: from expectation to reality?. Pharmacogenomics 6:7, 701-711
    CrossRef

  129. 129

    Ramón Cacabelos. (2005) Pharmacogenomics, nutrigenomics and therapeutic optimization in Alzheimer’s disease. Aging Health 1:2, 303-348
    CrossRef

  130. 130

    Zobair M. Younossi, Ancha Baranova, Katharine Ziegler, Luca Del Giacco, Karen Schlauch, Timothy L. Born, Hazem Elariny, Francesco Gorreta, Amy VanMeter, Abraham Younoszai, Janus P. Ong, Zachary Goodman, Vikas Chandhoke. (2005) A genomic and proteomic study of the spectrum of nonalcoholic fatty liver disease. Hepatology 42:3, 665-674
    CrossRef

  131. 131

    Zobair M. Younossi, Francesco Gorreta, Janus P. Ong, Karen Schlauch, Luca Del Giacco, Hazem Elariny, Amy Van Meter, Abraham Younoszai, Zachary Goodman, Anna Baranova, Alan Christensen, Geraldine Grant, Vikas Chandhoke. (2005) Hepatic gene expression in patients with obesity-related non-alcoholic steatohepatitis. Liver International 25:4, 760-771
    CrossRef

  132. 132

    Evelyn Tiffany-Castiglioni, Vijayanagaram Venkatraj, Yongchang Qian. (2005) Genetic Polymorphisms and Mechanisms of Neurotoxicity: Overview. NeuroToxicology 26:4, 641-649
    CrossRef

  133. 133

    Raquel Iniesta, Elisabet Guinó, Víctor Moreno. (2005) Análisis estadístico de polimorfismos genéticos en estudios epidemiológicos. Gaceta Sanitaria 19:4, 333-341
    CrossRef

  134. 134

    E. Berntorp, J. Astermark, S. M. Donfield, G. W. Nelson, J. Oldenburg, A. D. Shapiro, D. M. Dimichele, B. M. Ewenstein, E. D. Gomperts, C. A. Winkler, . (2005) Haemophilia Inhibitor Genetics Study - evaluation of a model for studies of complex diseases using linkage and association methods. Haemophilia 11:4, 427-429
    CrossRef

  135. 135

    Kirsty Challen, Hilary J Harris, Claire Julian-Reynier, Leo P ten Kate, Ulf Kristoffersson, Irmgard Nippert, Jörg Schmidtke, Caroline Benjamin, Rodney Harris. (2005) Genetic education and nongenetic health professionals: Educational providers and curricula in Europe. Genetics in Medicine 7:5, 302-310
    CrossRef

  136. 136

    Suzanne Feetham, Elizabeth J. Thomson, Ada Sue Hinshaw. (2005) Nursing Leadership in Genomics for Health and Society. Journal of Nursing Scholarship 37:2, 102-110
    CrossRef

  137. 137

    Jean Jenkins, Patricia A. Grady, Francis S. Collins. (2005) Nurses and the Genomic Revolution. Journal of Nursing Scholarship 37:2, 98-101
    CrossRef

  138. 138

    Ruth M. DeBusk. (2005) Editorial. Nutrition & Dietetics 62:2-3, 63-65
    CrossRef

  139. 139

    Teena M. McGuinness, Patricia Noonan, Janyce G. Dyer. (2005) Family History as a Tool for Psychiatric Nurses. Archives of Psychiatric Nursing 19:3, 116-124
    CrossRef

  140. 140

    Cynthia A. Moore, Muin J. Khoury, Linda A. Bradley. (2005) From Genetics to Genomics: Using Gene-Based Medicine to Prevent Disease and Promote Health in Children. Seminars in Perinatology 29:3, 135-143
    CrossRef

  141. 141

    Amanda A. Fox, Stanton K. Shernan, Simon C. Body, Charles D. Collard. (2005) Genetic Influences on Cardiac Surgical Outcomes. Journal of Cardiothoracic and Vascular Anesthesia 19:3, 379-391
    CrossRef

