METHODS FOR ENHANCING THE QUALITY OF LIFE OF A SENIOR ANIMAL

Information

  • Patent Application
  • 20110189303
  • Publication Number
    20110189303
  • Date Filed
    July 20, 2009
    15 years ago
  • Date Published
    August 04, 2011
    13 years ago
Abstract
The present invention relates to methods for enhancing the quality of life of a senior or super senior animal by feeding the animal a composition comprising at least one omega-3 polyunsaturated fatty acid and various combinations of amino acids, minerals, and antioxidants in amounts effective to enhance alertness, improve vitality, protect cartilage, maintain muscle mass, enhance digestibility, and improve skin and pelage quality. Beneficial changes in expression of genes associated with several biological pathways may be induced in an animal by feeding it said composition and are consistent with an enhancement in the quality of life of said animal.
Description
FIELD OF THE INVENTION

The present invention relates generally to methods for modulating biological functions associated with the aging process of an animal and particularly to using food compositions containing omega-3 polyunsaturated fatty acids for modulating biological functions associated with the aging process of a senior or super senior animal.


BACKGROUND OF THE INVENTION

Companion animals such as dogs and cats frequently require differing diets depending on their life stage (age), size, body composition, and breed. Both dog and cat nutrient requirements can be separated into three different life-stages, based on age: growing dogs (or cats), adult dogs (or cats), and senior dogs (or cats). The latter category, senior dogs (or cats), can be further separated into two stages, which include senior (or mature adult) and super senior (or geriatric). Dogs are further separated into different categories for regular breed dogs versus large-breed dogs.


Essential fatty acids, consisting of omega-3 and omega-6 polyunsaturated fatty acids, are critical nutrients for the health of an animal. These nutrients, however, either cannot be made by animals or cannot be made in sufficient amounts to elicit benefits and therefore must be consumed in an animal's diet. See, e.g., Hornstra, G., et al., “Essential fatty acids in pregnancy and early human development”, Eur. J. Obs. & Gyn. and Reprod. Biology, 61:57-62 (1995). It has previously been postulated that Docosahexaenoic Acid (“DHA”), an omega-3 polyunsaturated fatty acid, is effective in increasing the maze-learning ability and brain functions in aged mice. See, Lim, S.-Y., “Intakes of dietary docosahexaenoic acid ethyl ester and egg phosphatidylcholine improve maze-learning ability in young and old mice”, J. Nutr., 130:1629-1632 (2000).


Rogers discusses the theory of the potential use of antioxidants to slow the deterioration of cognitive function, particularly in the elderly. See Rogers, P., “A healthy body, a healthy mind: long-term impact of diet on mood and cognitive function”, Proceedings of the Nutrition Society, 60:135-143 (2001).


Despite the studies and developments relating to improving cognitive abilities, there continues to be a need for methods for enhancing the quality of life of senior animals, as measured by, e.g., enhanced alertness, improved vitality, cartilage protection, maintenance of muscle mass, enhanced digestibility, and improved skin and pelage quality in senior and super senior animals.


As previously reported, the super senior pet food composition described herein may be administered to achieve this result. Additionally, we now report herein our surprising discovery that the enhanced quality of life of senior and super senior animals achieved by the administration of the pet food compositions disclosed herein is reflected at the genomic level. Specifically, as described in detail in the Examples below, gene chip data indicate that the expression of genes that encode proteins associated with several biological pathways such as blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport are modified, i.e., in general, the majority are beneficially altered through administration to the animal of the super senior pet food compositions described herein.


SUMMARY OF THE INVENTION

The invention encompasses methods for improving or enhancing the quality of life of senior and super senior animals by feeding the animal a composition comprising at least about 9% by weight protein, at least about 5% by weight fat, and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid.


In one embodiment, the invention encompasses compositions effective to enhance an animal's quality of life, wherein enhanced quality of life is evidenced by improvement in one or more characteristics chosen from alertness, vitality, cartilage protection, muscle mass maintenance, digestibility, and skin and pelage quality.


In another embodiment, the invention encompasses compositions comprising at least one omega-3 polyunsaturated fatty acid chosen from docosahexaenoic acid (“DHA”) and eicosapentaenoic acid (“EPA”). In an additional embodiment, the method comprises feeding the animal a composition further comprising at least one antioxidant and at least one nutrient chosen from choline, manganese, methionine, cysteine, L-carnitine, lysine, and mixtures thereof.


In one embodiment, the invention encompasses compositions effective to improve or enhance the animal's quality of life, wherein enhanced quality of life is evidenced by improvement in one or more biological pathways chosen from blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport.


In another embodiment, the invention encompasses compositions effective to enhance the animal's quality of life, wherein enhanced quality of life is evidenced by a beneficial change in expression of one or more genes which encode proteins associated with or related to biological pathways chosen from blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport.


In yet another embodiment, the invention encompasses methods to treat an animal suffering from a disorder or disease associated with or related to a biological pathway chosen from blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport comprising administering to said animal an effective amount of a composition of the present invention. In one embodiment, the composition includes at least about 9% by weight protein, at least about 5% by weight fat, and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid. In a further embodiment said composition comprises at least one omega-3 polyunsaturated fatty acid chosen from docosahexaenoic acid (“DHA”) and eicosapentaenoic acid (“EPA”). In yet an additional embodiment, the composition further comprises at least one antioxidant and at least one nutrient chosen from choline, manganese, methionine, cysteine, L-carnitine, lysine, and mixtures thereof. In additional embodiments, the composition may comprise the components disclosed in Table 1 or Table 1A.


In another embodiment, the invention encompasses methods of measuring or characterizing the enhancement in the quality of life of an animal, particularly a senior or super senior animal, fed a composition described herein by quantitating the gene expression levels of one or more genes chosen from those disclosed in Tables 5-14 in said animal prior to and after feeding a composition disclosed herein and comparing said levels in the animal wherein an enhancement in the quality of life of said animal is reflected by a beneficial change in gene expression levels in said animal.


Another embodiment encompasses methods of altering the expression of at least one peptide in a mammal, the method comprising administering to the mammal a composition comprising at least about 9% by weight protein; at least about 5% by weight fat; and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid, wherein the at least one peptide is selected from the group consisting of X, Y and Z. With regard to the various embodiments presented herein, it is contemplated herein that the senior or super senior animal may be a senior or super senior large breed canine, regular breed canine, small breed canine or feline.


In another embodiment, the invention encompasses methods for screening one or more test compounds for its ability to alter the expression of at least one gene of interest in a mammal, the method comprising administering a control composition to a control group of mammals and determining the levels of expression of the at least one gene of interest, administering the one or more test compositions to an experimental group of mammals and determining the levels of expression of the least one gene of interest, wherein the test composition comprises at least about 9% by weight protein; at least about 5% by weight fat; and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid, and determining the differences in expression levels in the at least one gene of interest between the control and experimental groups of mammals after each group has been administered their respective compositions, wherein a difference in the expression levels of the at least one gene of interest indicates that the test composition is capable of altering the expression of the at least one gene of interest.


Another embodiment encompasses methods for screening one or more test compounds for its ability to alter the expression of at least one gene of interest in a mammal, the method comprising administering a control composition to a control group of mammals and determining the levels of expression of the at least one gene of interest, wherein the control composition comprises at least about 9% by weight protein; at least about 5% by weight fat; and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid, administering the one or more test compositions to an experimental group of mammals and determining the levels of expression of the least one gene of interest, and determining the differences in expression levels in the at least one gene of interest between the control and experimental groups of mammals after each group has been administered their respective compositions, wherein a difference in the expression levels of the at least one gene of interest indicates that the test composition is capable of altering the expression of the at least one gene of interest.


Other and further objects, features, and advantages of the present invention will be readily apparent to those skilled in the art.







DETAILED DESCRIPTION OF THE INVENTION
Definitions

It is contemplated that the invention described herein is not limited to the particular methodology, protocols, and reagents described as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention in any way.


Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the advantageous methods, devices and materials are now described. All publications mentioned herein are incorporated by reference for the purpose of describing and disclosing the materials and methodologies that are reported in the publication which might be used in connection with the invention.


In practicing the present invention, many conventional techniques in molecular biology may be used. These techniques are well known and are explained in, for example, F. M. Ausubel, Ed. Current Protocols in Molecular Biology, Volumes I, II, and III, (Wiley, New York), 1997; J. Sambrook, E. F. Fritsch., T. Maniatis, Eds., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989).


As used herein and in the appended claims, the singular forms “a”, “an”, and “the” include plural reference unless the context clearly dictates otherwise.


The terms “senior” or “mature adult” refers to the life-stage of an animal. For small or regular breed canines, the “senior” life stage is about 7 to about 10 years of age. For felines, the “senior” life stage is about 7 to about 12 years of age. For large breed canines, over 5 years of age represents “super senior” as described below.


The terms “super senior” or “geriatric” refers to a specific life-stage of an animal. For small or regular breed canines, the super senior stage is any age greater than 10 years of age. For large breed canines, the super senior stage is any age greater than 5 years of age. For felines, the super senior stage is any age greater than 12 years of age.


The term “large breed” canine means a canine that normally weighs about 55 pounds or more when an adult.


The term “regular breed” canine means a canine that normally weighs less than about 55 pounds when an adult.


The term “small breed” canine means a canine that weighs less than about 20 pounds when an adult.


The term “super senior pet food composition” refers to any and all of the pet food compositions disclosed herein.


The term “carbohydrate” as used herein includes polysaccharides (e.g., starches and dextrins) and sugars (e.g. sucrose, lactose, maltose, glucose, and fructose) that are metabolized for energy when hydrolyzed. Examples of carbohydrates suitable for inclusion in the compositions disclosed herein include, but are not limited to, corn, grain sorghum, wheat, barley, and rice.


The term “antioxidant” means a substance that is capable of reacting with free radicals and neutralizing them. Illustrative examples of such substances include beta-carotene, selenium, coenzyme Q10 (ubiquinone), luetin, tocotrienols, soy isoflavones, S-adenosylmethionine, glutathione, taurine, N-acetylcysteine, vitamin E, vitamin C, lipoic acid and L-carnitine. Examples of foods containing useful levels of one or more antioxidants include but are not limited to ginkgo biloba, green tea, broccoli, citrus pulp, grape pomace, tomato pomace, carrot spinach, and a wide variety of fruit meals and vegetable meals. It will be understood by one of skill in the art that while units of antioxidants may be provided herein as “ppm”, appropriate amounts of antioxidants may also be provided as “IU/kg” where appropriate and customary for a given antioxidant such as, e.g., Vitamin E.


The terms “beneficial change” in gene expression, or gene expression may be “beneficially altered” and like terms refer to a modification in gene expression (e.g., up or down regulation of mRNA levels) such that levels of proteins or peptide chains encoded by the genes may be correspondingly modified such that an associated biological pathway may be more likely to function normally, such as in a healthy adult animal and with less tendency to reflect pathological changes in the pathway that, e.g., may be typical of a super senior or geriatric animal. Generally, beneficial changes in gene expression relate to improved health and/or reduced propensity for disease in an animal. As used herein, measuring differences in “gene expression” and like terms refer to, e.g., characterizing whether expression of a gene is up or down regulated in an animal compared to a control level. Gene expression levels can assessed by determining mRNA levels for a corresponding gene, or they may be inferred by determining protein or peptide chain levels. To be clear, determining “gene expression” or “gene expression levels” as used herein includes, but is not limited to, determining either corresponding RNA levels or peptide/protein levels or both. The invention is not limited to a particular method for determining protein or peptide or RNA levels, all of which are well known in the art. Moreover, gene expression and gene expression levels can be assessed in any cell or tissue that is appropriate for expression of the gene of interest. In one embodiment, gene expression is assessed in blood cells. In a more specific embodiment, the blood cells are lymphocytes. In an even more specific embodiment, the cells are T-lymphocytes. Other cell types include, but are not limited to, muscle cells, nerve cells, glial cells, endothelial cells, skin cells, liver cells, kidney cells, bone cells, other types of blood cells, such as but not limited to, macrophages. The cells may be primary cells, i.e., taken directly from an animal, such as cells isolated from recently drawn blood. The cells may also be non-primary, i.e. an established cell line through passage or even an immortalized cell line, such that the methods determining gene expression levels can be performed on established animal cell lines, e.g., CHO cells, prior to administration of a composition to an animal.


