Advantages

Advantages of Domestic Species as Biomedical Models

Goals

a) to present examples of recent and historical milestones in the uses of domestic species for biomedical research, and b) to provide examples of domestic species as biomedical models that may be under-funded or not funded by NIH.

Since 1901 there have been 17 Nobel Prizes awarded to scientists that used cows, chickens, swine, horse and sheep as biomedical models during their studies [1]. General research areas that potentially could be advanced by use of these domestic species as biomedical models are listed in Table 1. The following are specific examples of some of the advantages and uses of domestic species as biomedical models:

 

Examples of Historical and Recent Milestones in Use of Domestic Species for Biomedical Research

Immunology and Immunobiology:

Vaccination: Edward Jenner used the cowpox virus for the first vaccine against small pox [2], which paved the way for modern vaccination programs against smallpox and other human and animal plagues [3].

Acquired Immunological Tolerance: John Hunter first described freemartin cattle (female born co-twin with a male) [4], and Lillie dissected placenta of freemartins and first described the intermingling of blood between twins [5]. By 1951, Medawar (shared the Nobel Prize in Physiology in 1960 with Burnet) reported successful skin allografts between freemartins and normal males [6]. Macfarlane Burnet discovered that vascular anastomosis results in blood chimerism in dizygotic twins in cattle and this explains the tolerance to allografts between twins. This work in cattle formed the basis for Dr. Burnet’s Nobel Prize in Physiology in 1960 [7]. It also explained in part the freemartin condition in female calves born co-twin with a male and has been followed by a series of studies on microchimerism in parous women [8].

Humoral Immunity: Glick and colleagues reported that the avian bursa of Fabricius was necessary for humoral immunity and antibody production [9], which was later followed by identification of B lymphocytes and acceptance of the idea that lymphocytes are responsible for adaptive immunity [10].

Xenotransplantation:

Since 1960, hundreds of thousands of heart valves of pigs and cattle have been implanted into human hearts (24,000 annually in U.S. and 68,000 worldwide) [11].

Cadaveric or live organ transplants do not meet the demand for organs. Although porcine xenografts may be the solution to this problem [3], the surface of pig cells contain α-1,3-galactosyltransferase, and primates including humans contain preformed antibodies against epitopes on this molecule. Consequently, use of pig organs for transplantation into humans is severely hampered. Recently, nuclear transfer technology produced α-1,3-galactosyltransferase “knockout” pigs. Hearts and kidneys from these pigs have been successfully transplanted without hyperacute rejection and severe microangiopathy into primates [12-15]. These major scientific breakthroughs are expected to greatly enhance the potential use of pigs as a source of donor organs, tissues and cells for humans [16-19].

Porcine islets can restore euglycemia in animal models thus pigs may also be a source for islets to treat diabetes in humans [20].


Reproductive biology:

Artificial Insemination was initially practiced in horses in 1322 then more extensively in bitches and horses in the late 1700’s and 1800’s. By the 1930’s, AI was used commercially in 1000’s of mares, cows and sheep [21, 22].

Cryopreservation of sperm was accomplished first in cattle [23] and later applied to humans [24].

Superovulation and embryo transfer: Although Heape performed the first embryo transfer in rabbits in the 1890’s, the revival of interest, initial transfers in species other than rabbits, and potential application of superovulation and embryo transfer to improve reproductive efficiency and enhance genetic selection were pioneered beginning in the 1930’s by Warwick, Berry and Horlacher and later by Hammond, Rowson and Willett and others in goats, sheep and cattle [25].

Cloning & nuclear transfer: Ian Wilmut used nuclear transfer procedures to successfully clone the first mammal, a sheep named Dolly, from a non-reproductive adult cell [26].

 

Developmental Biology:

Because of the ease of manipulating developing limbs in vivo, chickens have provided fate maps and enabled scientists to determine the cell-cell interactions that result in limb pattern. Indeed, over the past 15 years, the role of an increasing number of developmentally important genes has been uncovered. The principles that underlie limb development in chickens are applicable to other vertebrates including humans and there are increasing linkages with clinical genetics [27].


Cardiovascular Disease:

Heart failure occurs in at least 400,000 people annually in the U.S. alone [28]. While genetically engineered mice are excellent models [28], large animals such as calves and pigs, which have a heart size, musculature, geometry and physiology similar to humans [29], are more appropriate models for surgery, hemodynamics [30], artificial heart development [29-31], and etiology of heart failure [28].

 

Biochemistry:

Isolation and characterization of thyrotropin-releasing hormone (TRH) and gonadotropin-releasing hormone (GnRH): Pigs and sheep were used as sources of millions of hypothalami for the isolation and peptide sequence determination of TRH and GnRH by Schally and Guillemin, which led to the Nobel Prize for both scientists in 1977 [32, 33].

Prostaglandins were isolated from sheep seminal vesicles, crystallized, and structures of PGE and PGF determined in a series of studies by Bergstrom and Sjovall from 1957 to 1960 [34]. Bergstrom was awarded the Nobel Prize in 1982 along with Samuelsson and Vane for their work on prostaglandins [34].


Nutrition:

Milk fat secretion studies in ruminants led to the first models for understanding lipid synthesis and nutrient utilization. [35, 36]. This work conducted in the 1950’s and 60’s provided some of the seminal data on nutrient content in milk for nourishment of the neonate. In addition, the ultrastructural characterization of the mammary gland and resulting micrographs continue to be used to provide a basic understanding of milk secretion in humans.

 

Metabolic Diseases:

Malignant hyperthermia is a potentially deadly disorder triggered by anesthetic agents and resulting in uncontrolled skeletal muscle calcium flux and organ failure. The stress-susceptible pig model provided important clues to the genes involved in this disorder in humans and continues to be the primary animal model to elucidate the physiological and genetic basis for this disorder (for review see: [37]).

Diabetes: the source of insulin for treatment of diabetes in humans was primarily of bovine and porcine origin from 1920 to 1982 [38].