  142. 142

    M.J.E. Rijn, C.M. Duijn, A.J.C. Slooter. (2005) The impact of genetic testing on complex diseases. European Journal of Epidemiology 20:5, 383-388
    CrossRef

  143. 143

    Konstantinos N. Lazaridis, Gloria M. Petersen. (2005) Genomics, genetic epidemiology, and genomic medicine. Clinical Gastroenterology and Hepatology 3:4, 320-328
    CrossRef

  144. 144

    Ronald J Trent. DNA, RNA, Genes and Chromosomes. In: Molecular Medicine. Elsevier, 2005:17-I.
    CrossRef

  145. 145

    Vivian Ota Wang, Stanley Sue. (2005) In the Eye of the Storm: Race and Genomics in Research and Practice.. American Psychologist 60:1, 37-45
    CrossRef

  146. 146

    Allen C. Nunnally, Christopher J. Webster, Scott A. Brown, Gary A. Cohen. (2005) Genetic Patent Protection in the Pharmaceutical and Biotechnology Industries. Community Genetics 8:4, 209-216
    CrossRef

  147. 147

    Marc D. Schwartz, Beth N. Peshkin, Kenneth P. Tercyak, Kathryn L. Taylor, Heiddis Valdimarsdottir. (2005) Decision Making and Decision Support for Hereditary Breast-Ovarian Cancer Susceptibility.. Health Psychology 24:4, Suppl, S78-S84
    CrossRef

  148. 148

    F. Rodríguez de Castro, J. Solé-Violán, J.C. Rodríguez-Gallego. (2005) Variabilidad genética en la susceptibilidad y en la gravedad de la neumonía. Archivos de Bronconeumología 41, 21-29
    CrossRef

  149. 149

    DeWayne M. Pursley, Gary A. Silverman. Impact of the Human Genome Project on Neonatal Care. In: Avery's Diseases of the Newborn. Elsevier, 2005:171-185.
    CrossRef

  150. 150

    James A. Marcum. (2005) Biomechanical and phenomenological models of the body, the meaning of illness and quality of care. Medicine, Health Care and Philosophy 7:3, 311-320
    CrossRef

  151. 151

    Charles Gawad. (2005) Towards Molecular Medicine. American Journal of PharmacoGenomics 5:4, 207-211
    CrossRef

  152. 152

    Regina E. Ensenauer, Virginia V. Michels, Shanda S. Reinke. (2005) Genetic Testing: Practical, Ethical, and Counseling Considerations. Mayo Clinic Proceedings 80:1, 63-73
    CrossRef

  153. 153

    Julian Little, Linda Sharp, Muin J Khoury, Linda Bradley, Marta Gwinn. (2005) The Epidemiologic Approach to Pharmacogenomics. American Journal of PharmacoGenomics 5:1, 1-20
    CrossRef

  154. 154

    Ronald J Trent. Genomics, Proteomics and Bioinformatics. In: Molecular Medicine. Elsevier, 2005:119-III.
    CrossRef

  155. 155

    Bertrand L. Jaber, Brian J.G. Pereira, Joseph V. Bonventre, Vaidyanathapuram S. Balakrishnan. (2005) Polymorphism of host response genes: Implications in the pathogenesis and treatment of acute renal failure. Kidney International 67:1, 14-33
    CrossRef

  156. 156

    Kathleen Ries Merikangas, Nancy C. P. Low. (2004) The epidemiology of mood disorders. Current Psychiatry Reports 6:6, 411-421
    CrossRef

  157. 157

    R. Shah, B. Darne, D. Atar, E. Abadie, K.F. Adams, F. Zannad, . (2004) Pharmacogenomics in cardiovascular clinical trials. Fundamental and Clinical Pharmacology 18:6, 705-708
    CrossRef

