Genomics Research: Livestock Production

Similar documents
Fundamentals of Genomic Selection

The what, why and how of Genomics for the beef cattle breeder

Understanding genomic selection in poultry breeding

Domestic animal genomes meet animal breeding translational biology in the ag-biotech sector. Jerry Taylor University of Missouri-Columbia

QTL Mapping, MAS, and Genomic Selection

Whole Genome-Assisted Selection. Alison Van Eenennaam, Ph.D. Cooperative Extension Specialist

Traditional Genetic Improvement. Genetic variation is due to differences in DNA sequence. Adding DNA sequence data to traditional breeding.

The promise of genomics for animal improvement. Daniela Lourenco

J.P. Gibson The Institute for Genetics and Bioinformatics University of New England Australia

Genomic selection in the Australian sheep industry

Sequencing the Swine Genome: Progress and Prospects

From Genotype to Phenotype

2010 Annual Research Report

Implementing direct and indirect markers.

Obiettivi e recenti acquisizioni del progetto di ricerca SelMol

DNA Technologies and Production Markers

Marker Assisted Selection Where, When, and How. Lecture 18

Strategy for Applying Genome-Wide Selection in Dairy Cattle

High-density SNP Genotyping Analysis of Broiler Breeding Lines

Validating Genetic Markers

Question. In the last 100 years. What is Feed Efficiency? Genetics of Feed Efficiency and Applications for the Dairy Industry

BLUP and Genomic Selection

Introduction to Animal Breeding & Genomics

Ecological genomics and molecular adaptation: state of the Union and some research goals for the near future.

Strategy for applying genome-wide selection in dairy cattle

Genomic selection for sheep and beef systems, dispelling the myths for farmers. Dewi Jones January 2016

Genomic management of animal genetic diversity

Could modern methods of genetics improve the disease resistance in farm animals?

Melding genomics and quantitative genetics in sheep breeding programs: opportunities and limits

SNPs - GWAS - eqtls. Sebastian Schmeier

ANIMAL SCIENCE (ANSI)

Optimizing Traditional and Marker Assisted Evaluation in Beef Cattle

DESIGNS FOR QTL DETECTION IN LIVESTOCK AND THEIR IMPLICATIONS FOR MAS

Genetic dissection of complex traits, crop improvement through markerassisted selection, and genomic selection

A journey: opportunities & challenges of melding genomics into U.S. sheep breeding programs

SolCAP. Executive Commitee : David Douches Walter De Jong Robin Buell David Francis Alexandra Stone Lukas Mueller AllenVan Deynze

latestdevelopments relevant for the Ag sector André Eggen Agriculture Segment Manager, Europe

Genetics: Past, Present and Future

Marker types. Potato Association of America Frederiction August 9, Allen Van Deynze

Addressing the Phenomic Gap: Next generation genomic tools in beef

AGRO/ANSC/BIO/GENE/HORT 305 Fall, 2016 Overview of Genetics Lecture outline (Chpt 1, Genetics by Brooker) #1

Lecture 2: Height in Plants, Animals, and Humans. Michael Gore lecture notes Tucson Winter Institute version 18 Jan 2013

I.1 The Principle: Identification and Application of Molecular Markers

Genetics Effective Use of New and Existing Methods

Genomic selection applies to synthetic breeds

Genomic Selection in Breeding Programs BIOL 509 November 26, 2013

Genetics of Beef Cattle: Moving to the genomics era Matt Spangler, Assistant Professor, Animal Science, University of Nebraska-Lincoln

11/30/11. Lb output/beef cow. Corn Yields. Corn and Wheat

QTL mapping in domesticated and natural fish populations

Genomic information a tool for Assessment of Genetic diversity in Mithun

Crash-course in genomics

Gene Mapping in Natural Plant Populations Guilt by Association

QTL Mapping Using Multiple Markers Simultaneously

Marker-assisted selection in livestock case studies

Got Dairy? A brief introduction to dairy cattle genetics

The 150+ Tomato Genome (re-)sequence Project; Lessons Learned and Potential

Genomic Resources and Gene/QTL Discovery in Livestock

Trudy F C Mackay, Department of Genetics, North Carolina State University, Raleigh NC , USA.

Genomic Selection in Sheep Breeding Programs

RECOLAD. Introduction to the «atelier 1» Genetic approaches to improve adaptation to climate change in livestock

Human Chromosomes Section 14.1

A transmission disequilibrium test approach to screen for quantitative trait loci in two selected lines of Large White pigs

Revolutionizing genetics in the cattle industry. Extensive toolset. Proven technology. Collaborative approach.