As used herein, a “gene” is a DNA molecule where at least a portion of which is transcribed into an RNA molecule. The DNA molecule may or may not include non-transcribed regions and/or non-translated regions, such as but not limited to introns, promoters, enhancer regions, 5′ untranslated regions.


The methods include the genes listed herein, as well as homologs. Thus, the methods of the present invention are not limited to the genes whose database accession numbers are disclosed herein and include homologs thereof. As used herein, a homolog of a gene listed herein means a gene whose coding or non-coding sequence may vary slightly from the reference sequence but also codes for the same or “equivalent” protein or peptide in a different organism. For example, the methods of the present invention relate to expression of phospholipase A2 in at least a canine A homolog of the canine phospholipase A2 gene would include, but would not be limited to, the feline phospholipase A2 gene, the bovine phospholipase A2 gene, the porcine phospholipase A2 gene, the equine phospholipase A2 gene and the primate phospholipase A2 gene. Homologs also include variations in the coding or non-coding sequences that account for slight variations across species. For example, the present invention relates to the human phospholipase A2 gene, and a homolog thereof would include, but would not be limited to a monkey or chimpanzee phospholipase A2 gene.


As used herein, “improving” or “enhancing” the quality of life of an animal refers to as an improvement or enhancement in one or more characteristics chosen from alertness, vitality, protection of cartilage, maintenance of muscle mass, digestibility, and skin and pelage quality. Additionally, improvement/enhancement in blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport are also contemplated.


An “improvement” or an “enhancement” in a characteristic or biological pathway refers to a modification in said characteristic or biological pathway such that there is a tendency for the characteristic or pathway to appear and/or function normally and with less tendency to reflect pathological changes in the characteristic or pathway that, e.g., may be typical of a super senior animal.


As used herein, methods to “treat” an animal suffering from a disease or disorder is also meant to encompass methods to prevent and/or to ameliorate the disease or disorder as well.


As used herein, “genes associated with the aging process” or “aging genes” or like terms refers to those genes which may be involved in the process of senescence in an animal. These genes may include, e.g., genes that encode for proteins that have a role in a number of biological functions such as inflammation, DNA repair or cell survival, fat or cholesterol metabolism, protein synthesis, immune regulation, cell growth and cell death.


Similarly, the “aging process”, as the term is used herein, refers to the process of senescence in an animal and may include changes in biological functions such as, e.g., inflammation, DNA repair or cell survival, fat or cholesterol metabolism, protein synthesis, cell growth and cell death.


As used herein, the phrase “modulating biological functions associated with the aging process” refers to op-regulating or down-regulating genes, which may be involved in the process of senescence in an animal. These genes may include, e.g., genes that encode for proteins that have a role in a number of biological functions such as inflammation, DNA repair or cell survival, fat or cholesterol metabolism, protein synthesis, immune regulation, cell growth and cell death.


The Invention

The present invention encompasses compositions and methods for improving or enhancing the quality of life of a senior or super senior animal. The methods comprise feeding the animal a composition comprising at least about 9% by weight protein, at least about 5% by weight fat, and at least about 0.05% by weight omega-3 polyunsaturated fatty acid. The methods are useful for enhancing alertness, improving vitality, protecting cartilage, maintaining muscle mass, enhancing digestibility, and improving skin and pelage quality in a senior or super senior animal. The methods are also useful for improving in an animal one or more biological pathways chosen from blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and the electron transport pathway, such improvements also being reflected in overall beneficial changes at the genomic level. Methods for treating animals suffering from disorders or diseases associated with or related to these biological pathways comprising administering the compositions of the present invention are also contemplated herein.


Without being bound by theory, the benefits of the invention may be the result of physiological effects from the addition of omega-3 polyunsaturated fatty acids to a senior or super senior animal's diet. Similarly, the antioxidants, choline, and other nutrients may play a role in enhancing a senior or super senior animal's quality of life.


Although the methods of the present invention may improve an animal's quality of life by enhancing all of the above described characteristics or improving all of the described biological pathways, it is not necessary to demonstrate substantial improvements in each of the characteristics or pathways to achieve the “enhanced quality of life” as defined herein.


When the compositions are administered to a senior or super senior animal, the animal experiences an enhanced quality of life, e.g., exhibits or experiences one or more of enhanced alertness, improved vitality, protected cartilage, maintained muscle mass, enhanced digestibility, improved skin and pelage quality, as well as improvements in e.g., blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process and the electron transport pathway as indicated by overall beneficial changes at the genomic level. Methods for determining these measurements of quality of life are known to skilled artisans. For example, alertness can be measured by various means, including an analysis of metabolism and antioxidant markers, as well as through clinical studies with follow-up questions to participating pet owners. Potential metabolism markers may include ghrelin, GLP-1, thyroid hormone, and/or growth hormone. Potential markers of antioxidant status may include serum vitamin E, ORAC, glutathione peroxidase, alkanels, and/or cell damage indicators. Further, vitality can be measured by various means, including an analysis of metabolism and antioxidant markers, as well as through clinical studies with follow-up questions to participating pet owners. Similarly, cartilage protection can be measured by various means, including an analysis of arthritis biomarkers. Potential arthritis biomarkers may include type II collagen synthesis, matrix metaloproteinase, osteocalcin, alkaline phosphatase activity, COMP, and fragments of cartilage damage. Muscle mass maintenance can be measured by various means, including an analysis of body composition and digestibility can be measured by various means, including clinical studies with follow-up questions to participating pet owners and animal feeding to determine the percentage of nutrients digested Skin and pelage quality can be measured by various means, including clinical studies with follow-up questions to participating pet owners. Additionally, as discussed above, improvements in quality of life is also reflected at the genomic level, as evidenced by gene chip data which indicate beneficial changes on the expression of genes associated with various important biological pathways including blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and protection and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and the electron transport pathway. The identities of these genes are provided in the Examples below.


The methods of the invention are useful for enhancing the quality of life of humans and animals, including primates (e.g., monkeys, chimpanzees, etc.), companion animals (e.g., dogs, cats, horses, etc.), farm animals (e.g., goats, sheep, swine, cattle, etc.), laboratory animals (e.g., mice, rats, etc.), birds (e.g., domestic birds such as canaries, parrots, etc. and commercial birds such as chickens, ducks, turkeys, etc.), rodents (e.g., hamsters, guinea pigs, gerbils, rabbits, hedgehogs, ferrets, chinchillas, etc.), and wild, exotic, and zoo animals (e.g., wolves, bears, deer, etc.). In various embodiments, the animal is a cat, a dog, or a horse.


The compositions of the present invention are designed to enhance digestibility and improve chewability. Canine and feline foods are typically formulated based on life stage (age), size, body composition, and breed. Thus, some embodiments of the present invention include compositions that are formulated to address specific nutritional differences between regular or small breed dogs, large breed dogs, and cats.


The invention provides methods utilizing a variety of compositions containing at least one omega-3 polyunsaturated fatty acid. The compositions include foods, supplements, treats, and toys (typically chewable and consumable toys). The methods also provide the compositions to the designated animals over a period of time that is long enough to effectuate the improved quality of life. In one embodiment, the method provides the animal with a composition for at least thirty days.


The compositions for use in the methods of the present invention generally have an omega-3 polyunsaturated fatty acid content of at least about 0.02% (or about 0.05% to about 10%, or about 0.1% to about 6%) by weight on a dry matter basis. In some embodiments, the omega-3 polyunsaturated fatty acid is DHA. In other embodiments, the omega-3 polyunsaturated fatty acid is EPA. In still other embodiments, the omega-3 polyunsaturated fatty acid comprises a mixture of DHA and EPA.


In some embodiments, the composition containing omega-3 polyunsaturated fatty acid is a food. Although both liquid and solid foods are provided, solid foods are typically advantageous. Foods include both dry foods and wet foods. Some of the non-polyunsaturated fatty acid components of the food, and useful proportions, include those listed in Table 1.










TABLE 1






Proportion of the composition (% of dry weight of


Component
composition or parts per million)







Protein
about 9% to about 55%, or about 18% to about 30%, or



about 33% to about 55% or about 18% to about 20% or



about 33% to about 36%


Fat
about 7% to about 35%, or about 18% to about 35%, or



about 7% to about 24%, or about 14% to about 24%, or



about 14% to about 16% or about 18% to about 24%


Antioxidant
about 0 ppm to about 7500 ppm, or about 0.05 ppm to



about 3600 ppm, or about 250 to about 3600, or about 250



ppm to about 1650 ppm, or about 5 ppm to about 225 ppm,



or about 0.05 ppm to about 2.4 ppm









In one embodiment, the methods of this invention comprise feeding a super senior animal a composition in an amount effective to enhance the animal's quality of life. Such compositions generally comprise:

    • (a) 0.02% (or about 0.05% to about 10%, or about 0.1% to about 6%) of at least one omega-3 polyunsaturated fatty acid, and
    • (b) at least one of the following:
      • (i) about 10% to about 55% (or about 18% to about 30%, or about 33% to about 55% or about 18% to about 20% or about 33% to about 36%) protein,
      • (ii) about 7% to about 35% (or about 18% to about 35%, or about 7% to about 24%, or about 14% to about 24%, or about 14% to about 16% or about 18% to about 24%) fat, and
      • (iii) at least about 0.05 (or about 0.05 ppm or IU/kg to about 7500 ppm or IU/kg, or about 250 ppm or IU/kg to about 3600 ppm or IU/kg, or about 250 ppm or IU/kg to about 1650 ppm or IU/kg, or about 5 ppm or IU/kg to about 225 ppm or IU/kg, or about 0.05 ppm or IU/kg to about 2.4 ppm or IU/kg) antioxidant.


In another embodiment, the methods of this invention comprise feeding a super senior regular or small breed canine a composition in an amount effective to enhance the canine's quality of life. The composition generally comprises:

    • (a) at least one of the following:
      • (i) at least about 0.02% (or about 0.02% to about 0.3%, or about 0.05% to about 0.3%, or about 0.05% to about 0.2%) DHA, and
      • (ii) at least about 0.1% (or about 0.1% to about 0.5%, or about 0.2% to about 0.5%, or about 0.2% to about 0.3%) EPA,
    • (b) at least about 9% (or about 9% to about 30%, or about 18% to about 30%, or about 18% to about 20%) protein,
    • (c) at least about 7% (or about 7% to about 24%, or about 14% to about 24%, or about 14% to about 16%) fat, and
    • (d) at least one of the following:
      • (i) at least about 250 IU/kg (or about 250 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1000 IU/kg) vitamin E,
      • (iv) at least about 50 ppm (or about 50 ppm to about 500 ppm, or about 100 ppm to about 500 ppm, or about 100 ppm to about 301 ppm) vitamin C,
      • (v) at least about 600 ppm (or about 600 ppm to about 2400 ppm, or about 1260 ppm to about 2400 ppm, or about 1260 ppm to about 1545 ppm) taurine,
      • (vi) at least about 50 ppm (or about 50 ppm to about 200 ppm, or about 100 to about 160, or about 100 to about 155) lipoic acid, and
      • (vii) at least about 50 ppm (or about 50 ppm to about 500 ppm, or about 200 ppm to about 500 ppm, or about 200 ppm to about 350 ppm) carnitine.