Osteoporosis: Early work in the chicken led to the understanding of the importance of calcium metabolism and skeletal integrity [39] as well as the central role played by vitamin D in calcium absorption [40, 41].


Neurodegenerative Diseases:

Prions were first isolated by Prusiner (awarded Nobel Prize in Physiology or Medicine in 1997) from sheep and goats [42] and determined to be proteinaceous infectious pathogens that cause fatal neurodegenerative diseases including bovine spongiform encephalopathy (BSE), scrapie in sheep, and Creutzfeldt-Jakob disease (CJD) in humans [43].


New Therapeutics including pharming:

Interferon-τ, a novel cytokine discovered in sheep [44], is in clinical trials to evaluate its effectiveness as a treatment for multiple sclerosis [45-48].

Uteroferrin or TRAP (tartrate resistant acid phosphatase), a purple iron-binding glycoprotein with acid phosphatase activity, was isolated from uterine secretions of pigs [49]. This protein is a hematopoietic growth factor used to diagnose hairy cell leukemia, bone metastases in breast cancer patients [50] and bone turnover during osteoporosos [51, 52]. Pending approval TRAP may be used as a coating to improve bone grafts, bone cements and prosthetic devices by recruiting osteoclast progenitor cells from bone marrow or blood [53].

Inhibin-A and activin-A were first isolated from follicular fluids of sheep, cattle and pigs [54-58]. Currently, these dimeric glycoproteins are used as markers to diagnose Down Syndrome, and they may have value to diagnose complications associated with problem pregnancies in women including pre-eclampsia, early detection of pregnancy post IVF, miscarriages, and preterm labor, as well as ovarian cancer [59].

AMP or cationic anti-microbial peptides such as Iseganan and Micrologix are new anti-microbial agents derived from porcine leucocytes or bovine neutrophils that are in Phase II and III clinical trials [3].

Pharming: 17 different proteins are produced in cows, goats, sheep and pigs, and 10 of these proteins are produced at commercially feasible levels (>1 g liter) [60, 61] and many are in clinical trials [3]. For example, recombinant human α1-antitrypsin is produced in sheep to treat patients with pulmonary emphysema, a crippling lung disease caused by a lack of α1-antitrypsin in blood. Bovine lactoferrin, an iron-binding glycoprotein involved in host defense against infection and excessive inflammation, inhibits hepatitis C virus viremia in patients with chronic hepatitis C [62] and may have a novel role in improving cancer therapies [63]. The crystalline structure of human recombinant lactoferrin produced by transgenic cows is nearly identical to human lactoferrin [64]. Human recombinant anti-thrombin-III produced in goats is used as an anticoagulant to prevent thrombosis during surgery or delivery [65].


Current examples of use of domestic species as biomedical models that may not be funded or may be under-funded by NIH


Diseases and disorders:

Osteoporosis: Aged ovariectomized sheep fed calcium-wasting diets are a promising model for post menopausal osteoporosis because they are docile, easy to handle and house, and their bones are large enough to evaluate orthopedic implants [66]. Moreover, their skeletal turnover kinetics, hormonal profiles such as osteocalcin, responses of osteoblasts to hormones, and responses of sheep to estrogen replacement therapies are similar to humans [66].

Diabetes-induced accelerated atherosclerosis: Some 16 million people in the US have diabetes and 56 million may be insulin resistant [67]. Individuals with diabetes or insulin resistance have a 2- to 6-fold greater risk of developing atherosclerosis [67, 68]. Indeed, the most common cause of death in adult diabetics is coronary disease [69]. Swine have a cardiovascular system, metabolism and lipoprotein profiles similar to humans, and they develop spontaneous atherosclerosis with increased aging (see references in: [70]). Thus, the diabetes-induced accelerated atherosclerosis swine model, which has been well characterized, may be useful to elucidate the mechanisms causing accelerated atherosclerotic disease in humans with diabetes [70]. Even more useful to study diabetes and heart disease may be the feral pigs of Ossabaw Island, Georgia, because they develop precursors of diabetes and heart disease including raised blood pressure and hardened arteries within months of a regimen of high-fat diet and low exercise [71].

Anaphylaxis: In the U.S. it is estimated that there are 29,000 anaphylactic reactions to foods, especially peanuts, tree nuts, fish and shellfish, treated in emergency rooms and 125 to 150 deaths each year [72]. Pigs and calves are excellent models to study the mechanisms and treatments for allergic reactions to food because the gastrointestinal physiology and development of mucosal immunity in neonatal pigs is similar to humans [73], and neonatal swine and calves have hypersensitivity responses and adverse reactions similar to human allergic disease [74].

Asthma: It is estimated that 3 to 10% of the world’s population suffer from asthma and 100,000 die annually. Although allergic asthma does not occur in animals, animal models are critical to successful development of new therapies to treat this complex disease [75]. Sheep, horses and pigs for example have many asthma symptoms similar to humans, but may be an underutilized model for this disorder [75].

Sepsis: Sepsis occurs in 700,000 people and causes about 210,000 deaths annually in the U.S. alone [76]. Use of animal models to aid in development of new therapies to combat sepsis has met with little success because of the difficulty mimicking the clinical situation in humans leading to sepsis [77]. However, domestic species models enable investigators to evaluate therapies under conditions approximating clinical practice [78]. For example, sheep forced to inhale smoke and treated with bacteria undergo hyperdynamic sepsis similar to humans [79].


Reproductive biology:

Although ~123,000 cycles of assisted reproductive technology (ART) conducted in the 399 fertility clinics in the U.S. in 2003 resulted in ~36,000 births, 70% of the ART cycles were unsuccessful [80]. The recent discovery that women exhibit multiple FSH-induced wave-like patterns of follicular development throughout menstrual cycles [81-83] implies that cattle and horses, which like humans have well characterized follicular waves [84, 85] and a functional corpus luteum during their estrous cycles, may have significant advantages over the more traditional rodent models, especially for studies related to regulation of follicular development and oocyte quality during menstrual cycles to improve the success of assisted reproductive technologies [80].