  158. 158

    Lieve Nuytinck, Fred Shapiro. (2004) Mannose-binding lectin: laying the stepping stones from clinical research to personalized medicine. Personalized Medicine 1:1, 35-52
    CrossRef

  159. 159

    Laura M. Beskow, Jeffrey R. Botkin, Mary Daly, Eric T. Juengst, Lisa Soleymani Lehmann, Jon F. Merz, Rebecca Pentz, Nancy A. Press, Lainie Friedman Ross, Jeremy Sugarman, Lisa R. Susswein, Sharon F. Terry, Melissa A. Austin, Wylie Burke. (2004) Ethical issues in identifying and recruiting participants for familial genetic research. American Journal of Medical Genetics 130A:4, 424-431
    CrossRef

  160. 160

    K. Schreiber, T. Girard, C. H. Kindler. (2004) Bibliometric analysis of original molecular biology research in anaesthesia. Anaesthesia 59:10, 1002-1007
    CrossRef

  161. 161

    SHARON D. HORNER. (2004) Ethics and Genetics. Clinical Nurse Specialist 18:5, 228-231
    CrossRef

  162. 162

    Debra L. Schutte. (2004) The evolving role of genomics in shaping care for persons with dementia. Nursing Clinics of North America 39:3, 581-592
    CrossRef

  163. 163

    B. Godard, J. Marshall, C. Laberge, B. M. Knoppers. (2004) Strategies for consulting with the community: The cases of four large-scale genetic databases. Science and Engineering Ethics 10:3, 457-477
    CrossRef

  164. 164

    W.Gregory Feero. (2004) Genetic thrombophilia. Primary Care: Clinics in Office Practice 31:3, 685-709
    CrossRef

  165. 165

    Kathleen A. Calzone, Agnes Masny. (2004) Genetics and oncology nursing. Seminars in Oncology Nursing 20:3, 178-185
    CrossRef

  166. 166

    Stefano Rossetti, André T Hoogeveen, Nicoletta Sacchi. (2004) The MTG proteins: chromatin repression players with a passion for networking. Genomics 84:1, 1-9
    CrossRef

  167. 167

    Jose D Debes, Raul Urrutia. (2004) Bioinformatics tools to understand human diseases. Surgery 135:6, 579-585
    CrossRef

  168. 168

    Barkur S. Shastry. (2004) Role of SNP/haplotype map in gene discovery and drug development: An overview. Drug Development Research 62:2, 143-150
    CrossRef

  169. 169

    SG Suther, P Goodson. (2004) Texas physicians' perceptions of genomic medicine as an innovation. Clinical Genetics 65:5, 368-377
    CrossRef

  170. 170

    Richard R. Sharp, Michael A. Yudell, Samuel H. Wilson. (2004) Science and society: Shaping science policy in the age of genomics. Nature Reviews Genetics 5:4, 311-315
    CrossRef

  171. 171

    Sharon D. Horner, Elizabeth Abel, Kay Taylor, Delores Sands. (2004) Using theory to guide the diffusion of genetics content in nursing curricula. Nursing Outlook 52:2, 80-84
    CrossRef

  172. 172

    Moshe Zilberstein. (2004) Mining the human genome for new health therapies. Fertility and Sterility 81, 4-8
    CrossRef

  173. 173

    David A. Fishbain, Dana Fishbain, John Lewis, R. B. Cutler, Brandly Cole, H. L. Rosomoff, R. Steele Rosomoff. (2004) Genetic Testing for Enzymes of Drug Metabolism: Does It Have Clinical Utility for Pain Medicine at the Present Time? A Structured Review. Pain Medicine 5:1, 81-93
    CrossRef

  174. 174

    Ronald JA Trent, Bing Yu, Melody Caramins. (2004) Challenges for clinical genetic DNA testing. Expert Review of Molecular Diagnostics 4:2, 201-208
    CrossRef