Using Genomics to Improve the Genetic Potential and Management of Your Herd

Lecture 12. Genomics. Mapping. Definition Species sequencing ESTs. Why? Types of mapping Markers p & Types

January 1967 Denver, CO

Concepts of Genetics, 10e (Klug/Cummings/Spencer/Palladino) Chapter 1 Introduction to Genetics

LowInputBreeds & Genomic breeding

THE STUDY OF GENETICS is extremely

INTRODUCTION TO MOLECULAR GENETICS. Andrew McQuillin Molecular Psychiatry Laboratory UCL Division of Psychiatry 22 Sept 2017

GREG GIBSON SPENCER V. MUSE

Midterm 1 Results. Midterm 1 Akey/ Fields Median Number of Students. Exam Score

Section 4 - Guidelines for DNA Technology. Version October, 2017

Application of Modern Genetic Technologies to Improve Efficiency of Pig Production and Pork Quality

UNIT MOLECULAR GENETICS AND BIOTECHNOLOGY

Using RNAseq data to improve genomic selection in dairy cattle

GENETIC ENGINEERING OF LIVESTOCK IMPROVEMENT. Gordon E. Dickerson*, D.S.A. SUMMARY

Human Genomics. 1 P a g e

Genome wide association and genomic selection to speed up genetic improvement for meat quality in Hanwoo

TSB Collaborative Research: Utilising i sequence data and genomics to improve novel carcass traits in beef cattle

material, programs and services that assist our customers in maximizing profitability. The success of

Practical integration of genomic selection in dairy cattle breeding schemes

Quantitative Genetics

The use of multi-breed reference populations and multi-omic data to maximize accuracy of genomic prediction

Decoding genomic selection and the benefit for unconventional traits

Module 1 Principles of plant breeding

Developments in the Genetic Improvement of a Large Commercial Population in the New Zealand Sheep Industry

Use of New Breeding Techniques applied to cattle

Chapter 2 Genetic Control of Meat Quality Traits

2018 ICBF and Sheep Ireland Genetics Conference 20 th Anniversary. Dorian Garrick

Genetics of dairy production

DNA & DNA Replication

Practical value of genomics for the larger livestock industry in South Africa

Course Syllabus for FISH/CMBL 7660 Fall 2008

Lesson Overview. Studying the Human Genome. Lesson Overview Studying the Human Genome

Genetics Test. Multiple Choice Identify the choice that best completes the statement or answers the question.

The use of multi-breed reference populations and multi-omic data to maximize accuracy of genomic prediction

Carcass Traits Association with GH/AluI Gene Polymorphism in Indonesian Aceh Cattle

RP1: AN EXAMPLE OF REVERSE GENETICS APPROACH TO DESCRIBE COMMON RECESSIVE DEFECTS

CHAPTERS 16 & 17: DNA Technology

Transcription:

Genomics Research: Livestock Production Monika Sodhi Lawrence B. Schook Department of Animal Sciences, University of Illinois, Urbana, Illinois, U.S.A. INTRODUCTION Abstract The major emphasis of animal genomic research has been to increase the productivity and disease resistance ability of economically important farm animal breeds to meet the ever increasing demands of animal products. Traditionally, the approach of livestock improvement was based on the classical breeding where in the parents of the next generation were selected based on their phenotype without knowing the genetic variation underlying the traits of interest. To this end, genomic tools have helped to move away from this Black box approach and understand the complex genetic components of phenotypic variation. Through structural and comparative genomics it had become possible to identify the genomic regions/quantitative trait loci (QTL) harboring the performance altering genes. However, the QTL search followed by fine mapping could prove useful to a limited extent as detection of casual mutations underlying QTL s was time consuming and cost extensive. These limitations restricted the extensive implementation of marker assisted selection technology in the commercial breeds. Recent advances in genomic research including whole genome sequencing, expression array, and highly multiplexed and high throughput single nucleotide polymorphism (SNP) genotyping platforms have a large impact on the ability for genome mining and pinpoint genes that influence biological and economically important traits. In future, selection decision based on the genomic breeding values complemented with other quantitative genetic evaluation approaches can very well lead to improve genetic gain in livestock species. In this chapter, an effort has been made to describe the issues, scope and needs those confront the animal genomics community in today s industry driven era. Livestock science has an increased challenge to meet the food demand for an ever-expanding world population. Livestock production started 8000 10,000 years ago with the domestication of various livestock species. Along with domestication, livestock improvement was attempted following a classical breeding approach where parents of the next generation were selected on the basis of their phenotypes. This was somewhat a black-box approach as nothing in specific (DNA sequence, gene, or underlying traits?) was known and thus, it had limitations to improve the livestock production. Simultaneously, with the rise in income and urbanization, the demand/consumption of milk and meat has been increasing tremendously (Table 1, Fig. 1). During the 1996/1998 2020 period, the projected growth rates in developing countries for milk and meat are projected to be 3.0 and 2.9% annually, respectively. In addition, by 2020 2030, world demand for meat and dairy products is expected to increase by 40 50%. [2] To increase the production of meat and milk, it is not recommended to add on the number of livestock because of the associated pollution risks. Hence, introduction of new technologies to meet the ever-increasing demand for the livestock products is of great urgency. The biological mechanisms linking genetic variation with phenotype have been the major challenge for researchers. Understanding and capturing the genetic component of phenotypic variation could play an important role in productivity enhancement in the livestock species. The molecular genetics revolution in the 1980s and 1990s led to the emergence of a new scientific discipline, genomics, resulting from the convergence of genetics, molecular biology, and bioinformatics. The objective of genomics is systematic structural and functional analysis of complex genomes to understand what individual genes do and how they interact to control biological processes. The genomic tools have helped to know how to predict sequence to consequence and harness this genetic variation to facilitate stable changes in production, fertility, and health through selection. Advances in genomics have contributed significantly toward the quality and efficiency of livestock production such as commercial strains of chickens growing at implausible rates or pigs growing considerably faster, leaner, and utilizing much less feed and with increased litter size. 310 Encyclopedia of Biotechnology in Agriculture and Food DOI: 10.1081/E-EBAF-120043005 Copyright C 2011 by Taylor & Francis. All rights reserved.

Genomics Research: Livestock Production 311 Table 1 Projected food consumption trends of various livestock products to the year 2020 in developing world. Projected growth of consumption (%/yr) Total consumption (million metric tons) Percent of total world consumption Per capita consumption (kg) 1997 2020 1997 2020 2020 1997 2020 Beef 2.9 27 47 61 6 7 Pork 2.4 47 81 67 10 13 Poultry 3.9 29 49 64 7 8 Meat 3.0 111 188 65 25 30 Milk 2.9 194 391 57 43 62 Sources: From Delgado; [1] FAOSTAT statistical database. FAO, [2] http://faostat.fao.org/ default.aspx.; ftp://ftp.fao.org/docrep/nonfao/lead/x6155e/ x6155e00.pdf. Note: Consumption indicates direct use as food, measured as uncooked weight, bone in. Meat includes beef, pork, mutton, goat, and poultry. Milk represents cattle and buffalo milk and milk products in liquid milk equivalents. Metric tons and kilograms are three-year moving averages centered on the years shown. Genomics can be grossly divided into two basic areas: 1) structural genomics; and 2) functional genomics. Structural genomics corresponds to characterization of the physical nature of whole genome and functional genomics is the understanding of the genes transcribed and translated into protein products, their interactions, and also the regulation of these processes. 400 350 300 250 200 STRUCTURAL GENOMICS Structural genomics encompasses construction of genetic maps, physical maps, comparative maps, and ultimately, the determination of the complete DNA sequence of the genome of interest with a goal to understand the genome organization and locate DNA sequence variations, i.e., 700 600 500 400 300 150 100 200 50 100 0 China India Other East Asia Other South Asia Southeast Asia Latin America Brazil Sub- Saharan Africa Developing World Developed World World 0 Projected annual growth 1997 2020 Meat %/yr Total consumption in 2020 Meat million metric tons Per capita consumption in 2020 Meat kg Projected annual growth 1997 2020 Milk %/yr Total consumption in 2020 Milk million metric tons Per capita consumption in 2020 Milk kg Fig. 1 Trend of demand vs. projected production of meat and milk. Sources: From Delgado. [1] FAO annual data 2002, total and per capita meat consumption for 1997 are annual average of 1996 1998 values. Projections are from the July 2002 version of IMPACT.