Nutrition:

Atherosclerosis: Because of the similarity in size and other physiological, biochemical and anatomical properties to humans, including the tendency to overeat, pigs have advantages over rodent models to study atherosclerosis and cardiovascular disease. For example, feeding high-fat cholesterol diets to pigs results in atherosclerosis-like symptoms [86], but unless mouse models are genetically modified they are resistant [87].

Food intake: The dairy cow is a unique animal model to study the complex interactions of short-term signals regulating food intake, especially because the cow’s strong drive to eat, docile nature, and large size make it ideal to simultaneously monitor digestion and adsorption kinetics, endocrine responses, gene expression, metabolite pools and fluxes, and feeding behavior in response to various treatments [88].

Nutrition and food safety: Pigs and humans have a similar GI anatomy, morphology and physiology and thus have advantages over rodents as a biomedical model for nutrition studies as well as to evaluate safety of new food additives (see references in [89]). For example, the pig is an appropriate model to test the effects of neonatal Fe deficiency on anemia and the immune system [90] and effects of olestra, a fat substitute added commercially to foods, on nutrient availability [89].


Gene therapy:

Electrogene therapy was used to transfer the gene for a protease-resistant form of growth hormone releasing hormone (GHRH) into muscles of pigs, which increases secretion of growth hormone and growth for over 65 days [91]. Because of the similar physiology and anatomy of pigs and humans, pigs may be a more useful biomedical model compared with rodents to improve gene therapy to correct defects of GHRH secretion, which characterize conditions such as Turner’s syndrome, hypochrondroplasia, Crohn’s disease, intrauterine growth retardation or renal insufficiency [3].


Alcoholism and related disorder:

Pigs are the only species other than humans that prefer alcohol. In addition, their brain growth velocity is similar to humans and they are intelligent [92]. However, pigs may be underutilized as a model not only for alcoholism [93], but also effects of alcohol on prenatal development [92, 94] and alcohol induced atrial tachyarrhythmias, e.g., “holiday heart syndrome”. [95]. Sheep also have significant advantages over rodents for studies on prenatal effects of alcohol because they have brain growth and gestation characteristics similar to humans and are well established as models for fetal physiology and long-term instrumentation [92].


Oncology:

Lung cancer kills more than one million people annually [96] and adenocarcinoma is increasing in frequency and a significant cause of lung cancers [97]. Because sheep develop ovine pulmonary adenocarcinoma (incidence is 2 to 30%, see references in [98]), a naturally occurring adenocarcinoma caused by jaagsiekte retrovirus, they are an excellent model to test therapeutic agents and to study the pathogenesis, natural history, epidemiology and host susceptibility of the disease [98].

Cutaneous melanoma is one of the most deadly forms of cancer and it is increasing at a rate faster than any other cancer [99, 100]. Some lines of minipigs have a high incidence of melanoma and develop spontaneous melanomas before 3 months of age thus they have advantages over rodent models, especially because spontaneous melanomas are rare in rodents, and precursor melanoma cells in genetic “knockout” mice are from the dermis rather than the epidermis as in humans [100, 101].

Ovarian cancer afflicts ~1 of 70 women in the US and it is one of the most lethal and difficult to detect (fewer than 25% detected at curable stage) of cancers that afflict women [102, 103]. Chickens are an important biomedical model for etiological studies and to evaluate chemopreventive and therapeutic agents because they are the only known species other than humans that exhibit spontaneous ovarian cancer resulting from transformation of ovarian surface epithelial cells [102-106].

 

Genomics:

Genomic variation is clearly a major factor in host resistance to pathogens in humans and animals. Identification of specific genome sequences that predispose susceptibility/resistance to disease will be fundamental to averting accidental or terrorist-initiated epidemics, and to developing models of human gene/pathogen interaction. Genomic variation also underlies traits such as growth, body composition, lactation, and reproductive health. The working draft of genome sequences of agriculturally important animals will, therefore, provide an invaluable resource for discovery of genes and their functions to benefit human health, animal health and production animal agriculture [107]. For example, numerous advances have been made in swine biomedical model genomics. For a review, see [108]. Moreover, insights from genomics in domestic species have already contributed to the discovery of human diseases [109]. Specifically, a mutation in the MC4R gene is associated with fatness in pigs [110] and also results in obesity in humans [111], and discovery of the limbin gene that causes chondrodysplastic dwarfism in Japanese Brown cattle [112] aided in discovery of the gene responsible for Ellis-van Creneld syndrome [113], which causes dwarfism in humans [109].

Quantitative traits: Cattle have long been studied for complex traits influenced by multiple genes as well as environmental factors. These so called “quantitative traits” are now targeted by the human health research community. Cardiovascular health, obesity, and several cancers are examples of complex traits segregating in breeding populations of cattle. The understanding of what makes cattle breeds different with respect to reproduction, lactation, growth, bone structure, fat deposition, altitude and heat tolerance, and resistance to specific pathogens will be invaluable in elucidating related physiological processes important to human health.


Infectious disease and vaccine development:

Pathogenesis and immunity are best studied in animal models in which the pathogen is naturally occurring. While there are infectious diseases in which rodents serve as relevant models, restriction to rodents, primarily mice, ignores the diversity of animal species with microbial diseases that recapitulate the human disease and represents a loss of research opportunity when the most representative model is not investigated. This often reflects a lack of knowledge in the biomedical community of the occurrence of alternative domestic animal models or that the domestic animal model has not been developed to the point where the key questions can be addressed. The study of the equine lentivirus EIAV as a model of HIV [114, 115], Anaplasma and Babesia in cattle as models of vector-borne pathogens [116-118], and scrapie in sheep as a model of spongioform encephalopathy [119] are only a few examples of projects that have provided novel insight into human diseases. In addition, the cow is an underutilized model for uterine infection and immunity [120]. Enhanced research into domestic animal models of infectious disease is necessary to expand our knowledge of fundamentals in transmission and pathogenesis of infectious agents.