  175. 175

    Sonja A. Rasmussen, Cynthia A. Moore. (2004) Public health approach to birth defects, developmental disabilities, and genetic conditions. American Journal of Medical Genetics 125C:1, 1-3
    CrossRef

  176. 176

    F Martin-Sanchez, I Iakovidis, S Nørager, V Maojo, P de Groen, J Van der Lei, T Jones, K Abraham-Fuchs, R Apweiler, A Babic, R Baud, V Breton, P Cinquin, P Doupi, M Dugas, R Eils, R Engelbrecht, P Ghazal, P Jehenson, C Kulikowski, K Lampe, G De Moor, S Orphanoudakis, N Rossing, B Sarachan, A Sousa, G Spekowius, G Thireos, G Zahlmann, J Zvárová, I Hermosilla, F.J Vicente. (2004) Synergy between medical informatics and bioinformatics: facilitating genomic medicine for future health care. Journal of Biomedical Informatics 37:1, 30-42
    CrossRef

  177. 177

    Joseph Pearlberg, Joshua LaBaer. (2004) Protein expression clone repositories for functional proteomics. Current Opinion in Chemical Biology 8:1, 98-102
    CrossRef

  178. 178

    Theresa A. Beery, Marcia J. Hern. (2004) Genetic Practice, Education, and Research. Clinical Nurse Specialist 18:3, 126-132
    CrossRef

  179. 179

    Kari Hemminki, Xinjun Li, Kamila Czene. (2004) Familial risk of cancer: Data for clinical counseling and cancer genetics. International Journal of Cancer 108:1, 109-114
    CrossRef

  180. 180

    Stephanie J. Green, Donna K. Pauler. (2004) Statistics in clinical trials. Current Oncology Reports 6:1, 36-41
    CrossRef

  181. 181

    Sandrine Guis, Jean-Pierre Mattéi, Frédéric Lioté. (2003) Drug-induced and toxic myopathies. Best Practice & Research Clinical Rheumatology 17:6, 877-907
    CrossRef

  182. 182

    Dale Halsey Lea. (2003) How genetics changes daily practice. Nursing Management (Springhouse) 34:11, 19-24
    CrossRef

  183. 183

    Manuel Favre, Christophe Butticaz, Brian Stevenson, C. Victor Jongeneel, Amalio Telenti. (2003) High Frequency of Alternative Splicing of Human Genes Participating in the HIV-1 Life Cycle. JAIDS Journal of Acquired Immune Deficiency Syndromes 34:2, 127-133
    CrossRef

  184. 184

    Guttmacher, Alan E., Collins, Francis S., . (2003) Welcome to the Genomic Era. New England Journal of Medicine 349:10, 996-998
    Free Full Text

  185. 185

    Guttmacher, Alan E., Collins, Francis S., , Burke, Wylie, . (2003) Genomics as a Probe for Disease Biology. New England Journal of Medicine 349:10, 969-974
    Free Full Text

  186. 186

    David Schottenfeld. (2003) “What's past is prologue”: a tribute to Abraham Lilienfeld. Annals of Epidemiology 13:8, 549-551
    CrossRef

  187. 187

    Jun Yoshioka, Richard T. Lee. (2003) Cardiovascular genomics. Cardiovascular Pathology 12:5, 249-254
    CrossRef

  188. 188

    Ramón Cacabelos. (2003) The application of functional genomics to Alzheimer’s disease. Pharmacogenomics 4:5, 597-621
    CrossRef

  189. 189

    Robert E. Taylor. (2003) Pharmacological and Cultural Considerations in Alcohol Treatment Clinical Trials: Issues in Clinical Research Related to Race and Ethnicity. Alcoholism: Clinical & Experimental Research 27:8, 1345-1348
    CrossRef