In another embodiment, the methods of this invention comprise feeding a super senior large breed canine a composition in an amount effective to enhance the canine's quality of life. The compositions generally comprise:

    • (a) at least one of the following:
      • (i) at least about 0.02% (or about 0.02% to about 0.3%, or about 0.05% to about 0.3%, or about 0.05% to about 0.2%) DHA, and
      • (ii) at least about 0.1% (or about 0.1% to about 0.5%, or about 0.2% to about 0.5%, or about 0.2% to about 0.3%) EPA,
    • (b) at least about 9% (or about 9% to about 30%, or about 18% to about 30%, or about 18% to about 20%) protein,
    • (c) at least about 7% (or about 7% to about 24%, or about 14% to about 24%, or about 14% to about 16%) fat, and
    • (d) at least one of the following:
      • (i) at least about 250 IU/kg (or about 250 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1000 IU/kg) vitamin E,
      • (viii) at least about 50 ppm (or about 50 ppm to about 500 ppm, or about 100 ppm to about 500 ppm, or about 100 ppm to about 301 ppm) vitamin C,
      • (ix) at least about 600 ppm (or about 600 ppm to about 2400 ppm, or about 1260 ppm to about 2400 ppm, or about 1260 ppm to about 1575 ppm) taurine, and
      • (x) at least about 50 ppm (or about 50 ppm to about 200 ppm, or about 100 to about 160, or about 100 to about 155) lipoic acid, and
      • (xi) at least about 50 ppm (or about 50 ppm to about 500 ppm, or about 200 ppm to about 500 ppm, or about 200 ppm to about 350 ppm) carnitine.


In another embodiment, the methods of this invention comprise feeding a super senior feline a composition in an amount effective to enhance the feline's quality of life. The compositions generally comprise:

    • (a) at least one of the following:
      • (i) at least about 0.05% (or about 0.05% to about 0.30%, or about 0.1% to about 0.30%, or about 0.1% to about 0.2%) DHA, and
      • (ii) at least about 0.1% (or about 0.1% to about 0.5%, or about 0.2% to about 0.5%, or about 0.2% to about 0.3%) EPA,
    • (b) at least about 15% (or about 15% to about 55%, or about 30% to about 55%, or about 33% to about 36%) protein,
    • (c) at least about 9% (or about 9% to about 35%, or about 18% to about 35%, or about 18% to about 24%) fat, and
    • (d) at least one of the following:
      • (i) at least about 250 IU/kg (or about 250 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1100 IU/kg) vitamin E,
      • (xii) at least about 50 ppm (or about 50 ppm to about 300 ppm, or about 100 ppm to about 300 ppm, or about 100 ppm to about 200 ppm) vitamin C,
      • (xiii) at least about 1100 ppm (or about 1100 ppm to about 3500 ppm, or about 2300 ppm to about 3500 ppm, or about 2300 ppm to about 2350 ppm) taurine, and
      • (xiv) at least about 200 ppm (or about 200 to about 750 ppm, or about 400 ppm to about 750 ppm, or about 400 to about 525 ppm) carnitine, and
      • (xv) at least about 0.05% (or about 0.05% to about 0.6%, or about 0.1% to about 0.6%, or about 0.1% to about 0.4%) cystine.


In another embodiment, the methods of this invention comprise feeding a super senior animal a composition in an amount effective to enhance the animal's alertness and vitality. The composition generally comprises:

    • (a) 0.02% (or about 0.05% to about 10%, or about 0.1% to about 6%) at least one omega-3 polyunsaturated fatty acid, and
    • (b) at least one of the following:
      • (xvi) about 10% to about 55% (or about 18% to about 30%, or about 33% to about 55% or about 18% to about 20% or about 33% to about 36%) protein,
      • (xvii) about 7% to about 35% (or about 18% to about 35%, or about 7% to about 24%, or about 14% to about 24%, or about 14% to about 16% or about 18% to about 24%) fat,
      • (xviii) at least about 0.05 (or about 0.05 ppm to about 7500 ppm, or about 250 to about 3600, or about 250 ppm to about 1650 ppm, or about 5 ppm to about 225 ppm, or about 0.05 ppm to about 2.4 ppm) antioxidant, and
      • (xix) at least about 1000 ppm (or about 1000 ppm to about 5000 ppm, about 3300 ppm to about 5000 ppm, or about 2000 ppm to about 3000 ppm, or about 3000 ppm to about 4000 ppm) choline.


In another embodiment, the methods of this invention comprise feeding a super senior regular or small breed canine a composition in an amount effective to enhance the canine's alertness and vitality. The composition generally comprises:

    • (a) at least one of the following:
      • (i) at least about 0.02% (or about 0.02% to about 0.3%, or about 0.05% to about 0.3%, or about 0.05% to about 0.2%) DHA, and (ii) at least about 0.1% (or about 0.1% to about 0.5%, or about 0.2% to about 0.5%, or about 0.2% to about 0.3%) EPA,
    • (b) at least about 9% (or about 9% to about 30%, or about 18% to about 30%, or about 18% to about 20%) protein,
    • (c) at least about 7% (or about 7% to about 24%, or about 14% to about 24%, or about 14% to about 16%) fat,
    • (d) at least one of the following:
      • (i) at least about 250 IU/kg (or about 250 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1000 IU/kg) vitamin E,
      • (xx) at least about 50 ppm (or about 50 ppm to about 500 ppm, or about 100 ppm to about 500 ppm, or about 100 ppm to about 301 ppm) vitamin C,
      • (xxi) at least about 600 ppm (or about 600 ppm to about 2400 ppm, or about 1260 ppm to about 2400 ppm, or about 1260 ppm to about 1545 ppm) taurine, and
      • (xxii) at least about 50 ppm (or about 50 ppm to about 200 ppm, or about 100 to about 160, or about 100 to about 155) lipoic acid, and
      • (xxiii) at least about 50 ppm (or about 50 ppm to about 500 ppm, or about 200 ppm to about 500 ppm, or about 200 ppm to about 350 ppm) carnitine,
    • (e) at least about 1000 ppm (or about 1000 ppm to about 3200 ppm, or about 2000 ppm to about 3200 ppm, or about 2000 ppm to about 2500 ppm) choline,
    • (f) at least about 50 ppm (or about 50 ppm to about 150 ppm, or about 100 ppm to about 150 ppm, or about 100 ppm to about 110 ppm) manganese, and
    • (g) at least about 0.4% (or about 0.4% to about 2%, or about 0.9% to about 2%, or about 0.9% to about 1.2%) lysine, and
    • (h) at least about 0.4% to about 1.5% methionine.


In another embodiment, the methods of this invention comprise feeding a super senior large breed canine a composition in an amount effective to enhance the canine's alertness and vitality. The composition generally comprises:

    • (a) at least one of the following:
      • (i) at least about 0.02% (or about 0.02% to about 0.3%, or about 0.05% to about 0.3%, or about 0.05% to about 0.2%) DHA, and
      • (ii) at least about 0.1% (or about 0.1% to about 0.5%, or about 0.2% to about 0.5%, or about 0.2% to about 0.3%) EPA,
    • (b) at least about 9% (or about 9% to about 30%, or about 18% to about 30%, or about 18% to about 20%) protein,
    • (c) at least about 7% (or about 7% to about 24%, or about 14% to about 24%, or about 14% to about 16%) fat,
    • (d) at least one of the following:
      • (i) at least about 250 IU/kg (or about 250 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1000 IU/kg) vitamin E,
      • (xxiv) at least about 50 ppm (or about 50 ppm to about 500 ppm, or about 100 ppm to about 500 ppm, or about 100 ppm to about 301 ppm) vitamin C,
      • (xxv) at least about 600 ppm (or about 600 ppm to about 2400 ppm, or about 1260 ppm to about 2400 ppm, or about 1260 ppm to about 1575 ppm) taurine, and
      • (xxvi) at least about 50 ppm (or about 50 ppm to about 200 ppm, or about 100 to about 160, or about 100 to about 155) lipoic acid, and
      • (xxvii) at least about 50 ppm (or about 50 ppm to about 500 ppm, or about 200 ppm to about 500 ppm, or about 200 ppm to about 350 ppm) carnitine,
    • (e) at least about 1000 ppm (or about 1000 ppm to about 3200 ppm, or about 2000 ppm to about 3200 ppm, or about 2000 ppm to about 2500 ppm) choline,
    • (f) at least about 50 ppm (or about 50 ppm to about 150 ppm, or about 100 ppm to about 150 ppm, or about 100 ppm to about 110 ppm) manganese, and
    • (g) at least about 0.4% (or about 0.4% to about 2%, or about 0.9% to about 2%, or about 0.9% to about 1.2%) lysine, and
    • (h) at least about 0.4% to about 1.5% methionine.


In another embodiment, the methods of this invention comprise feeding a super senior feline a composition in an amount effective to enhance the feline's alertness and vitality. The composition generally comprises:

    • (a) at least one of the following:
      • (i) at least about 0.05% (or about 0.05% to about 0.30%, or about 0.1% to about 0.30%, or about 0.1% to about 0.2%) DHA, and
      • (ii) at least about 0.1% (or about 0.1% to about 0.5%, or about 0.2% to about 0.5%, or about 0.2% to about 0.3%) EPA,
    • (b) at least about 15% (or about 15% to about 55%, or about 30% to about 55%, or about 33% to about 36%) protein,
    • (c) at least about 9% (or about 9% to about 35%, or about 18% to about 35%, or about 18% to about 24%) fat,
    • (d) at least one of the following:
      • (i) at least about 250 IU/kg (or about 250 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1500 IU/kg, or about 500 IU/kg to about 1100 IU/kg) vitamin E,
      • (xxviii) at least about 50 ppm (or about 50 ppm to about 300 ppm, or about 100 ppm to about 300 ppm, or about 100 ppm to about 200 ppm) vitamin C,
      • (xxix) at least about 1100 ppm (or about 1100 ppm to about 3500 ppm, or about 2300 ppm to about 3500 ppm, or about 2300 ppm to about 2350 ppm) taurine, and
      • (xxx) at least about 200 ppm (or about 200 to about 750 ppm, or about 400 ppm to about 750 ppm, or about 400 to about 525 ppm) carnitine, and
      • (xxxi) at least about 0.05% (or about 0.05% to about 0.6%, or about 0.1% to about 0.6%, or about 0.1% to about 0.4%) cystine,
    • (e) at least about 1600 ppm (or about 1600 ppm to about 5000 ppm, or about 3300 ppm to about 5000 ppm, or about 3300 ppm to about 3400 ppm) choline,
    • (f) at least about 50 ppm (or about 50 ppm to about 150 ppm, or about 100 ppm to about 150 ppm, or about 100 ppm to about 110 ppm) manganese, and
    • (g) at least about 0.7% (or about 0.7% to about 3%, or about 1.4% to about 3%, or about 1.4% to about 1.7%) lysine, and
    • (h) at least about 0.4% to about 1.5% methionine.


In another embodiment, this invention provides a method for improving the quality of life of a senior or super senior small or regular breed canine The method comprises feeding the canine a composition comprising:

    • about 60% to about 70% by weight carbohydrate;
    • about 15% to about 25% by weight protein chosen from animal protein and vegetable protein;
    • about 5% to about 7% by weight fat chosen from animal fat and vegetable fat;
    • about 2.5% to about 4% by weight of at least one omega-3 polyunsaturated fatty acids;
    • about 1% to about 4% by weight fiber;
    • about 1% to about 2% by weight minerals; and
    • about 0.5 to about 1.5% by weight vitamins.


In another embodiment, this invention provides a method for improving the quality of life of a senior or super senior large breed canine. The method comprises feeding the canine a composition comprising:

    • about 60% to about 70% by weight carbohydrate;
    • about 15% to about 25% by weight protein chosen from animal protein and vegetable protein;
    • about 5% to 10% by weight fat chosen from animal fat and vegetable fat;
    • about 3% to about 5% by weight of at least one omega-3 polyunsaturated fatty acids;
    • about 1% to about 4% by weight fiber;
    • about 0.5% to about 1% by weight minerals; and
    • about 0.75 to about 1.25% by weight vitamins.