Rotavirus causes severe gastroenteritis in infants and young children and animals worldwide. Gnotobiotic pigs are excellent models because their gastrointestinal physiology and development of mucosal immunity and susceptibility to infection with human rotavirus strains resembles humans. In addition, the pigs are born virus-free and without maternal antibodies, which eliminates confounding variables and enables assessment of true primary immune responses [121].

Vaccine development: Development and testing of novel vaccines in domestic animals is both informative as models and as a means to directly control spread of zoonotic diseases. Domestic animals respond to vaccination with marked variation in responses among individuals, similar to the responses of humans but unlike inbred mouse models [122, 123]. This variation in response allows testing of novel vaccine constructs to enhance responses of individual “poor responders” as well as to examine the effects of this variation in immunity among individuals at the population level. Both issues are of fundamental importance in improving vaccination as a means to improve human health. In addition, the development and deployment of animal vaccines have been used to directly control disease spread to humans, the most striking example perhaps being rabies virus, in which human health is protected essentially entirely through vaccination of animals, rather than humans themselves [124]. This concept is applicable to a variety of food and water-borne pathogens, vector-borne diseases, and may extend to prevention of transmissible spongioform encephalopathies (prion diseases) [125, 126].


Table 1. Research areas (not prioritized) that potentially could be advanced by use of agricultural animals as biomedical models. ______________________________________________________________________

Epigenetics and environment: effect of photoperiod, global warming, seasonality, and elevation on modification of gene function

Reproduction: gametogenesis, gonadal function, infertility

Aging: skeletal diseases, especially chicken and pig models; bone metabolism and osteoarthritis, especially the horse model; reproduction, especially beef cattle, mares

Obesity: genetic, dietary, hormonal influences on pre- and post-natal adipose tissue development using pig model [86, 89]

Pregnancy: placental growth, angiogenesis, congenital and birth defects, developmental biology especially chickens, fetal programming especially sheep to study stress, malnutrition, effects of exposure of fetuses to androgens and environmental toxins on adults, molecular/cellular basis of parturition and premature birth

Diabetes Types I and II [70].

Therapeutics: xenotransplantation [11, 127], gene therapy, pharming [91, 127], stem cells, “Farmaceuticals” [3]

Toxicology [128], environmental endocrine disrupters

Neurobiology: behavior, stress, learning, pheromonal communication, neuroendocrinology

Immunology: autoimmune disease, inflammation, innate and mucosal

Cardiovascular disorders such as diet-induced artherosclerosis [86], and lethal cardia tachyarrhythmias (ventricular fibrillation) using miniature or normal pigs

Nutrition: energetic balance including homeostatic mechanism, regulation of metabolism, use of neonatal piglet as pediatric model for studies of nutrition,metabolism and gastroenterology (see references in:[89])

Ophthalmology: retinal degeneration, retinitis pigmentosa [129], macular degeneration

Comparative physiology (e.g., Understanding of what makes cattle breeds different with respect to reproduction, lactation, growth, bone structure, fat deposition, altitude and heat tolerance, and resistance to specific pathogens will be invaluable in elucidating related physiological processes important to human health)

Interventional Radiology [130]

Biomechanics

Renal biology

Diseases: Transmissible Spongiform Encephalopathies (TSE); Respiratory Syncytial Virus (RSV); Crohn’s Disease; sexually transmitted diseases (STD); enteric including Transmissible

Gastroenteritis (TGE); viral, E. coli 01578; cancer including prostate, breast, ovary (chicken), hematopoiesis, leukemia; cattle as a model for salmonellosis, tuberculosis and cryptosporidiosis; pathogen transmission of emerging diseases that infect animals and humans such as use of cattle to study tick-borne infections

Disorders: liver, epilepsy, and sleep such as narcolepsy

Microbial ecology

Go Back


REFERENCES

1. FBR. Nobel Prizes The Payoff from Animal Research.

2. Barquet N, Domingo P. Smallpox: The triumph over the Most Terrible of the Ministers of Death. Annals of Internal Medicine 1997; 127: 635-642.

3. Kues WA, Niemann H. The contribution of farm animals to human health. TRENDS in Biotechnology 2004; 22: 287-294.

4. Hunter J. On the Free Martin. Royal Coll Surg 1779; XX.

5. Lillie FR. The theory of the free-martin. Sceince 1916; 43: 611.

6. Anderson D, Billingham RE, Lamkin GH, Medawar PB. Use of skin grafting to distinguish between monzygotic and dizygotic twins in cattle.  Heredity 1951; 5: 379.

7. Fenner F. Frank Macfarlane Burnet 1899-1985. Historical Records of Australian Science 1987; 7: 1-37.

8. Ichinohe T, Maruya E, Saji H. Long-term feto-maternal microchimerism: nature's hidden clue for alternative donor hematopoietic cell transplantation. International Journal of Hematology 2002; 76: 229-237.

9. Glick B, Chang TS, Jaap RG. The bursa of fabricius and antibody production. Poultry Science 1956; 35: 224-225.

10. Monroe JG. B-cell positive selection and peripheral homeostasis. Immunological Reviews 2004; 197: 5-9.

11. Schoen FJ, Levy RJ. Tissue heart valves: Current challenges and future research perspectives. Journal of Biomedical Materials Research 1999; 47: 439-465.

12. Tseng Y-L, Kuwaki K, Dor FJMF, Shimizu A, Houser S, Hisashi Y, Yamada K, Robson SC, Awwad M, Schuurman H-J, Sachs DH, Cooper DKC. alpha 1,3-Galactosyltransferase gene-knockout pig heart transplantation in baboons with survival approaching 6 months. Xenotransplantation 2005; 80: 1493-1500.

13. Kuwaki K, Tseng Y-L, Dor FJMF, Shimizu A, Houser SL, Sanderson TM, Lancos CJ, Prabharasuth DD, Cheng J, Moran K, Hisashi Y, Mueller N, Yamada K, Greenstein JL, Hawley RJ, Patience C, Awwad M, Fishman JA, Robson SC, Schuurman H-J, Sachs DH, Cooper DKC. Heart transplantation in baboons using [alpha]1,3-galactosyltransferase gene-knockout pigs as donors: initial experience. Nat Med 2005; 11: 29-31.