  190. 190

    Randy Todd, Merideth August. (2003) Molecular approaches to the diagnosis of sporadic and nevoid basal cell carcinoma syndrome-associated odontogenic keratocysts. Oral and Maxillofacial Surgery Clinics of North America 15:3, 447-461
    CrossRef

  191. 191

    Joann Genovich-Richards. (2003) Interview with a Quality Leader: Dale Schumacher on Strategic Quality Planning and Research. Journal for Healthcare Quality 25:4, 16-39
    CrossRef

  192. 192

    Basil Hanss, Leslie A. Bruggeman. (2003) Applications of gene therapy to kidney disease. Current Opinion in Nephrology and Hypertension 12:4, 439-445
    CrossRef

  193. 193

    Regina E. Ensenauer, Shanda S. Reinke, Michael J. Ackerman, David J. Tester, David A.H. Whiteman, Ayalew Tefferi. (2003) Primer on Medical Genomics Part VIII: Essentials of Medical Genetics for the Practicing Physician. Mayo Clinic Proceedings 78:7, 846-857
    CrossRef

  194. 194

    Muin J. Khoury. (2003) Genetics and genomics in practice: The continuum from genetic disease to genetic information in health and disease. Genetics in Medicine 5:4, 261-268
    CrossRef

  195. 195

    Luisa Mestroni. (2003) Genomic medicine and atrial fibrillation. Journal of the American College of Cardiology 41:12, 2193-2196
    CrossRef

  196. 196

    Robert J Zagursky, Stephen B Olmsted, David P Russell, Joe L Wooters. (2003) Bioinformatics: how it is being used to identify bacterial vaccine candidates. Expert Review of Vaccines 2:3, 417-436
    CrossRef

  197. 197

    Kathryn A Phillips, David Veenstra, Stephanie Van Bebber, Julie Sakowski. (2003) An introduction to cost-effectiveness and cost–benefit analysis of pharmacogenomics. Pharmacogenomics 4:3, 231-239
    CrossRef

  198. 198

    Seth E Dobrin, Dietrich A Stephan. (2003) Integrating microarrays into disease-gene identification strategies. Expert Review of Molecular Diagnostics 3:3, 375-385
    CrossRef

  199. 199

    Vincent Mooser, Jose M. Ordovas. (2003) ‘Omic’ approaches and lipid metabolism: are these new technologies holding their promises?. Current Opinion in Lipidology 14:2, 115-119
    CrossRef

  200. 200

    Jesús Villar, Carlos Flores, Sebastián Méndez-Alvarez. (2003) Genetic susceptibility to acute lung injury. Critical Care Medicine 31:Supplement, S272-S275
    CrossRef

  201. 201

    Rasih Atilla Ener, Sharon B. Meglathery, William A. Van Decker, Rollin M. Gallagher. (2003) Serotonin Syndrome and Other Serotonergic Disorders. Pain Medicine 4:1, 63-74
    CrossRef

  202. 202

    Quanhe Yang, Muin J. Khoury, Lorenzo Botto, J.M. Friedman, W. Dana Flanders. (2003) Improving the Prediction of Complex Diseases by Testing for Multiple Disease-Susceptibility Genes. The American Journal of Human Genetics 72:3, 636-649
    CrossRef

  203. 203

    (2003) Genomic Medicine. New England Journal of Medicine 348:8, 759-760
    Free Full Text

  204. 204

    Goldstein, David B., . (2003) Pharmacogenetics in the Laboratory and the Clinic. New England Journal of Medicine 348:6, 553-556
    Free Full Text

  205. 205

    Phyllis Griffin Epps. (2003) Policy Before Practice. American Journal of PharmacoGenomics 3:6, 405-418
    CrossRef

  206. 206

    K.R. MERIKANGAS. (2003) Implications of Genomics for Public Health: The Role of Genetic Epidemiology. Cold Spring Harbor Symposia on Quantitative Biology 68:0, 359-364
    CrossRef

  207. 207

    Varmus, Harold, . (2002) Getting Ready for Gene-Based Medicine. New England Journal of Medicine 347:19, 1526-1527
    Free Full Text

Glossary

Glossary

  1. Allele:

    — An alternative form of a gene.