In another embodiment, this invention provides a method for improving the quality of life of a senior or super senior feline. The method comprises feeding the feline a composition comprising:

    • about 30% to about 35% by weight carbohydrate;
    • about 35% to about 50% by weight protein chosen from animal protein and vegetable protein;
    • about 12% to about 15% by weight fat chosen from animal fat and vegetable fat;
    • about 1% to about 2% by weight of at least one omega-3 polyunsaturated fatty acids;
    • about 1% to about 5% by weight fiber;
    • about 1% to about 2% by weight minerals; and
    • about 1% to about 2% by weight vitamins.


In a further embodiment, this invention provides a method for improving the quality of life of a senior or super senior animal comprising feeding the animal (e.g., small, regular or large breed canine or feline, as the case may be) a composition comprising the components as indicated in Table 1A below:









TABLE 1A







Chemical composition of Super Senior


Foods











Small/Regular





Breed
Large Breed


Nutrient Component
Canine
Canine
Feline













Crude Protein, %
20.1
19.34
35.73


Fat, %
16.45
16.92
22.47


Calcium, %
0.71
0.73
0.94


Phosphorus, %
0.61
0.68
0.77


EPA, %
0.32
0.32
0.23


DHA, %
0.22
0.22
0.32


Linoleic Acid, %
3.96
4.04
5.05


Total N-3 fatty acids, %
1.3
2.24
1.14


Total N-6 fatty acids, %
3.96
3.99
5.09


Taurine, ppm
1400
15.25
2100


Carnitine, ppm
314
337
367


Methioinine, %
1
1.19
1.32


Cystine, %
0.25
0.24
0.47


Manganese, ppm
87
100
104


Vitamin E, IU/kg
1492
1525
1292


Vitamin C, ppm
127
261
141


Lipoic Acid, ppm*
101
135





*Lipoic acid based on formulated, not analyzed values.






The compositions for use in the methods of this invention further comprise at least one nutrient chosen from manganese, methionine, cysteine, mixtures of methionine and cysteine, L-carnitine, lysine, and arginine. Specific advantageous amounts for each component in a composition will depend on a variety of factors including, for example, the species of animal consuming the composition; the particular components included in the composition; the age, weight, general health, sex, and diet of the animal; the animal's consumption rate, and the like. Thus, the component amounts may vary widely, and may even deviate from the proportions given herein.


The omega-3 fatty acids may be obtained from a variety of sources. One convenient source is fish oils from, for example, menhaden, mackerel, herring, anchovy, and salmon. DHA and EPA are typical fatty acids present in such fish oils, and, together often make up a significant portion of the oil, such as about 25% to about 38% of the oil.


When the composition is an animal food, vitamins and minerals preferably are included in amounts required to avoid deficiency and maintain health. These amounts are readily available in the art. The National Research Council (NRC), for example, provides recommended amounts of such ingredients for farm animals. See, e.g., Nutrient Requirements of Swine (10th Rev. Ed., Nat'l Academy Press, Wash. D.C., 197298), Nutrient Requirements of Poultry (9th Rev. Ed., Nat'l Academy Press, Wash. D.C., 1994), Nutrient Requirements of Horses (Fifth Rev. Ed., Nat'l Academy Press, Wash. D.C., 1989), Nutrient Requirements of Dogs and Cats (Nat'l Academy Press, Wash. D.C., 2006). The American Feed Control Officials (AAFCO), for example, provides recommended amounts of such ingredients for dogs and cats. See American Feed Control Officials, Inc., Official publication, pp. 126-140 (2003). Examples of vitamins useful as food additives include vitamin A, B1, B2, B6, B12, C, D, E, K, H (biotin), K, folic acid, inositol, niacin, and pantothenic acid. Examples of minerals and trace elements useful as food additives include calcium, phosphorus, sodium, potassium, magnesium, copper, zinc, chloride, and iron salts.


The methods of the present invention include compositions that may further contain other additives known in the art. Preferably, such additives are present in amounts that do not impair the purpose and effect provided by the invention. Examples of additives include, for example, substances with a stabilizing effect, processing aids, substances that enhance palatability, coloring substances, and substances that provide nutritional benefits.


Stabilizing substances include, for example, substances that tend to increase the shelf life of the composition. Potentially suitable examples of such substances include, for example, preservatives, antioxidants, synergists and sequestrants, packaging gases, stabilizers, emulsifiers, thickeners, gelling agents, and humectants. Examples of emulsifiers and/or thickening agents include, for example, gelatin, cellulose ethers, starch, starch esters, starch ethers, and modified starches.


Additives for coloring, palatability (“pal enhancers”), and nutritional purposes include, for example, colorants (e.g., iron oxide, such as the red, yellow, or brown forms); sodium chloride, potassium citrate, potassium chloride, and other edible salts; vitamins; minerals; and flavoring. Such additives are known in the art. See, e.g., U.S. Pat. No. 3,202,514. See also, U.S. Pat. No. 4,997,671. Flavorants include, for example, dairy product flavorants (e.g., milk or cheese), meat flavorants (e.g., bacon, liver, beef, poultry, or fish), oleoresin, pinacol, and the various flavorants identified in the trade by a FEMA (Flavor Extract Manufacturers Association) number. Flavorants help provide additional palatability, and are known in the art. See, e.g., U.S. Pat. No. 4,997,672. See also, U.S. Pat. No. 5,004,624. See also, U.S. Pat. No. 5,114,704. See also, U.S. Pat. No. 5,532,010. See also, U.S. Pat. No. 6,379,727. The concentration of such additives in the composition typically may be up to about 5% by weight. In some embodiments, the concentration of such additives (particularly where such additives are primarily nutritional balancing agents, such as vitamins and minerals) is about 0% to about 2.0% by weight. In some embodiments, the concentration of such additives (again, particularly where such additives are primarily nutritional balancing agents) is about 0% to about 1.0% by weight.


Supplements include, for example, a feed used with another feed to improve the nutritive balance or performance of the total. Supplements include compositions that are fed undiluted as a supplement to other feeds, offered free choice with other parts of an animal's ration that are separately available, or diluted and mixed with an animal's regular feed to produce a complete feed. The AAFCO, for example, provides a discussion relating to supplements in the American Feed Control Officials, Inc. Official Publication, p. 220 (2003). Supplements may be in various forms including, for example, powders, liquids, syrups, pills, encapsulated compositions, and the like.


Treats include, for example, compositions that are given to an animal to entice the animal to eat during a non-meal time. Treats for canines include, for example, dog bones. Treats may be nutritional, wherein the composition comprises one or more nutrients, and may, for example, have a composition as described above for food. Non-nutritional treats encompass any other treats that are non-toxic.


Toys include, for example, chewable toys. Toys for dogs include, for example, artificial bones. There is a wide range of suitable toys currently marketed. See, e.g., U.S. Pat. No. 5,339,771 (and references disclosed in U.S. Pat. No. 5,339,771). See also, e.g., U.S. Pat. No. 5,419,283 (and references disclosed in U.S. Pat. No. 5,419,283). The invention provides both partially consumable toys (e.g., toys comprising plastic components) and fully consumable toys (e.g., rawhides and various artificial bones). It should be further recognized that this invention provides toys for both human and non-human use, particularly for companion, farm, and zoo animal use, and particularly for dog, cat, or bird use.


A “food” is a nutritionally complete diet for the intended recipient animal (e.g., domestic cat or domestic dog). A “nutritionally complete diet” is a diet that includes sufficient nutrients for maintenance of normal health of a healthy animal on the diet. The methods of this invention utilize compositions that are not intended to be restricted by any specific listing of proteinaceous or fat ingredients or product form. The compositions can be prepared in, for example, a dry, canned, wet, or intermediate moisture form using conventional pet food processes. In some embodiments, the moisture content is about 10% to about 90% of the total weight of the composition. In other embodiments, the moisture content is about 65% to about 75% of the total weight of the composition.


In preparing a composition for use with the methods of the present invention, any ingredient (e.g., fish oil) generally may, for example, be incorporated into the composition during the processing of the formulation, such as during and/or after mixing of other components of the composition. Distribution of these components into the composition can be accomplished by conventional means. In one embodiment, ground animal and poultry proteinaceous tissues are mixed with the other ingredients, including fish oils, cereal grains, other nutritionally balancing ingredients, special-purpose additives (e.g., vitamin and mineral mixtures, inorganic salts, cellulose and beet pulp, bulking agents, and the like); and water that is sufficient for processing is also added. These ingredients preferably are mixed in a vessel suitable for heating while blending the components. Heating of the mixture may be effected using any suitable manner, such as, for example, by direct steam injection or by using a vessel fitted with a heat exchanger. Following the addition of the last ingredient, the mixture is heated to a temperature range of about 50° F. (10° C.) to about 212° F. (100° C.). In some embodiments, the mixture is heated to a temperature range of about 70° F. (21° C.) to about 140° F. (60° C.). Temperatures outside these ranges are generally acceptable, but may be commercially impractical without use of other processing aids. When heated to the appropriate temperature, the material will typically be in the form of a thick liquid. The thick liquid is filled into cans. A lid is applied, and the container is hermetically sealed. The sealed can is then placed into conventional equipment designed to sterilize the contents. This is usually accomplished by heating to temperatures of greater than about 230° F. (110° C.) for an appropriate time, which is dependent on, for example, the temperature used and the composition.


Methods of the present invention include utilizing compositions that can be prepared in a dry form using conventional processes. In one embodiment, dry ingredients, including, for example, animal protein sources, plant protein sources, grains, etc., are ground and mixed together. Moist or liquid ingredients, including fats, oils, animal protein sources, water, etc., are then added to and mixed with the dry mix. The mixture is then processed into kibbles or similar dry pieces. Kibble is often formed using an extrusion process in which the mixture of dry and wet ingredients is subjected to mechanical work at a high pressure and temperature, and forced through small openings and cut off into kibble by a rotating knife. The wet kibble is then dried and optionally coated with one or more topical coatings which may include, for example, flavors, fats, oils, powders, and the like. Kibble also can be made from the dough using a baking process, rather than extrusion, wherein the dough is placed into a mold before dry-heat processing.


The compositions are also designed to be easier to chew. Canine and feline foods are typically formulated based on life stage (age), size, body composition, and breed. In the methods of this invention, some embodiments of the compositions address specific nutritional differences between super senior regular or small breed dogs, large breed dogs, and cats.


All percentages expressed herein are on a weight by dry matter basis unless specifically stated otherwise.


As noted previously, this invention is directed, in part, to a method for enhancing the quality of life of an animal. The method comprises feeding a senior or super senior animal a composition in an amount effective to enhance alertness, improve vitality, protect cartilage, maintain muscle mass, enhance digestibility, and improve skin and pelage quality. Additionally, we now report herein our surprising discovery that the enhanced quality of life of an animal achieved by administration of the compositions of the present invention is reflected at the genomic level. While it may be that a change in expression of any one gene disclosed in the tables presented below may result in beneficial or deleterious biological effects, the data presented herein indicate that, overall, the observed expression profiles are consistent with the beneficial biological effects seen in vivo after administration of the diets disclosed herein. Specifically, gene chip data indicate that the expression of genes that encode proteins associated with or related to several biological pathways such as blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport are, for the most part, beneficially altered through administration to the animal of compositions described herein. Thus, the invention also relates to methods of measuring or characterizing the enhancement in the quality of life of an animal, particularly a senior or super senior animal, fed a composition described herein by quantitating the gene expression levels of one or more genes chosen from those disclosed in Tables 5-14 in said animal prior to and after feeding a composition disclosed herein and comparing said levels in the animal wherein an enhancement in the quality of life of said animal is reflected by a beneficial change in gene expression levels in said animal.


Quantitation of gene expression may be carried out in numerous ways familiar to one of skill in the art and include such techniques as RT PCR as well as gene chip assays and Northern blotting. Thus, it is contemplated herein that the expression levels detected may be used, for example, in methods to measure enhancement in the quality of life of an animal as disclosed herein.