14. Yamada K, Yazawa K, Shimizu A, Iwanaga T, Hisashi Y, Nuhn M, O'Malley P, Nobori S, Vagefi PA, Patience C, Fishman J, Cooper DKC, Hawley RJ, Greenstein J, Schuurman H-J, Awwad M, Sykes M, Sachs DH. Marked prolongation of porcine renal xenograft survival in baboons through the use of [alpha]1,3-galactosyltransferase gene-knockout donors and the cotransplantation of vascularized thymic tissue. Nat Med 2005; 11: 32-34.

15. Dor FJMF, Kuwaki K, Tseng YL, Shimizu A, Houser SL, Yamada K, Hawley RJ, Patience C, Awwad M, Fishman JA, Robson SC, Sachs DH, Schuuman HJ, Cooper DKC. Potential of aspirin to inhibit thrombotic microangiopathy in alpha1,3-galactosyltransferse gene-knockout pig hearts after transplantation in baboons.  Transplantation Proceedings 2005; 37: 489-490.

16. Buhler L, Friedman T, Iacomini J, Cooper DKC. Xenotransplantation-state of the art-update 1999. Frontiers in Bioscience 1999; 4: 416-432.

17. Wheeler MB, Walters EM, Clark SG. Transgenic animals in biomedicine and agriculture: outlook for the future. Animal Reproduction Science 2003; 79: 265-289.

18. Lai L, Kolber-Simonds D, Park K-W, Cheong H-T, Greenstein JL, Im G-S, Samuel M, Bonk A, Rieke A, Day BN, Murphy CN, Carter DB, Hawley RJ, Prather RS. Production of alpha -1,3-Galactosyltransferase Knockout Pigs by Nuclear Transfer Cloning. Science 2002; 295: 1089-1092.

19. Kolber-Simonds D, Lai L, Watt SR, Denaro M, Arn S, Augenstein ML, Betthauser J, Carter DB, Greenstein JL, Hao Y, Im G-S, Liu Z, Mell GD, Murphy CN, Park K-W, Rieke A, Ryan DJJ, Sachs DH, Forsberg EJ, Prather RS, Hawley RJ. Production of {alpha}-1,3-galactosyltransferase null pigs by means of nuclear transfer with fibroblasts bearing loss of heterozygosity mutations. PNAS 2004; 101: 7335-7340.

20. MacKenzie DA, Hullett DA, Sollinger HW. Xenogeneic transplantation of procine islets: an overview. Transplantation 2003; 76: 887-891.

21. Dalton JC. Factors important to the efficiency of artificial insemination in single-ovulating and superovulated cattle. Blacksburg, Virginia: Virginia Polytechnic Institute and State University 1999.

22. Allen WR. The Development and Application of the Modern Reproductive Technologies to Horse Breeding. Reproduction in Domestic Animals 2005; 40: 310-329.

23. Polge C, Smith AU, Parkes AS. Revival of spermatoza after vitrification and dehydration at low temperatures. Nature 1949; 164: 666.

24. Foote RH. Cryopreservation of spermatozoa and artificial insemination: past, present and future. J Androl 1982; 3: 85-100.

25. Betteridge KJ. A history of farm animal embryo transfer and some associated techniques. Anim Reprod Sci 2003; 79: 203-244.

26. Campbell KHS, McWhir J, Ritchie WA, Wilmut I. Sheep cloned by nuclear transfer from a cultured cell line. Nature 1996; 380: 64-66.

27. Tickle C. The contribution of chicken embryology to the understanding of vertebrate limb development. Mech Dev 2004; 121: 1019-1029.

28. Cohn JN, Bristow MR, Chien KR, Colucci WS, Frazier OH, Leinwand LA, Lorell BH, Moss AJ, Sonnenblick EH, Walsh RA, Mockrin SC, Reinlib L. Report of the National Heart, Lung, and Blood Institute Special Emphasis Panel on Heart Failure Research. Circulation 1997; 95: 766-770.

29. Chinchoy E, Soule CL, Houlton AJ, Gallagher WJ, Hjelle MA, Laske TG, Morissette J, Iaizzo PA. Isolated four-chamber working swine heart model. Ann Thorac Surg 2000; 70: 1607-1614.

30. McCarthy PM, Fukamachi K, Fukumura F, Muramoto K, Golding LAR, Harasaki H. The Cleveland Clinic-Nimbus total artificial heart: In vivo hemodynamic performance in calves and preclinical studies. J Thorac Cardiovasc Surg 1994; 108: 420-428.

31. DeBakey ME. The Odyssey of the Artificial Heart. Artificial Organs 2000; 24: 405-411.

32. Schally AV. Nobel Lecture-Aspects of hypothalamic regulation of the pituitary gland with major emphasis on its implications for the control of reproductive processes [www.nobel.se/medicine/laureates/1977/schally-lecture.html]. 1977.

33. Guillemin R. Nobel Lecture-Peptides in the brain: the new endocrinology of the neuron [http://www.nobel.se/medicine/laureates/1977/guillemin-lecture.html]. 1977.

34. Flower RJ. Prostaglandins, bioassay and inflammation. British Journal of Pharmacology 2006; 147: S182-S192.

35. McCarthy RD, Patton, S. Biosynthesis of glycerides in freshly secreted milk. Nature 1964; 202: 347-349.

36. Patton S, Josephson, D.V. Methionine: origin of sunlight flavor in milk. Science 1953; 118: 211.

37. Jurkat-Rott K, McCarthy T, Lehmann-Horn F. Genetics and pathogenesis of malignant hyperthermia. Muscle Nerve 2000; 23: 4-17.

38. Cramer P, Martinet J. Biotechnology for Diabetes: Insulin and insulin analogues. In: Bioimpact; 2004: 1-21.

39. Mueller WJ, Schraer R, Schraer H. Calcium metabolism and skeletal dynamics of laying pullets. J Nutr 1964; 84: 20-26.