  2. Alternative splicing:

    — A regulatory mechanism by which variations in the incorporation of a gene's exons, or coding regions, into messenger RNA lead to the production of more than one related protein, or isoform.

  3. Autosomes:

    — All of the chromosomes except for the sex chromosomes and the mitochondrial chromosome.

  4. Centromere:

    — The constricted region near the center of a chromosome that has a critical role in cell division.

  5. Codon:

    — A three-base sequence of DNA or RNA that specifies a single amino acid.

  6. Conservative mutation:

    — A change in a DNA or RNA sequence that leads to the replacement of one amino acid with a biochemically similar one.

  7. Epigenetic:

    — A term describing nonmutational phenomena, such as methylation and histone modification, that modify the expression of a gene.

  8. Exon:

    — A region of a gene that codes for a protein.

  9. Frame-shift mutation:

    — The addition or deletion of a number of DNA bases that is not a multiple of three, thus causing a shift in the reading frame of the gene. This shift leads to a change in the reading frame of all parts of the gene that are downstream from the mutation, often leading to a premature stop codon and ultimately, to a truncated protein.

  10. Gain-of-function mutation:

    — A mutation that produces a protein that takes on a new or enhanced function.

  11. Genomics:

    — The study of the functions and interactions of all the genes in the genome, including their interactions with environmental factors.

  12. Genotype:

    — A person's genetic makeup, as reflected by his or her DNA sequence.

  13. Haplotype:

    — A group of nearby alleles that are inherited together.

  14. Heterozygous:

    — Having two different alleles at a specific autosomal (or X chromosome in a female) gene locus.

  15. Homozygous:

    — Having two identical alleles at a specific autosomal (or X chromosome in a female) gene locus.

  16. Intron:

    — A region of a gene that does not code for a protein.

  17. Linkage disequilibrium:

    — The nonrandom association in a population of alleles at nearby loci.

  18. Loss-of-function mutation:

    — A mutation that decreases the production or function of a protein (or does both).

  19. Missense mutation:

    — Substitution of a single DNA base that results in a codon that specifies an alternative amino acid.

  20. Monogenic:

    — Caused by a mutation in a single gene.

  21. Motif:

    — A DNA-sequence pattern within a gene that, because of its similarity to sequences in other known genes, suggests a possible function of the gene, its protein product, or both.

  22. Multifactorial:

    — Caused by the interaction of multiple genetic and environmental factors.

  23. Nonconservative mutation:

    — A change in the DNA or RNA sequence that leads to the replacement of one amino acid with a very dissimilar one.

  24. Nonsense mutation:

    — Substitution of a single DNA base that results in a stop codon, thus leading to the truncation of a protein.

  25. Penetrance:

    — The likelihood that a person carrying a particular mutant gene will have an altered phenotype.

  26. Phenotype:

    — The clinical presentation or expression of a specific gene or genes, environmental factors, or both.

  27. Point mutation:

    — The substitution of a single DNA base in the normal DNA sequence.

  28. Regulatory mutation:

    — A mutation in a region of the genome that does not encode a protein but affects the expression of a gene.

  29. Repeat sequence:

    — A stretch of DNA bases that occurs in the genome in multiple identical or closely related copies.

  30. Silent mutation:

    — Substitution of a single DNA base that produces no change in the amino acid sequence of the encoded protein.

  31. Single-nucleotide polymorphism (SNP):

    — A common variant in the genome sequence; the human genome contains about 10 million SNPs.

  32. Stop codon:

    — A codon that leads to the termination of a protein rather than to the addition of an amino acid. The three stop codons are TGA, TAA, and TAG.

Letters

Trends

Most Viewed (Last Week)