There are certain age-induced changes in gene expression patterns (see, for example, P. Tollet-Egnell et al., Molecular Endocrinology, 15(2):308-318 (2001)). Without being bound by theory, such changes in gene expression patterns may be related to senescence, the aging mechanism. C-K Lee et al., Science, 285:1390-1393 (1999) reported that alterations in the gene expression profile of the aging process in mice can be completely or partially prevented by caloric restriction. We have found that, surprisingly, the changes in expression of certain genes as an animal, such as a dog, ages from a healthy adult animal to a geriatric animal can be reversed by a diet of super senior dog food according to the present invention. Thus, comparing the gene expression pattern in a healthy adult dog to the gene expression pattern in a geriatric dog, one finds certain genes expressed higher (“up”) in the geriatric dog while other genes are expressed lower (“down”). Surprisingly, we have found that by feeding a diet of super senior dog food according to the present invention to a geriatric dog, the gene expression pattern can be reversed. That is, comparing the gene expression pattern in a geriatric dog fed a control diet to the gene expression pattern in a geriatric dog fed a diet of super senior dog food of the present invention, one finds that certain genes are expressed higher (“up”) under the control dog food regimen, while other genes are expressed lower (“down”) under the control dog food regimen. The result is that the geriatric dogs under the super senior dog food diet of the present invention had their gene expression profiles altered towards that of healthy adult dogs. Comparing the list of genes that correlate in the opposite sense to the healthy adult dog/geriatric dog expression pattern, we found genes provided in Tables 15-20 below that surprisingly demonstrate that the super senior dog food of the present invention can reverse the alteration in expression that certain genes undergo as a part of the aging process. Thus, the quality of life of geriatric animals can be benefited by modifying the aging process in that the gene expression pattern of certain genes are altered towards that of a healthy adult dog from the pattern of a geriatric dog.


Accordingly, this invention is directed, in part, to a method for enhancing the quality of life of an animal comprising feeding a senior or super senior animal a composition in an amount effective to alter the gene expression pattern of certain genes (provided on Tables 15-20 where the direction of adult vs geriatric is the same as the direction of super senior vs control) towards the pattern of a healthy adult dog form the pattern of a geriatric dog. The method enhances the quality of life of an animal by modifying the expression of genes associated with the aging process such that the gene expression pattern is altered towards that of a healthy adult animal from that of a geriatric animal.


In one aspect, this invention is directed to a method for improving the quality of life of a senior or super senior animal comprising feeding the animal a composition comprising at least about 9% by weight protein; at least about 5% by weight fat; and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid, wherein the method comprises feeding the animal the composition in an amount effective to enhance the animal's quality of life, wherein enhanced quality of life is evidenced by a change in expression of one or more genes which encode proteins associated with the aging process. As described herein, these genes are generally referred to as genes associated with the aging process, however, it should be noted that these genes specifically may be related to biological pathways chosen from, e.g., inflammation, DNA repair, cell survival, fat or cholesterol metabolism, immune regulation, protein synthesis, cell growth and cell death.


In an embodiment of this aspect, the change in expression is of one or more genes listed on Tables 15-19 and wherein the change in expression is towards the expression level in a healthy adult animal as compared to the expression level in a geriatric animal.


In another embodiment of this aspect, the animal is a dog.


In another aspect, this invention is directed to a method for improving the quality of life of a senior or super senior animal comprising feeding the animal a composition comprising at least about 9% by weight protein; at least about 5% by weight fat; and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid, wherein the method comprises feeding the animal the composition in an amount effective to enhance the animal's quality of life, wherein enhanced quality of life is evidenced by a change in expression of one or more genes listed on Table 20 and wherein the change in expression is towards the expression level in a healthy adult animal as compared to the expression level in a geriatric animal.


In an embodiment of this aspect, the animal is a dog.


It is also contemplated herein that the invention relates to methods for treating an animal suffering from disorders or disease associated with or relating to any one of more of the following biological pathways: blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport comprising administering to the animal an effective amount of a food composition of the present invention.


This invention is not limited to the particular methodology, protocols, and reagents described herein because they may vary. Further, the terminology used herein is for the purpose of describing particular embodiments only and is not intended to limit the scope of the present invention. The terms “comprise”, “comprises”, and “comprising” are to be interpreted inclusively rather than exclusively.


Unless defined otherwise, all technical and scientific terms and any acronyms used herein have the same meanings as commonly understood by one of ordinary skill in the art in the field of the invention. Many modifications and variations of the present invention are possible in light of the above teachings. It is therefore to be understood that within the scope of the appended claims the invention may be practiced otherwise than as specifically described.


All patents, patent applications, and publications mentioned herein are incorporated herein by reference in their entirety. However, where there is a conflict between a definition in the present disclosure and that of a cited reference, the present disclosure controls.


EXAMPLES

This invention can be further illustrated by the following examples, although it will be understood that these examples are included merely for purposes of illustration and are not intended to limit the scope of the invention unless otherwise specifically indicated.


Example 1

A composition formulated for senior or super senior regular or small breed canines is described in Table 2.









TABLE 2







Ingredient Composition for Canine Regular or Small Breed


Super Senior










Ingredient
% of composition














Carbohydrate
65.83



Animal Protein
14.31



Vegetable Protein
6.05



Animal/Vegetable Fat
6.60



Omega Fat
3.38



Fiber
1.42



Minerals
1.63



Vitamins
0.78










Example 2

A composition formulated for senior or super senior large breed canines is described in Table 3.









TABLE 3







Ingredient Composition for Canine Large Breed Super Senior










Ingredient
% of composition














Carbohydrate
65.15



Animal Protein
14.79



Vegetable Protein
6.45



Animal/Vegetable Fat
6.23



Omega Fat
4.12



Fiber
1.30



Minerals
0.91



Vitamins
1.05










Example 3

A composition formulated for senior or super senior felines is described in Table 4.









TABLE 4







Ingredient Composition for Feline Super Senior










Ingredient
% of composition














Carbohydrate
31.47



Animal Protein
25.57



Vegetable Protein
20.14



Animal/Vegetable Fat
13.31



Omega Fat
1.61



Fiber
4.80



Minerals
1.77



Vitamins
1.34










Example 4
Genomic Analysis of Control vs. Super Senior Pet Food

To further characterize the nutritional benefits of the super senior pet food compositions of the present invention, gene expression profiles from animals fed the compositions compared to control animals are assayed and the results are described in detail below.


Materials and Methods:
Study Design:

Blood samples are drawn from 9 Beagles according to conventional methods before and after feeding for 14 days on Super Senior K9 diet (a total of 18 samples). Each sample taken after the 14-day trial is compared to its own control.


Isolation of Lymphocytes from Canine Blood


Reagents:

4 ml canine blood, heparin or EDTA tubes, Hank's Balanced Salt Solution (Gibco 14175-095), HEPES buffer (Gibco 15630-080), Accu-Paque (Accurate Chemical & Scientific Corp AN3100).


Materials/Equipment:

Transfer pipettes (VWR 14670-147), 14 ml centrifuge tubes w/ caps, 9″ Pasteur pipettes, 1.5 ml microcentrifuge tubes (VWR 20170-038), centrifuge tube racks, microcentrifuge tube bale, waste container, Beckman Coulter Allegra 25R Centrifuge, SN AJC01J015Eppendorf Centrifuge, 5417C.


Solutions:

Hank's Balanced Salt Solution (HBSS) w/25 mM HEPES buffer solution is made by adding 12.8 ml of HEPES buffer solution to a 500 ml bottle of HBSS. Hank's Balanced Salt Solution and Accu-Paque need to be removed from the refrigerator and placed at room temperature at least 30 minutes before beginning the lymphocyte isolation. Both solutions should be place back in the refrigerator (4° C.) immediately following their use.


Procedure:


1. Measure 4 ml of HBSS w/ HEPES into the correct number of 14 ml centrifuge tubes (one tube for each 4 ml draw of blood)


2. Using a transfer pipette, transfer 4 ml blood from the Vacutainer® tubes to the 14 ml centrifuge tube containing the HBSS w/ HEPES.


3. Mix the sample well using the transfer pipette to pipette up and down for 30 seconds.


4. Insert a 9″ Pasteur pipette into each of the 14 ml centrifuge tubes. Make sure the bottom tip of the Pasteur pipette touches the bottom of the tube.


5. Using a transfer pipette, slowly add 4 ml of Accu-Paque by running the liquid down the inside of the Pasteur pipette allowing gravity to layer the Accu-Paque under the diluted blood sample.


6. Plug the top of the Pasteur pipette using your finger and gently remove the pipette.


7. Centrifuge the tubes at 800×g for 20 minutes at room temperature. For puppy blood a longer centrifugation of 45 minutes is necessary to allow for a good separation of RBC's from WBC's.


8. Using a transfer pipette, carefully remove the top layer to within 0.5 cm of the middle opaque layer and discard.


9. Using a new transfer pipette, carefully remove the middle opaque layer and transfer to a 1.5 ml microcentrifuge tube. Be careful not to transfer any of the bottom layers.


10. Centrifuge the microcentrifuge tubes at 13,200 rpm for 3.5 minutes at room temperature.


11. Carefully remove the supernatant and flash freeze the remaining pellet (lymphocytes) in liquid nitrogen. Store the final samples at −80° C.


RNA Isolation:
Reagents:

Deionized H2O, Absolute ethanol (Sigma E7023), RNA Storage Solution (Ambion 7000), RNase Zap® (Ambion 9780), Buffer RLT, Buffer RW1 and Buffer RPE (provided in the RNeasy Mini Kit).


Equipment/Materials:

RNeasy Mini Kit (Qiagen 74104), QIAshredder spin columns (Qiagen 79656), P1000 Pipetman pipette (Rainin), P200 Pipetman pipette (Rainin), 100-100 μl filtered pipette tips (USA Scientific 1126-7810), 1-200 μA filtered pipette tips (USA Scientific 1120-8810), sterile transfer pipettes (VWR 14670-147), 55 ml sterile solution basin (VWR 21007-974), 2 waste containers (one for liquid, one for tips/pipettes), 1.5 ml sterile microcentrifuge tubes (VWR 20170-038), Microcentrifuge tube rack, permanent marker, Eppendorf Microcentrifuge, model #5417C.


Procedure:

1. Loosen the pellet in the microcentrifuge tubes by thawing slightly and then flick the tube to dislodge the pellet.


2. Add the appropriate volume of Buffer RLT (in this case use 600 μl). Vortex or pipette to mix.


3. Transfer sample to a QIAshredder tube to homogenize the sample. Centrifuge for 2 minutes at 14,000 rpm. Discard spin column but keep the collection tube and its contents.


4. Add one volume (600 μl) of 70% ethanol to the homogenized lysate and mix by pipetting.


5. Apply a 600 μl aliquot of the sample to an RNeasy mini column placed in a 2 ml collection tube. Close tube gently and centrifuge for 15 sec at 14,000 rpm. Discard the flow-through. Add the second 600 μl aliquot of the cell lysate to the same spin column and repeat. Discard flow-through.


6. Reuse the collection tube from step 5. Add 700 μl Buffer RW1 to the column. Centrifuge for 15 sec at 14,000 rpm. Discard the flow-through and collection tube.


7. Transfer the column to a new 2 ml collection tube and pipette 500 μl Buffer RPE onto the column. Centrifuge for 15 sec at 14,000 rpm to wash the column. Discard the flow-through but save the collection tube for step 8.


8. Add another 500 ml Buffer RPE to the column. Centrifuge for 2 min at 14,000 rpm to dry the membrane.


9. Transfer the column to a new 1.5 ml collection tube. Pipette 10 μl of RNA Storage Solution directly onto the membrane. Centrifuge for 1 min at 14,000 rpm to elute the RNA. Add a second volume of 5 μl of RNA Storage Solution directly to the membrane and spin for an additional minute. Store the final elution of RNA at −80° C.


RNA Probe Preparation and Hybridization.
Reagent:

Ovation TM Biotin System v1.0 for probe preps.


Protocol:

User Guide (Cat#D01002, version Oct. 27, 2004, NuGEN Technologies, Inc). The experimental procedure is followed as described in the user guide. All probe preparation starts with 50 ng of total RNA.