40. Wasserman RH, Taylor AN. Vitamin D3-induced calcium binding protein in chick intestinal mucosa. . Science 1966; 152: 791-793.

41. Jande SS, Maler L, Lawson EM. Immunohistochemical mapping of vitamin D-dependent calcium binding protein in brain. Nature 1981; 294: 765-767.

42. Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science 1982; 216: 136-144.

43. Prusiner SB. Prions. PNAS 1998; 95: 13363-13383.

44. Imakawa K, Anthony RV, Kazemi M, Marotti KR, Polites HG, Roberts RM. Interferon-like sequence of ovine trophoblast protein secreted by embryonic trophoectoderm. Nature 1987; 330: 377-379.

45. Soos JM, Mujtaba GM, Subramaniam PS, Streit WJ, Johnson HM. Oral feeding of interferon s can prevent acute and chronic relapsing forms of experimental allergic encephalomyelitis. J Neuroimmunol 1997; 75: 43-51.

46. Soos JM, Johnson HM. Interferon tau prospects for clinical use in autoimmune disease. Bio Drugs 1999; 11: 125-126.

47. Olek MJ, Smith D.J., Cook SL, Khoury SJ, Weiner HL. Phase I study of oral recombinant ovine interferon tau in relapsing-remitting multiple sclerosis. . Neurology 2001; 56(Suppl 3): A76-A77.

48. Lutton JD, Winston R, Rodman TC. Multiple Sclerosis: Etiological Mechanisms and Future Directions. Experimental Biology and Medicine 2004; 229: 12-20.

49. Schlosnagle DC, Bazer FW, Tsibris JCM, Roberts MR. J Biol Chem 1974; 249: 7574-7579.

50. Capeller B, Caffier H, Sutterline MW, Deitl J. Evaluation of tartrate-resistant acid phosphatase (TRAP) 5b as serum marker of bone metastases in human breast cancer. Anticancer Res 2003; 2003: 1011-1015.

51. Bazer FW, Worthington-White D, Fliss MF, Gross S. Uteroferrin: a progesterone-induced hematopoietic growth factor of uterine origin Exp Hematol 1991; 19: 910-915.

52. Halleen JM, Alatalo SL, Janckila AJ, Woitge HW, Seibel MJ, Vaananen HK. Serum Tartrate-resistant Acid Phosphatase 5b Is a Specific and Sensitive Marker of Bone Resorption. Clin Chem 2001; 47: 597-600.

53. Lee MY, Evre DR, Weis MAE. Trap-coated bone grafts and prostheses. In: Office UP (ed.), vol. 60/044,033. US; 1997.

54. Vale W, Rivier J, Vaughan J, McClintock R, Corrigan A, Woo W, Karr D, Spiess J. Purification and characterization of an FSH releasing protein from porcine ovarian follicular fluid. Nature 1986; 321: 776-782.

55. Leversha LJ, Robertson DM, de Vos FL, Morgan FJ, Hearn MTW, Wettenhall REH, Findlay JK, Burger HG, de Kretser DM. Isolation of inhibin from ovine follicular fluid. Journal of Endocrinology 1987; 113: 213-221.

56. de Kretser DM, Robertson DM. The isolation and physiology of inhibin and related proteins. Biology of Reproduction 1989; 40: 33-47.

57. Miyamoto K, Hasegawa Y, Fukuda M, Igarashi M. Demonstration of high molecular weight forms of inhibin in bovine follicular fluid (bFF) by using monoclonal antibodies to bFF 32K inhibin. Biochemical and Biophysical Research Communications 1986; 136: 1103-1109.

58. Knight PG. Identification and purification of inhibin and inhibin-related proteins. J Reprod Fert 1991; Suppl 43: 111-123.

59. The ABCs of Inhibins and Activins. In.

60. Rudolph NS. Biopharmaceutical production in transgenic livestock. TIBTECH 1999; 17: 367-374.

61. Das RC. Production of therapeutic proteins from transgenic animals. BioBusiness 2001: 60-64.

62. Tanaka K, Ikeda M, Nozaki A, Kato N, Tsuda H, Saito S, Sekihara H. Lactoferrin Inhibits Hepatitis C Virus Viremia in Patients with Chronic Hepatitis C: A Pilot Study. Cancer Science 1999; 90: 367-371.

63. Varadhachary A, Wolf JS, Petrak K, O'Malley Jr. BW, Spadaro M, Curcio C, Forni G, Pericle F. Oral lactoferrin inhibits growth of established tumors and potentiates conventional chemotherapy. . International Journal of Cancer 2004; 111: 398-403.

64. Thomassen EAJ, van Veen HA, van Berkel PHC, Nuijens JH, Abrahams JP. The protein structure of recombinant human lactoferrin produced in the milk of transgenic cows closely matches the structure of human milk-derived lactoferrin. Transgenic Research 2005; 14: 397-405.

65. Konkle BA, Bauer KA, Weinstein R, Greist A, Holmes HE, Bonfiglio J. Use of recombinant human antithrombin in patients with congenital antithrombin deficiency undergoing surgical procedures. Transfusion 2003; 43: 390-394.

66. Turner AS. Animal models of osteoporosis - Necessity and limitations. European Cells and Materials 2001; 1: 66-81.

67. Valenzl KD. The link between diabetes and heart disease. In: DIABETES/HEART RESEARCH, vol. Winter; 2002: 15-16.

68. Kannel WB, McGee DL. Diabetes and cardiovascular disease: the Framingham study. JAMA 1979; 241: 2035-2038.

69. Wingard DL, Barrett-Connor E. Heart disease and diabetes. In: Diabetes in America, vol. NIH publication no. 95-1468, 2nd ed. Bethesda: NIH; 1995: 429-448.

70. Gerrity RG, Natarajan R, Nadler JL, Kimsey T. Diabetes-Induced Accelerated Atherosclerosis in Swine. Diabetes 2001; 50: 1654-1665.

71. Whitfield J. news@nature.com. In: Fat pigs ape obese humans; 2003.

72. Sampson HA. Food anaphylaxis. Br Med Bull 2000; 56: 925-935.

73. Helm RM, Ermel RW, Frick OL. Nonmurine animal models of food allergy. Environmental Health Perspectives 2003; 111: 239-244.