312 Genomics Research: Livestock Production Table 2 Status of quantitative trait loci identified in different livestock species. Species QTLs Traits Pig 1831 316 Cattle 1123 101 Chicken 657 112 Sheep 53 28 Source: From http://www.animalgenome.org/qtldb. polymorphisms. Structural genomics started in 1980s with the genetic mapping to identify the locations on the chromosome that might contain the performance altering genes, which however, was not a very successful approach as the sequence of the DNA was not known. The development of new techniques enabled identification of genes that underlie the genetic variation of production traits in livestock species. Although scientists have been searching for quantitative trait locus (QTL, a region that contains a gene [or locus] affecting a quantitative trait) in livestock for over 40 years, genome-wide QTL studies could not be conducted until genetic markers that spanned the entire genome were developed. For construction of high-density genetic maps, microsatellite markers (repetitive sequences) were the markers of choice because of their high polymorphism and abundant distribution throughout the genome. Microsatellite-based high-density genetic maps facilitated the fine mapping of hereditary traits of agricultural importance, characterization of meiosis, and provided a foundation for physical map construction. Resource populations required for construction of genetic maps were developed by crossing widely divergent breeds representing different alleles having large effects on QTLs. Worldwide resource populations are available for most of the livestock species and has been successfully used to identify numerous economically important trait loci (ETL) in a variety of livestock species (Table 2). The information related to livestock QTL is available in the animal quantitative trait locus database (AnimalQTLdb, http://www.animalgenome.org/qtldb). Over the years, QTL studies have contributed considerably to the development of more efficient selection procedures employing genetic markers, a strategy called marker-assisted selection (MAS). Presently several gene tests are being used in MAS programs to improve milk and meat quality, growth and reproduction in various livestock species (Table 3). QTL studies have improved the swine industry the most with several gene tests being used under MAS program for various production traits in over 60% of the pigs produced in United States. [4] However, mapping of QTL is not a very straightforward approach. It has been very difficult to search for a gene or mutation causing a QTL effect in an area surrounded by millions of unknown DNA sequences in the region. To locate these genes, the concept of comparative genomics was applied. Comparative genomics utilizes the DNA sequence similarities between species. Although the arrangement of DNA sequences is not identical across species, large regions of the genome/functional elements have been conserved throughout evolution. Hence, it is possible to harness the information available in the gene-rich mammalian species and extrapolate the same in livestock. Comparative mapping has been very effectively used to identify a number of genes in livestock. [5] The best example of the use of comparative genomics in livestock is the localization of chromosomal region containing the double muscling gene. Two research groups were able to localize the QTL effect to bovine chromosome 2 in the mid-1990s based on its location in the mouse genome. However, the specific gene was annotated in cattle by comparative mapping after the identification of myostatin gene in mice having a large impact on muscle development. The myostatin or GDF8 gene was mapped to a region of the human genome syntenic to that of bovine chromosome 2 and a single nucleotide switch from guanine to adenine at codon 313 in the gene that caused the double muscling effect was identified. Grobet et al. [6] were able to inactivate the GDF8 gene postnatally and demonstrated that these animals achieved the same level of muscle hypertrophy as animals with inactive myostatin genes. Such information is beneficial to the cattle industry as the percentage of lean in the carcass could be increased without the associated problems of dystocia because of the heavy muscled fetus. A few other genes that have been mapped in cattle through the QTL search followed by comparative fine mapping approaches include the thyroglobulin and calpastatin genes affecting the degree of marbling and meat tenderness; diacylglycerol acetyltransferase (DGAT) having an affect on fat deposition in milk and leptin, a protein important in energy metabolism. Linkage maps, QTL searches, comparative mapping, and fine mapping were useful, but time-consuming and cost extensive. Most of the economically important traits in livestock are under the control of numerous genes simultaneously, and interact both with one another as well as the production environment. Searching these genes one by one and establishing the genotype phenotype association was agigantictask.later,inthelasthalfof1990s,theefforts started for the whole genome sequencing of human and mouse using high-throughput sequencing (National Human Genome Research Institute, NHGRI) and whole genome shotgun (Celera genomics) approach. The animal scientists took advantage of infrastructure built by the NHGRI and efforts were initiated for sequencing the major livestock species. The first drafts of the sequence of chicken and cattle genomes were completed in 2005 (http://www.genome. gov/12512874). In 2006, the sequencing of the horse and the pig genomes was also begun, with the first draft of the horse sequence completing in 2007, and the