Gene Chip Procedures:

The Genechips used for the test is the Canine Genome 2.0 Array (Affymetrix). This Genechip contains 44,000 probe sets. Detailed sequence information for each unique probe identification number is available from the manufacturer.


Gene Expression Analysis:

Normalization is performed using MAS 5 provided in GCOS Affymetrix software (version 1.2). Expression levels for the genes analyzed are indicated on the tables included in the examples below, where an upward facing arrow refers to “up regulation” or increase and a downward facing arrow indicates “down regulation” in gene expression. Similarly, in some tables, upward or downward facing arrows also indicate increases or decreases in activity of certain proteins involved in a particular pathway, and are otherwise self explanatory.


Gene List Selection:

15,411 genes are chosen for further analysis based on their “present” calls in at least 9 out of 18 samples.


Results of the gene chip analysis indicate that 1088 genes are differentially expressed between the control and Super Senior diet treated groups. The expression levels of these 1088 genes are statistically significant when grouped by ‘diet’; using a parametric test where the variances is not assumed to be equal (Welch t-test). The p-value cutoff is 0.01 with no multiple testing correction. Under those selection criteria only about 154 genes would be expected to pass the restriction by chance. The genomic data is discussed in detail below.


Results:

Effect of Nutrition on Genes Associated with Pain and Inflammation


Based on an analysis of the gene chip data, at the P<0.01 level, expression levels of 1,088 genes changed compared to control expression levels (10 were up regulated and the rest down regulated). At the P<0.001 level, data indicate that expression in 35 genes is down regulated in beagles fed the super senior food. Nine of these down regulated genes are identified as related to the inflammatory and pain response. Down regulation of these genes may be predicted to result in pain relief, cartilage protection (less damage) and reduction in inflammatory responses. The compositions disclosed herein may be part of a therapeutic regimen to treat animals suffering from pain and/or inflammatory diseases. These genes and their putative role in inflammation and pain response are provided below in Tables 5-6.









TABLE 5







Genes involved in inflammation and pain response (P < 0.001)

















% match of








probe





Best Current BLAST
sequence to
Probe Target


Genes
Also Known As
Probe
Annotation
BLAST hit
Sequence
















Phospholipase
IPLA2GAMMA,
CfaAffx.6431.
PREDICTED: Canis
100
GGAGCCATGCATTT



A2
IPLA2-2
1.S1_s_at

familiaris similar to


TATGACAGTCAAAC





intracellular

GTGGGAAAATATTC





membrane-associated

TTAAGGACAGAATG





calcium-independent

GGATCCTCGCTAAT





phospholipase A2

GATTGAAACAGCAA





gamma; transcript

GAAACCCTTCATGT





variant 3

CCTAAGGATGGAG





(LOC475880); mRNA

GTTTGCTTCTGAAT







AACCCTTCAGCGCT







AGCAATGCACGAGT







GCAAATGTCTTTGG







CCTGACGTCCCATT







AGAGTGCATTGTGT







CCCTGGGCACCGG







GCGTTATGAGAGTG







ATGTGAGAAACTCT







GTGACATCTACAAG







CTTGAAAACCAAAC







TGTCTAATGTCATT







AACAGTGCTACAGA







TACAGAAGAAGTCC







ACGTAATGCTTGAT







GGTCTTTTACCTCC







TGACACCTATTTTA







GAT





Dipeptidase 2
Putative
CfaAffx.31124.
PREDICTED: Canis
82.197
GTGCTGCAATGCAA



dipeptidase
1.S1_at

familiaris similar to


CCTGTTAGCTAACG





dipeptidase 2

TGTCCACTGTGGCA





(LOC611083); mRNA

GTTCCCACGCATCC







CTGCCCTGGAAGC







CCCACAGTGCTGAC







TCTCCATCCCTCAG







ATCACTTTGACTAC







ATCAGGGCAGTCAT







TGGATCCAAGTTCA







TTGGAATTGGTGGA







GATTATGATGGGGC







CAGACGTTTCCCTC







AGGGGCTGGAGGA







TGTGTCCACATACC







CAGTTCTGATAGAG







GAGTTGCTGAGGC







GTGGCTGGAGTAG







GGAAGAGCTCCAG







GGTGTCCTTCGAG







GAAACCTACTGCGG







GTCTTTGGACAGGT







GGAACAGGTACGG







GAGGCAAGCAAGG







GGCAAAGGCCCTT







GGAGGATGAGTTC







CCGGATGAGCAGC







TGAGCAGCTCTTGC







CGCTCCGTTCTCTC







ACGTCTGCATCAGA







CACAGTACCCTGCT







CCATACCAGAAACT







AACTGAGATTTCAC







CTGAGTGGTCCCCT







AAACAGTCATTGTC







AAAATCTCTCCCCA







TCATGGCCCCAGG







CCTCATAGTTATTG







CTGCTTGT





Thromboxane
Thromboxane A
CfaAffx.6939.
PREDICTED: Canis
100
ATCGCTGGCTATGA


synthase
synthase 1,
1.S1_s_at

familiaris similar to


GATCATCACCAACA



Thromboxane A

Thromboxane-A

CGCTCTCTTTTGCC



synthase, Platelet,

synthase (TXA

ACCTACCTCCTGGC



Cytochrome P450,

synthase) (TXS)

CACCAACCCTGACT



subfamily V,

(LOC482771); mRNA

GCCAAGAGAAGCTT



CYP5, CYP5A1,



CTGGCAGAGGTGG



Thromboxane



ACAGCTTTAAGGAG



synthatase, TXA



AAATATACGGCCCT



synthase, TXS



TGACTACTGCAGCC







TCCAGGAAGGCCT







GCCCTACCTGGACA







TGGTGATTGCGGA







GACCTTGAGGATCT







ACCCCCCGGCTTTC







AGGTTCACACGGG







AGGCGGCGCGGGA







CTGCGAGGTGCGG







GGACAGCGCATCC







CCGCGGGCGCCGT







GGTGGAGGTGGCC







GTGGGCGCCCTGC







ACCGTGACCCTGA







GTACTGGCCACAAC







CGGAGACCTTCAAC







CCCGAGAGGTTCAA







GGCCGAGGCGCAG







CGACGACAGCAAC







CCTTCACCTACCTG







CCGTTCGGCGCGG







GCCCCCGGAGCTG







CCTCGGGGTGCGG







CTGGGGCTGCTGG







AGGTCAAGCTGAC







GCTGCTGCAGGTC







CTGCACCAGTTCCG







GTTCGAGGCCTGC







CCGGAGACGCAGG







TACCACTGCAGCTA







GACTCCAAATCTGC







CCTAGGTCCAAAGA







ATGGCATCTACATC







AAGATTGTCTCCCG







CT





Ubiquitin
Ubiquitin protein
CfaAffx.275.1.
PREDICTED: Pan
97.19626
GATTTGGCCCGTGA


conjugating
ligase, Ubiquitin
S1_s_at

troglodytes


CCCTCCAGCACAAT


enzyme
carrier protein,

LOC461941

GTTCTGCAGGTCCT


E2D 3
E2(17)KB 3,

(LOC461941); mRNA

GTTTGGGATGATAT



Ubiquitin



GTTTCATTGGCAAG



conjugating



CCACAATTATAGGA



enzyme E2-17 kDa



CCTAATGACAGCCC



3, UBC4/5,



ATATCAAGG



UBCH5C





NEDD8
Neural precursor
Cfa.12556.1.A
PREDICTED: Canis
99.12473
GGAATGGGCTACTC


ultimate
cell expressed,
1_s_at

familiaris similar to


TACTCATGCAGNCA


buster-1
developmentally

NEDD8 ultimate

AGCAGGNCCTGCA



down regulated 8,

buster-1 (NY-REN-18

TCAGGCCAGTGGG



Ubiquitin like

antigen)

AACCTGGACGAAG



protein NEDD8

(LOC475542); mRNA

CCCTGAAGATTCTT







CTCAGCAATCCTCA







GATGTGGTGGTTAA







ATGATTCAGATCCT







GAAACGANCAACCA







GCAAGAAAGTCCTT







CCCAGGAAAACATT







GACCAACTGGTGTA







CATGGGCTTCGAC







GCTGTGGTGGCTG







ATGCTGCCTTGAGA







GTGTTCAGGGGAAA







CGTGCAGCTGGCA







GCTCAGNCCCTCG







CCCACAACGGAGG







AACTCTTCCTCCTG







ACCTGCAGCTCTTG







GTGGAAGACTCTTC







ATCAACGCCATCCA







CGTCCCCTTCCGAC







TCCGCAGGTACCTC







TAGTGCCTCAACAG







ATGAAGATATGGAA







ACCGAAGCTGTCAA







TGAAATACTGGAAG







ATATTCCAGAACAT







GAAGAAGATTATCT







TGACTCAACACTGG







AAG





Mitogen-
p38, Mitogen
CfaAffx.2947.

Homo sapiens

97.84946
GAGATGGAGTCCT


activated
activated protein
1.S1_at
mitogen-activated

GAGCACCTGGTTTC


protein
kinase 14,

protein kinase 14;

TGTTTTGTTGATCC


kinase 14
Cytokine

transcript variant 2;

CACTTCACTGTGAG


(p38)
suppressive

mRNA (cDNA clone

GGGAAGGCCTTTTC



antiinflammatory

MGC: 34610

ATGGGAACTCTCCA



drug binding

IMAGE: 5181064);

AATATCATTC



protein 1, CSBP1,

complete cds



CSAID binding



protein 1, Stress



activated protein



kinase 2A,



SAPK2A, p38



MAP kinase, p38



alpha, RK, MXI2,



Cytokine



suppressive



antiinflammatory



drug binding



protein 2, CSBP2,



CSAID binding



protein 2





Matrix
MMP 19
Cfa.4573.1.A1_at

Homo sapiens cDNA

48.93048
GTAGTTGATTCCTG


metalloproteinase


FLJ38021 fis; clone

GTTCGCCTTTCCTC


19


CTONG2012847

TTGGGTCCCATAGG


(MMP-19)




TTCGAATCCCCTTC







TACCTCAGTCGGGA







GTACTGTCCTCCAT







GGTGCTTCCCTTCC







TCTCCTTAATGTGG







GGAAGACCATGGG







GCAATGCATGGCG







CAGGACCTGCCTC







CCCCAAAAGCAGTC







TACTTGCTCCACGG







AGAGAGAACTGGG







TCCACGTGCCAGA







GTCTTGCCCTTTGG







CCCAGAGTAGCCT







GGTCTTCATGGCTG







TATGGGAGACAAGT







GCCTTCTCTGCTTC







TTGTTGTAGGTGAT







GCTAATCTCCTTAA







CCAAACCTTTGTCC







CAGCCGCTAATCTG







TTCTAACTCTCCCT







CCTCNTGATTCTCC







TGCTCAAAGTCTGT







TC





Tissue
TIMP-1
Cfa.3680.1.S1_s_at

Canis familiaris TIMP

99.4
AGATGTTCAAGGGT


Inhibitor of


metallopeptidase

TTCAGCGCCTTGGG


metalloproteinases


inhibitor 1 (TIMP1);

GAATGCCTCGGACA


(TIMP-1)


mRNA

TCCGCTTCGTCGAC







ACCCCCGCCCTGG







AGAGCGTCTGCGG







ATACTTGCACAGGT







CCCAGAACCGCAG







CGAGGAGTTTCTGG







TCGCCGGAAACCT







GCGGGACGGACAC







TTGCAGATCAACAC







CTGCAGTTTCGTGG







CCCCGTGGAGCAG







CCTGAGTACCGCTC







AGCGCCGGGGCTT







CACCAAGACCTATG







CTGCTGGCTGTGA







GGGGTGCACAGTG







TTTACCTGTTCATC







CATCCCCTGCAAAC







TGCAGAGTGACACT







CACTGCTTGTGGAC







GGACCAGTTCCTCA







CAGGCTCTGACAAG







GGTTTCCAGAGCC







GCCACCTGGCCTG







CCTGCCAAGAGAG







CCAGGGATATGCAC







CTGGCAGTCCCTG







CGGCCCCGGATGG







CCTAAATCCTACTC







CCCGTGGAAGCCA







AAGCCTGCACAGTG







TTCACCCCACTTCC







CACTCCTGTCTTTC







TTTATCCAAAA





Fatty acid
Oleamide
CfaAffx.7308.
PREDICTED: Canis
63.33333
GAAGTGGAGTAGG


amide
hydrolase
1.S1_x_at

familiaris similar to


TGCCGCTGTTGCTG


hydrolase
Anandamide

Ubiquinol-cytochrome

CTGGTGTTGAATTC


(FAAH)
amidohydrolase

c reductase complex

AGAACTGTAGCGG



FAAH

11 kDa protein;