74. Barratt MEJ, Strachan PJ, Porter P. Antibody mechanisms implicated in digestive disturbances following ingestion of soya protein in calves and piglets. Clin Exp Immunol 1978; 31: 305-312.

75. Szelenyl I. Animal models of branchial asthma. Inflammation Research 2000; 49: 639-654.

76. Riedemann NC, Guo R-F, Ward PA. The enigma of sepsis. J. Clin. Invest. 2003; 112: 460-467.

77. Michie HR. The value of animal models in the development of new drugs for the treatment of the sepsis syndrome. J Antimicrobial Chemotherapy 1998; 41 (Suppl A): 47-49.

78. Marshall JC, Creery D. Pre-clinical models of sepsis. Sepsis 1998; 2: 187-197.

79. Kazunori M, Bjertnaes LJ, Schmalstieg FC, McGuire R, Cox RA, Hawkins HK, Herndon DN, Treber LD, Traber DL. A novel animal model of sepsis after acute lung injury in sheep. Critical Care Medicine 2002; 30: 2083-2090.

80. CDC. 2003 Assisted Reproductive Technology (ART) Report: Introduction to the 2003 National Report. In; 2005.

81. Baerwald AR AG, Pierson RA. A new model for the menstrual cycle in women. In: Proceedings of the 11th World Congress on Human Reproduction; 2002; Montreal.

82. Baerwald AR, Adams GP, Pierson RA. Characterization of ovarian follicular waves in women. Biol Reprod 2003; 69: 1023-1031.

83. Baerwald AR, Adams GP, Pierson RA. A new model for ovarian follicular development during the human menstrual cycle. Fertil Steril 2003; 80: 116-122.

84. Adams GP. Comparative patterns of follicle development and selection in ruminants. J Reprod Fert 1999; 54: 17-32.

85. Ginther OJ, Wiltbank MC, Fricke PM, Gibbons JR, Kot K. Selection of the dominant follicle in cattle. Biol Reprod 1996; 55: 1187-1194.

86. Bergen WG, Mersmann HJ. Comparative Aspects of Lipid Metabolism: Impact on Contemporary Research and Use of Animal Models. J. Nutr. 2005; 135: 2499-2502.

87. Breslow JL. Mouse Models of Atherosclerosis. Science 1996; 272: 685-688.

88. Allen MS, Bradford BJ, Harvatine KJ. The cow as a model to study food intake regulation. Annu Rev Nutr 2005; 25: 532-547.

89. Cooper DA, Berry DA, Spendel VA, Kiorpes AL, Peters JC. The Domestic Pig as a Model for Evaluating Olestra's Nutritional Effects. J. Nutr. 1997; 127: 1555S-.

90. Svoboda M, Drabek J, Krejci J, Rehakova Z, Faldyna M. Impairment of the Peripheral Lymphoid Compartment in Iron-deficient Piglets. Journal of Veterinary Medicine Series B 2004; 51: 231-237.

91. Draghia-Akli R, Fiorotto ML, Hill LA, Malone PB, Deaver DR, Schwartz RJ. Myogenic expression of an injectable protease-resistant growth hormone-releasing hormone augments long-term growth in pigs. Nat Biotech 1999; 17: 1179-1183.

92. Cudd TA. Animal Model Systems for the Study of Alcohol Teratology. Experimental Biology and Medicine 2005; 230: 389-393.

93. Dexter JD, Tumbleson ME, Hutcheson DP, Middleton CC. Sinclair (S-1) miniature swine as a model for the study of human alcoholism. Ann. N.Y. Acad. Sci. 1976; 273: 188-193.

94. Kubotsu SL, Jianbo H, Carmahan KG, deAvila J, Ott TL, Mirando M. The effects of chronic ethanol consumption during early pregnancy on conceptus health and uterine function in pigs. Alcoholism 2003; 27: 712-719.

95. Zaballos M, Almendral J, Anadon MJ, Gonzalez P, Navia J. Comparative effects of thiopental and propofol on atrial vulnerability: electrophysiological study in a porcine model including acute alcoholic intoxication. Br. J. Anaesth. 2004; 93: 414-421.

96. Greenlee RT, Murray T, Bolden S, Wingo PA. Cancer statistics, 2000. CA Cancer J Clin 2000; 50: 7-33.

97. Greco RJ, Steiner RM, Goldman S, Cotler H, Patchefsky A, Cohn HE. Bronchoalveolar cell carcinoma of the lung. Ann Thorac Surg 1986; 41: 652-656.

98. Palmarini M, Fan H. Retrovirus-Induced Ovine Pulmonary Adenocarcinoma, an Animal Model for Lung Cancer. J Natl Cancer Inst 2001; 93: 1603-1614.

99. Misfeldt ML, Grimm DR. Sinclair miniature swine: an animal model of human melanoma. . Vet Immunol Immunopathol 1994; 43: 167-175.

100. Vincent-Naulleau S, et al. Clinical and histopathological characterization of cutaneous melanomas in the melanoblastoma-bearing Libechov minipig model. Pigment Cell Res 2004; 17: 24-35.

101. Kusewitt DF, Ley RD. Animal models of melanoma. Cancer Surv 1996; 26: 35-70.

102. Rodriguez-Burford C, Barnes MN, Berry W, Partridge EE, Grizzle WE. Immunohistochemical expression of molecular markers in an avian model: a potential model for preclinical evaluation of agents for ovarian cancer chemoprevention. Gynecologic oncology 2001; 81: 373-379.

103. Mills GB. Mechanisms Underlying Chemoprevention of Ovarian Cancer. Clin Cancer Res 2002; 8: 7-10.

104. Fredrickson TN. Ovarian tumors of the hen. . Environ Health Perspect 1987; 73: 35-51.

105. Murdoch WJ. Carcinogenic Potential of Ovulatory Genotoxicity. Biol Reprod 2005; 73: 586-590.

106. Murdoch WJ, Van Kirk EA, Alexander BM. DNA Damages in Ovarian Surface Epithelial Cells of Ovulatory Hens. Experimental Biology and Medicine 2005; 230: 429-433.