Genomics Research: Livestock Production 313 Table 3 Commercial DNA tests available for different livestock species. Species Trait category Cattle dairy Cattle Beef Sheep Pig Congenital BLAD CMS RYR defects Citrulinaemia Citrulinaemia DUMPS Congenital myasthenic syndrome Freemartinism Freemartinism MSUD MSUD Mannosidosis Mannosidosis CVM Genetic disorder α-mannosidosis α-mannosidosis β-mannosidosis β-mannosidosis Bovine hereditary zinc deficiency Bovine hereditary zinc deficiency Dwarfism Dwarfism Factor XI deficiency Pompes disease (E7) Factor IX deficiency Pompes disease (E13) α-mannosidosis Myophosphorylase deficiency Hypotrichosis and oligodontia Inherited congenital myoclonus Deficiency of uridine Monophosphate synthetase Weaver syndrome Protoporphyria Platelet bleeding Platelet bleeding Albinism Muscular hypertrophy Muscular hypertrophy Prp F18 Disease MC1R/MSHR MC1R/MSHR MC1R/MSHR Appearance Red coat color Red coat color CKIT Black coat color Black coat color TYR or Albinism MGF or Roan Milk quality κ-casein β-lactoglobulin FMO3 Milk yield and composition κ-casein GRH DGAT Meat quality TG RYR CAST RN/PRKAG3 Growth and composition Reproduction Feed intake CAPN1 CAPN3 RN/PRKAG3 RYR GH1 Myostatin Callipyge MC4R Booroola Inverdale Hanna >15 PICmarq IGF-2 MC4R Source: From J. Anim. Sci. [3]

314 Genomics Research: Livestock Production Economically important/ Complex trait loci Structural genomics Linkage maps QTL searches, comparative mapping and fine mapping association studies Functional genomics Microarray analysis Nucleotide polymorphism Variations in transcript levels Genetical genomics/eqtl Fig. 2 Integrative approaches applied to decipher economically important traits. pig sequence draft has also become available recently (http://www.ensembl.org/sus scrofa). Taking advantage of the availability of sequence draft, single nucleotide polymorphism (SNP) arrays have already been developed for chicken (Illumina TM 18K iselect chip), cattle (Bovine 50K Illumina TM iselect chip), pig (Illumina TM iselect pig DNA chip, 60K), and sheep (Illumina TM OvineSNP50) and are under development for other livestock species. SNP chip provides a platform where hundreds or thousands of markers can be genotyped in a cost effective manner. SNP arraybased association studies are being extended to traits like behavior, disease resistance, and structural and environmental soundness. [7] With the availability of species-specific SNP chips, it is now possible to undertake whole genome scans [8] and genomic selection approaches can be accomplished to correlate phenotype and genotype across the whole genome simultaneously, rather than by one locus at a time. Thus, SNP-based platforms are available to predict the molecular genetic value of individual animals for specific traits, viz., marbling and tenderness in beef cattle. SNP chips also provide a higher resolution map of the genome that can be used to detect region of chromosomal abnormalities, e.g., aneuploidies, microdeletions, microduplications, and loss of heterozygosity (LOH). As a variety of diseases are linked to such chromosomal abnormalities, SNP chips have the power to identify such regions and provide insight about the diseases and further suggest targets for intervention. [9] FUNCTIONAL GENOMICS QTL mapping is a powerful method and a number of QTLs have been reported in various livestock species, but only afewofthesehavebeenlocalizedatthegenelevel. [10] A gap still exists in the knowledge to localize QTL and then, to identify the underlying genes and their function which reduces the ability to harness the potentials of genomics. With the recent completion of the genome sequence of the major livestock species and availability of high-throughout technology, a functional genomics approach can now be used for the assignment of function to the identified genes and also for the organization and control of genetic pathways combining to determine the complex phenotype. The use of microarray technology for global profiling of gene expression is the primary method of functional genomics and has made it possible to compare expression levels for active genes under a variety of genetic and environmental conditions. Examples showing the power of this technology include identification of set of genes conferring resistance to Marek s disease in chicken and Trypanosoma congolense infection in cattle. Another powerful application of microarray technology is to synergistically harness the powers of recombination and functional analyses in a combined approach known as Genetical Genomics, or expression QTL (eqtl) mapping (Fig. 2). Such an approach has the potential to identify cis-acting and trans-acting genes in the region of the functional QTL and master regulators of quantitative trait variation as well, wherein polymorphisms at a single locus may regulate/control variation in gene expression of many other genes. Genetical genomics approach allows one to understand the groups of genes and governing variation in complex traits. Integrated information on phenotypic traits, pedigree structure, molecular markers, and gene expression can be used for estimating heritability of mrna transcript abundances so as to infer the regulatory gene network and map eqtl. Such studies will help in answering the questions concerning genes involved in meat and milk production, meat and milk quality and composition, and response to production systems/husbandry stressors. Eventually, this