GACATGGGGCTGG





mitochondrial

AGGACGAGCAAAA





precursor

GATGCTGACCGGG





(Mitochondrial hinge

TCCGGAGATCCCAA





protein) (Cytochrome

GGAGGATCCCCTAA





C1; nonheme 11 kDa

CAACAGTGAGAGA





protein) (Complex III

GCAATGCGAGCAG





subunit VIII);

CTGGAGAAATGTGT





transcript variant 2

AAAGGCTCGGGAG





(LOC608530); mRNA

CGGCTAGAGCTCT







GTGACCAGCGTGTA







TCCTCCAGGTCACA







GACAGAGGAGGAT







TGCACAGAGGAGC







TCTTTGACTTCCTG







CATGCAAGGGACC







ACTGTGTGGCCCAC







AAACTCTTTAACAG







CTTG
















TABLE 6







Summary of down-regulated enzyme roles involved


in the eicosanoid pathway (inflammatory response)











Gene Expression





Compared to


Gene
Control
Results in
Role





Phospholipase

↓ in arachidonic
↓ in 2-series inflammatory


A2

release from
response




phospholipids


Thromboxane

↓ Thromboxane A2
↓ platelet aggregation,


synthase


vasoconstriction, lymphocyte





proliferation and





bronchoconstriction




↓ Thromboxane B2
↓ vasoconstriction


Dipeptidase 2

↓ Leukotriene E4
↓ component of slow-reactive





substance of anaphylaxis,





microvascular vasoconstrictor





and bronchoconstriction


Ubiquitin

↓ ubiquination or
↓ MMP Production


conjugating

activation of TAK1,


enzyme E2D 3

IRAK and TRAF


(and NEDD8


ultimate


buster-1)


Mitogen

↓ in c-Jun promotor
↓ MMP Production


activated


protein kinase


14 (p38)


MMP-19

↓ MMP-19
↓ in T-cell derived MMP-19





which has been implicated in





rheumatoid arthritis


TIMP-1

↓ TIMP-1
Deactivates MMP's





concentration is directly





related to MMP concentration


Fatty acid

↑ anandmide
↓ pain response


amide


hydrolase









Effect of Nutrition on Genes Involved in Heart Health and Blood Coagulation

At the P<0.001 and P<0.01 level, 12 genes are identified to be related to heart health through regulation of the eicosanoid pathway and blood coagulation pathway. The genes are responsible for blood coagulation through platelet activation and aggregation. The down regulation of these genes through nutrition can prevent inappropriate blood clotting which may result in heart or brain related disorders. The compositions of the present invention may be part of a therapeutic regimen to treat animals suffering from disorders or diseases of the blood, heart or brain. These genes and their putative role in vivo are described in Tables 7 and 8 below.









TABLE 7







Genes involved in heart health and blood coagulation

















% match of







Best current
probe





BLAST
sequence to


Gene
Probe
P-value
annotation
BLAST hit
Probe Target Seq.
















Glycoprotein
Cfa.3503.1.
<0.01

Canis familiaris

98.57143
TGTGGGTCCGAGCTAACAGCTA



Ib
S1_at

glycoprotein lb

CGTGGGGCCTCTGATGGCAGG





mRNA; complete

ACGGCGGCCCTCTGCCCTGAG





cds

CCTGGGTCGTGGGCAGGACCT







GCTAGGTACGGTGGGCGTTAG







GTACTCCAGCCACAGCCTCTGA







GGCGACGGTGGGCAGTTTGGG







GACCTTGAGAGGCTGTGATGG







GCCCTCCTATCAGGATCTTGCT







GGGGGTGGGTGGGCAGGGAG







CACAGGATTGGGGGGAGGCCT







TAAGCACCTTTTCTGGGTCAGA







AGCCTCCTCTCCGCATTGCATG







TGCAACCTCAGTGAAGCAGCAT







GGGCAGGGGAGCCGGACGGG







CCACCCAACAGAGCTCCTTATG







CTGCAGGAGGGGTTCACAGAC







CACTCGGACATCACCATCACCT







TGGGGGGGGTGCTTGAGGGAA







AAGCAAATTGAACAGAGCGTGA







TTCTCACGTGCAGGTACCTAAG







GGAACTGGGGAAGAGATGCAC







CAAGACGAGAGCCCTCGTCATC







CCTGGGGAGCCCAAGCCTAGG







GGTTTTCTTCCTCTTCCCGTTTA







GCATTTTCCACCATCGTATGTTAC





Platelet
CfaAffx.4809.
<0.01
PREDICTED:
50
AGTTTTGACCAATTCGCTCTGT


glycoprotein
1.S1_at


Canis familiaris


ACAAGGAGGGGGACACTGAGC


VI


similar to

CCCACAAGCAATCTGCAGAACA





glycoprotein VI

GTACTGGGCCAATTTCCCCATC





(platelet)

ACCGCAGTGACTGTTGCCCACA





(LOC484303);

GTGGGATCTACCGATGCTATAG





mRNA

CTTTTCCAGCAAGTTCCCGTAC







CTGTGGTCAGCCCCCAGCGAC







CCCCTGGAGCTTGTGGTAACAG







GTGAGGGAGATGCAGTCCAAG







CCTTTCTTCTTCAGCTCTTGCAT







ACTCTGGTGGAAGTTCCAGGG







GAGGGGCCAACAGTGCCTTCT







AGGACTATCACTGTCTCTCCAA







AGGGGTCAGACTCTCCAACTG







GTCTTGCTCACCAGCACTACAC







CAAGGGCAATCTGGTCCGGATA







TGCCTTGGAGCTGTGATTCTAA







TACTCCTGGTGGGAATTCTGGC







AGAAGATTGGCACAGCAGAAAG







AAACCCCTGTTGCTCCGGGTCA







GAGCTGTCCACAGGCCACTCC







CACCCCTCCCACAGACCCAGAA







ACCACACAGTCATCAGGATGGG







GGTCGACCAGATGGCCATAAC







CAT





Platelet
CfaAffx.7430.
<0.01
PREDICTED:
100
TCTGGGCTGCCACGGAGGCCA


glycoprotein
1.S1_at


Canis familiaris


CCAACGACTGCCCCGCAGAGT


IX precursor


similar to Platelet

GCACCTGCCAGACCCTGGAGA





glycoprotein IX

CCATGGGGCTGTGGGTGGACT





precursor (GPIX)

GCAGGGGGCGGGGACTCAAGG





(CD42A)

CCCTGCCCGCCCTGCCGGTCC





(LOC609630);

ACACCCGCCACCTCCTGCTGG





mRNA

CCAATAACAGCCTCCGCTCCGT







GCCCCCTGGTGCCTTCGACCA







CCTGCCTGGGCTGCAGATCCT







CGACGTGATGCACAACCCCTG







GCACTGTGACTGCAGCCTCACC







TACCTGCGTCTCTGGCTGGAG







GACCACACGCCCGAGGCCTTG







CTGCAGGTCCGCTGTGCCAGC







CCCGCGCTGGCCACCACCCGG







CCGCTGGGCTGGCTGACGGGC







TACGAGCTGGGCAGCTGCGGC







TGGCAGCTACAGGCACCCTGG







ACCTA





Coagulation
CfaAffx.14964.
<0.01
PREDICTED:
99.6008
ATCTCTCAGGCAACATCGTCTT


factor XIII A
1.S1_s_at


Canis familiaris


CTACACCGGGGTCTCCAAGAC


chain


similar to

GGAATTCAAGAAGGAGACATTT


precursor


Coagulation

GAAGTGACACTGGAGCCCTTGT





factor XIII A

CTTTCAAGAGAGAGGAGGTGCT





chain precursor

GATCAGAGCGGGCGAGTACAT





(Coagulation

GGGCCAGCTGCTAGAGCAAGC





factor XIIIa)

ATACCTGCACTTCTTTGTCACA





(Protein-

GCGCGTGTCAATGAGTCCAAG





glutamine

GATATTCTGGCCAAGCAGAAGT





gamma-

CCACCGTGCTGACGATCCCCC





glutamyltransferase

AGCTCATCATCAAGGTCCGTGG





A chain)

CGCCAAGATGGTTGGTTCTGAC





(Transglutaminase

ATGGTGGTGACAGTTGAGTTCA





A chain);

CCAATCCCCTGAAAGAAACTCT





transcript variant

GCGGAATGTGTGGATACACCTG





1 (LOC478711);

GATGGTCCTGGAGTGATAAAGC





mRNA

CAATGAGGAAGATGTTCCGTGA







AATCCAGCCCANTGCCACCATA







CAATGGGAAGAAGTGTGTCGAC







CCTGGGTGTCTGGCCCTCGGA







AGCTGATAGCCAGCATGACGA







GTGACTCCCTGAGACACGTGTA







TG





Thromboxane
CfaAffx.6939.
<0.001
PREDICTED:
100
ATCGCTGGCTATGAGATCATCA


synthase
1.S1_s_at


Canis familiaris


CCAACACGCTCTCTTTTGCCAC





similar to

CTACCTCCTGGCCACCAACCCT





Thromboxane-A

GACTGCCAAGAGAAGCTTCTGG





synthase (TXA

CAGAGGTGGACAGCTTTAAGGA





synthase) (TXS)

GAAATATACGGCCCTTGACTAC





(LOC482771);

TGCAGCCTCCAGGAAGGCCTG





mRNA

CCCTACCTGGACATGGTGATTG







CGGAGACCTTGAGGATCTACCC







CCCGGCTTTCAGGTTCACACGG







GAGGCGGCGCGGGACTGCGA







GGTGCGGGGACAGCGCATCCC







CGCGGGCGCCGTGGTGGAGGT







GGCCGTGGGCGCCCTGCACCG







TGACCCTGAGTACTGGCCACAA







CCGGAGACCTTCAACCCCGAG







AGGTTCAAGGCCGAGGCGCAG







CGACGACAGCAACCCTTCACCT







ACCTGCCGTTCGGCGCGGGCC







CCCGGAGCTGCCTCGGGGTGC







GGCTGGGGCTGCTGGAGGTCA







AGCTGACGCTGCTGCAGGTCC







TGCACCAGTTCCGGTTCGAGG







CCTGCCCGGAGACGCAGGTAC







CACTGCAGCTAGACTCCAAATC







TGCCCTAGGTCCAAAGAATGGC







ATCTACATCAAGATTGTCTCCC







GCT





Dystrobrevin
CfaAffx.15541.
<0.01
PREDICTED:
99.65986
GGCAACATGTCGTCCATGGAG


binding
1.S1_s_at


Canis familiaris


GTCAACATCGACATGCTGGAGC


protein 1


similar to

AGATGGACCTGATGGACATCTC


isoform a


dystrobrevin

TGACCAGGAGGCCCTGGACGT





binding protein 1

CTTCCTGAACTCCGGCGCTGAA





isoform a

GACAACACGGTGCCGTCTCCG





(LOC610315);