107. Roher G, Beever JE, Rothschild MF, Schook L, Gibbs R, Weinstock G. Porcine sequencing white paper. In: Porcine genomic sequencing initiative: 1-10.

108. Lunney JK. Advances in swine biomedical model genomics. International Journal of Biological Sciences 2007; 3: 179-184.

109. Womack JE. Advances in livestock genomics: Opening the barn door. Genome Res. 2005; 15: 1699-1705.

110. Kim KS, Larsen N, Short T, Plastow G, Rothschild MF. A missense variant of the porcine melanocortin-4 receptor (MC4R) gene is associated with fatness, growth, and feed intake traits. Mammalian Genome 2000; 11: 131-135.

111. Farooqi IS, Yeo GSH, Keogh JM, Aminian S, Jebb SA, Butler G, Cheetham T, O'Rahilly S. Dominant and recessive inheritance of morbid obesity associated with melanocortin 4 receptor deficiency. J. Clin. Invest. 2000; 106: 271-279.

112. Takeda H, Takami M, Oguni T, Tsuji T, Yoneda K, Sato H, Ihara N, Itoh T, Kata SR, Mishina Y, Womack JE, Moritomo Y, Sugimoto Y, Kunieda T. Positional cloning of the gene LIMBIN responsible for bovine chondrodysplastic dwarfism. PNAS 2002; 99: 10549-10554.

113. Galdzicka M, Patnala, S., Hirshman, M.G., Cai, J.-F., Nitowsky, H., Egeland, J.A., Ginns, E.I. A new gene, EVC2, is mutated in Ellis-van Creveld syndrome. Mol Gene Metabo 2002; 77: 291-295.

114. Craigo JK, Steckbeck JD, Durkin S, Howe L, Cook SJ, Issel C, Montelaro RC. Discerning an effective balance between equine infectious anemia virus attenuation and vaccine efficacy. Journal of Virology 2005; 79: 2666-2677.

115. Mealey RH, Sharif A, Ellis SA, Littke MH, Leib SR, McGuire TC. Early detection of dominant Env-specific and subdominant Gag-specific CD8+ lymphocytes in equine infectious anemia virus-infected horses using major histocompatibility complex class l/peptide tetrameric complexes. . Virology 2005; 339: 110-126.

116. Allred DR, Al-Khedery B. Antigenic variation and cytoadhesion in Babesia bovis and Plasmodium falciparum: different logics achieve the same goal. Molecular and Biochemical Parasitology 2004; 134: 27-35.

117. Barbour AG, Restrepo BI. Antigenic variation in vector-borne pathogens. Emerging Infectious Diseases 2000; 6: 449-457.

118. Barry JD, Marcello L, Morrision LJ, Read AF, Lythgoe K, Jones N, Carrington M, Blandin G, Bohme U, Caler E, Hertz-Fowler C, Renauld H, El-Sayed N, Berriman M. What the genome sequence is revealing about trypanosome antigenic variation. Biochemical Society Transactions 2005; 33: 986-989.

119. Prusiner SB, DeArmond SJ. Molecular biology and pathology of scrapie and the prion. Brain Pathology 1991; 1: 297-310.

120. Hearath S, Dobson H, Bryant CE, Sheldon LM. Use of the cow as a large animal model of uterine infection and immunity. Journal of Reproductive Immunology 2006; 69: 13-22.

121. Yuan L, Ward LA, Rosen BI, Thanh LT, Saif LJ. Systemic and intestinal antibody-secreting cell responses and correlates of protective immunity to human rotavirus in a gnotobiotic pig model of diseasel J Virol 1996; 70: 3075-3083.

122. Glass EJ. Genetic variation and responses to vaccines. Animal Health Research Reviews 2004; 5: 197-208.

123. Ovsyannikova IG, Dhiman N, Jacobson RM, Poland GA. Human leukocyte antigen polymorphisms: variable humoral immune responses to viral vaccines. Expert Reviews of Vaccines 2006; 5: 33-43.

124. Bleck TP, Rupprecht CE. Rabies virus. In: Mandell GL, Bennett JE, Dolin R (eds.), Principles and Practice of Infectious Diseases. Philadelphia: Churchill-Livingstone.

125. Goni F, Knudsen E, Schreiber F, Pankiewicz J, Carp R, Meeker HC, Rubenstein R, Brown DR, Sy MS, Chabalgooity JA, Sigurdsson EM, Wisniewski T. Mucosal vaccination delays or prevents prion infection via an oral route. Neuroscience 2005; 133: 413-421.

126. Tsao JI, Wootton JT, Bunikis J, Luna MG, Fish D, Barbour AG. An ecological approach to preventing human infection: vaccinating wild mouse reservoirs intervenes in the Lyme disease cycle. . PNAS 2004; 101: 18164-18164.

127. Niemann H, Kues W, Carnwath JW. Transgenic farm animals: present and future. Rev sci tech Off int Epiz 2005; 24: 285-298.

128. Harrington GW, Magee PN, Pylypiw Jr. HMW, Kozeniauskas R, Bevill RF, Nelson DR, Thurmon JC. The formation, disposition and hepatic metabolism of dimethylnitrosamine in the pig. Drug Metabolism and Disposition 1990; 18: 626-631.

129. Petters RM, Alexander CA, Wells KD, Collins EB, Sommer JR, Blanton MR, Rojas G, Hao Y, Flowers WL, Banin E, Cideciyan AV, Jacobson SG, Wong F. Genetically engineered large animal model for studying cone photoreceptor survival and degeneration in retinitis pigmentosa. Nat Biotech 1997; 15: 965-970.

130. Dondelinger RF, Ghysels MP, Brisbois D, Donkers E, Snaps FR, Saunders J, Deviere J. Relevant radiological anatomy of the pig as a training model in interventional radiology. Eur Radiol 1998; 8: 1254-1273.