Genomics Research: Livestock Production 315 knowledge will further aid in selection and management of livestock. IMPLICATIONS OF GENOMICS The most important utilization of the genomic data toward livestock production would be the use in selective breeding. Traditionally, the selection methods are based on the estimated breeding value (EBV) for traits of economic importance that are estimated from phenotypic records, genetic covariances among traits, and pedigree relationships in populations. In most cases, however, the actual location, identity, and functional role of the QTL remain unknown. In addition, efficiency of traditional selection practices is low when the traits are difficult to measure and have low heritability. Solution to these problems is gene-assisted selection, which is easily applied to traits that are difficult to measure, and have low heritability and slow genetic change. With the availability of the high-throughput platforms (SNP array), it is possible to incorporate DNA information from markers into EBVs. Selection decisions based on genomic breeding values (GEBV) are calculated as the sum of effects of dense genetic markers or haplotypes across the entire genome and thus, all the QTLs contributing to variation in a trait are captured. The QTL effects are first measured on a large reference populations with phenotypic records and then, only marker information is required to calculate GEBV in subsequent generations. GEBV holds special importance in developing countries where structured resource populations are not available for most of the livestock species. GEBV approach has already been evaluated in United States, New Zealand, Australia, and the Netherlands and is expected to double the rate of genetic gain in the dairy industry. [11] SUMMARY The demand for livestock products is projected to grow significantly in the developing countries. Traditional breeding methods are not well equipped to meet this increasing demand of meat and dairy products. The integrated use of structural and functional genomics has the potential to speed up the livestock production progress through understanding the genotype phenotype relationship and genotype environment interaction of livestock genomes as a whole. Selection decisions based on GEBV are made possible with the advances in the whole genome sequencing and high-throughput SNP panels. This approach of GEBV to select animals with desired allelic combinations is cost effective and can even target non-measurable traits. REFERENCES 1. Delgado, C.L. Rising consumption of meat and milk in developing countries has created a new food revolution. J. Nutr. 2003, 133,3907S 3910S. 2. Food and Agriculture Organization of the United Nations. FAOSTAT Statistical Database. FAO, 2009, http:// faostat.fao.org/default.aspx; ftp://ftp.fao.org/docrep/nonfao/ lead/x6155e/x6155e00.pdf. 3. Dekkers, J.C.M. Commercial application of marker- and gene-assisted selection in livestock: Strategies and lessons. J. Anim. Sci. 2004, 82,E313 E328. 4. Rothschild, M.F. Porcine genomics delivers new tools and results: this little piggy did more than just go to market. Genet. Res. 2004, 83,1 6. 5. O Brien, S.J.; Menotti-Raymond, M.; Murphy, W.J.; Nash, W.G.; Wienberg, J.; Stanyon, R.; Copeland, N.G.; Jenkins, N.A.; Womack, J.E.; Marshall Graves, J.A. The promise of comparative genomics in mammals. Science 1999, 286, 458 462, 479 481. 6. Grobet, L.; Pirottin, D.; Farnir, F.; Poncelet, D.; Royo, L.J.; Brouwers, B.; Christians, E.; Desmecht, D.; Coignoul, F.; Kahn, R.; Georges, M. Modulating skeletal muscle mass by postnatal, muscle-specific inactivation of the myostatin gene. Genesis 2003, 35,227 238. 7. Rothschild, M.F.; Plastow, G.S. Impact of genomics on animal agriculture and opportunities for animal health. Trends Biotechnol. 2007, 26,21 25. 8. Meuwissen, T.H.E.; Hayes, B.J.; Goddard, M.E. Prediction of total genetic value using genome-wide dense marker maps. Genetics 2001, 157,1819 1829. 9. Scharpf, R.B.; Ting, J.C.; Pevsner, J. BIOINFORMATICS SNP chip: R classes and methods for SNP array data. Bioinformatics 2008, 23,627 628. 10. Andersson, L.; Georges, M. Domestic animal genomics: deciphering the genetics of complex traits. Nat. Rev. Genet. 2004, 5,202 212. 11. Hayes, B.J.; Bowman, P.J.; Chamberlain, A.J.; Goddard, M.E. Invited review: genomic selection in dairy cattle: progress and challenges. J. Dairy Sci. 2009, 92,433 443.