GTCTCAGGGCCTGGCTCGGGG





mRNA

GACAGTCGGCAGGAAATCACG







CTCCGGGTTCCAGATCCCGCC







GAATCGCAAGCTGAGCCTCCTC







CCTCGCCGTGTGCCTGTCCTGA







GCTGGCCGCCCCGGCCCCCGG







CGACGGTGAGGCCCCCGTGGT







CCAGTCTGACGAGGAG





Integrin beta-7
Cfa.11961.1.
<0.01
PREDICTED:
99.0909
ATTACAACGTGACTCTGGCTTT


precursor
A1_s_at


Canis familiaris


GGTCCCTGTCCTGGATGACGG





similar to Integrin

CTGGTGCAAAGAGAGGACCCT





beta-7 precursor

AGACNAACCAGCTGCTGTTCTT





(LOC477598);

CCTGGTGGAGGAGGAANCCGG





mRNA

AGGCATGGTTGTGTTGACAGTG







AGACCCCAAGAGAGAGGCGCG







GATCACACCCAGGCCATCGTG







CTGGGCTGTGTAGGGGGCATC







GTGGCAGTGGGGCTGGGGCTG







GTCCTGGCTTACCGGCTCTCTG







TGGAAATCTACGNCCGCCGAG







AATTTAGCCGCTTTGAGAAGGA







GCAGAAGCACCTCAACTGGAA







GCAGGAAAACAATCCTCTCTAC







AGAAGCGCC





integrin-linked
Cfa.465.1.S1_s_at
<0.01
PREDICTED:
100
TGGGCGCATGTATGCACCTGC


kinase



Canis familiaris


CTGGGTGGCCCCTGAAGCTCT





similar to integrin

GCAGAAGAAGCCTGAAGATACA





linked kinase;

AACAGACGCTCAGCAGATATGT





transcript variant

GGAGTTTTGCAGTGCTTCTGTG





1 (LOC476836);

GGAACTGGTGACGAGGGAGGT





mRNA

ACCCTTTGCTGACCTCTCCAAC







ATGGAGATTGGAATGAAGGTGG







CACTGGAAGGCCTTCGGCCTA







CTATCCCACCAGGCATTTCCCC







CCATGTGTGTAAGCTCATGAAG







ATCTGCATGAATGAAGACCCTG







CTAAGCGGCCCAAGTTTGACAT







GATTGTGCCTATCCTGGAGAAG







ATGCAGGACAAGTAGAGCTGG







AAAGCCCTTGCCTAAACTCCAG







AGGTGTCAGGACACGGTTAGG







GGAGTGTGTCTCCCCAAAGCA







GCAGGC





Thrombospondin 1
Cfa.21204.1.
<0.01
PREDICTED:
54.83871
ATACGAATGCAGAGATTCCTAA



S1_at


Canis familiaris


TCAAACTGTTGATCAAAAGACT





similar to

GATCCTAACCAATGCTGGTGTT





thrombospondin

GCACCTTCTGGAACCACGGGC





1 precursor

TTAAGAAAACCCCCAGGATCAC





(LOC487486);

TCCTCCCTGCCTTTTCTCTGCTT





mRNA

GCATATCATTGTGGACACCTAG







AATACGGGACTTGCCTCGAGAC







CATGCNNNNNTCCAAATCAGAC







TNNNNNNGTAGCCTCTGAACGC







GAAGAGAATCTTCCAAGAGCAT







GAACAG





Thrombospondin
CfaAffx.18675.
<0.01
PREDICTED:
100
GAAGCCCTTGATGGATACTGTG


repeat
1.S1_s_at


Canis familiaris


AACGGGAACAGGCTATAAAGAC


containing 1


similar to

CCACCACCACTCCTGTTGCCAC





extracellular

CACCCTCCTAGCCCTGCCCGC





matrix protein 1

GATGAGTGCTTTGCCCGTCAGG





isoform 1

CGCCATACCCCAACTATGACCG





precursor

GGACATCCTGACCCTTGATTTC





(LOC608791);

AGCCAAGTTACCCCCAACCTCA





mRNA

TGCAACATCTCTGTGGAAATGG







AAGACTTCTCACCAAGCATAAA







CAGATTCCTGGGCTGATCCGGA







ACATGACTGCCCACTGCTGTGA







CCTGCCATTTCCAGAGCAGGCC







TGCTGTGCTGAGGAGGAGAAAT







CGGCCTTCATTGCAGACTTGTG







TGGTTCCCGACGTAACTTCTGG







CGAGACTCTGCCCTCTGCTGTA







ACCTGAATCCTGGAGATGAACA







GACCAACTGCTTCAACACTTAT







TATCTGAGGAATGTGGCTCTAG







TGGCTGGAGACAAT





Thrombospondin
CfaAffx.16694.
<0.01
PREDICTED:
98.13084
TGGTTGTAGCTCCTCACTTGTC


type 1
1.S1_at


Canis familiaris


CAAGACCGAAGCAGCAACCAAA


motif, 17


similar to lines

CTGAACTTAGCCTTTGGGCTGC





homolog 1

TCTTGGTAGTCACAGAAATGCC





isoform 1

CACGCTTCAGTCCCCTGGGCTT





(LOC607902);

CCAATGCTTCTGGACCTCTGAA





mRNA

CCAGCCTGTGATGTCCAAGGAA







CCCCACGTCACGCTCCAGGCT







GCTGCTGGTCTGTCTCCCCCAC







AAGCTTCTCAAAGTCTGGTAGA







TTATGACAGCTCTGATGATTCT







GAAGTAGAAGTCACAGACCAGC







ACTCAACAAACAGTAAACAAAC







ATCTTTACAGCAAGAAGCAAAG







AAGAAATTTCAGGACACAGTTA







GAACAGGTCCAGATGAAAAAGA







ACTTAGCATGGAGCCTCAATCA







AGGCCTCTGGTTCCAGAACAAT







CTAATATTAATATTCCCTTCTCT







GTTGACTGTGACATCTCCAAAG







TAGGAATATCTTACAGGACACT







GAAGTGCTTTCAGGAGCTACAG







GGTGCCATTTACCGTTTGCAGA







AAAAAAATCTTTTCCCCTATAAT







GCCACA





Angio-
Cfa.8616.1.
<0.001

Canis familiaris

64.77273
GCGGACTGTGTTCCAACCCCTT


associated
A1_s_at

angio-associated

CAGCCGACTTGCCCCCTCCGT


migratory cell


migratory cell

CCCTTCTCTTAAGAGACCCATC


protein


protein (AAMP)

CCTTGGCCCCCCACCCCACCC


(AAMP)


gene; complete

TCACCCAGACCTGCGGGTCCC





cds

TCAGAGGGGGGTCAGGCCTCT







TTCTCTTTCACCTTCATTTGCTG







GCGTGAGCTGCGGGGGTGTGT







GTTTGTATGTGGGGAGTAGGTG







TTTGAGGTTCCCGTTCTTTCCC







TTCCCAAGTCTCTGGGGGTGGA







AAGGAGGAAGAGATATTAGTTA







CAGA
















TABLE 8







Summary of down regulated enzyme roles


involved in heart health and blood coagulation










Gene




Expression



compared


Gene
to Control
Role





Glycoprotein Ib

GP-Ib, a surface membrane




protein of platelets,




participates in the formation




of platelet plugs by binding




to the A1 domain of von




Willebrand factor, which is




already bound to the




subendothelium.


Platelet glycoprotein VI

Collagen receptor belonging




to the immunoglobulin-like




protein family that is




essential for platelet




interactions with collagen


Platelet glycoprotein IX

The GPIb-V-IX complex


precursor

functions as the von




Willebrand factor receptor




and mediates von




Willebrand factor-




dependent platelet adhesion




to blood vessels. The




adhesion of platelets to




injured vascular surfaces in




the arterial circulation is a




critical initiating event in




hemostasis


Coagulation factor XIII A

Factor XIII is activated by


chain precursor

thrombin and calcium ion to




a transglutaminase that




catalyzes




the formation of gamma-




glutamyl-epsilon-lysine




cross-links between fibrin




chains, thus




stabilizing the fibrin clot.


Thromboxane synthase

↓ platelet aggregation,




vasoconstriction,




lymphocyte proliferation




and bronchoconstriction


Angio-associated

contains a heparin-binding


migratory cell protein

domain (dissociation


(AAMP)

constant, 14 pmol) and




mediates heparin-sensitive




cell adhesion


Dystrobrevin binding

Plays a role in the


protein 1 isoform a

biogenesis of lysosome-




related organelles such as




platelet dense




granule and melanosomes


Thrombospondin 1

Adhesive glycoprotein that




mediates cell-to-cell and




cell-to-matrix interactions.




Can bind to fibrinogen,




fibronectin, laminin, type V




collagen and integrins




alpha-V/beta-1, alpha-




V/beta-3 and alpha-IIb/beta-




3.


Thrombospondin type 1

Metalloprotease activity


motif, 17


Thrombospondin repeat



containing 1


Integrin beta-7 precursor

Integrin alpha-4/beta-7




(Peyer's patches-specific




homing receptor LPAM-1)




is expected to play a role in




adhesive interactions of




leukocytes. It is a receptor




for fibronectin and




recognizes one or more




domains within the




alternatively spliced CS-1




region of fibronectin.




Integrin alpha-4/beta-7 is




also a receptor for




MADCAM1 and VCAM1.




It recognizes the sequence




L-D-T in MADCAM1.




Integrin alpha-E/beta-7




(HML-1) is a receptor for




E-cadherin.


Integrin linked kinase

Receptor-proximal protein




kinase regulating integrin-




mediated signal




transduction. May act as a




mediator of inside-out




integrin signaling. Focal




adhesion protein part of the




complex ILK-PINCH. This




complex is considered to be




one of the convergence




points of integrin- and




growth factor-signaling




pathway. Could be




implicated in mediating cell




architecture, adhesion to




integrin substrates and




anchorage-dependent




growth in epithelial cells.




Phosphorylates beta-1 and




beta-3 integrin subunit on




serine and threonine




residues, but also AKT1




and GSK3B.










Effect of Nutrition on Genes Involved with Muscle and Bone Regulation


Ten down regulated genes are identified as related to body composition through regulation of bone and muscle. The genes spare muscle and bone deterioration by reducing nitric oxide production and glucocorticoid degradation of muscle. Down regulation of these genes results in a decrease in nitric oxide production and glucocorticoid response. The compositions disclosed herein may be part of a therapeutic regimen to treat animals suffering from diseases or disorders associated with or relating to muscle or bone. These genes and their putative role in muscle and bone regulation are detailed in Tables 9 and 10 below.









TABLE 9







Genes involved in muscle and bone regulation

















% match of








probe





Best current BLAST
sequence to


Gene
Probe
P-value
annotation
BLAST hit
Probe Target Sequence
















Capping
Cfa.1044.1.
0.001
PREDICTED: Canis
44.87179
AGGTCCCGTAACACCGGCAT



Protein
S1_at


familiaris similar to F-


CGCGACCGCACAGCGCCAT





actin capping protein

CTCCCCAGAATAAAGCCCAG





beta subunit

TAAACACCCCTGNNNNNNAN





(LOC478209); mRNA

NNNNANNNNNCACCACGTTT







TGCTATCAGAACTCTCCTTGT







TTCCAGAGCCCGTGTGCTTT







TGTTTGCCCCAGCCCC





Calmodulin
Cfa.4168.1.
0.01
PREDICTED: Canis
52.54237
CCACCCATGGTGACGATGAC



S1_at


familiaris similar to


ACACATCCTGGTGGCATGCG





calmodulin 1;

TGTGTTGGTTTAGCGTTGTCT





transcript variant 3

GCGTTGTACTAGAGCGAAAA





(LOC480416); mRNA

TGGGTGTCAGGCTTGTCACC







ATTCACACAGAAATTTAAAAA







AAAAAAAAAAANNNNGANAA







AAAACCTTTACCAAGGGAGC







ATCTTTGGACTCTCTGTTTTT







AAAACCTCCTGAACCATGAC







TTGGAGCCAGCAGATTAGGC







TGTGGCTGTGGACTTCAGCA







CAACCATCAACATTGCTGATC







AAGAAATTACAATATACGTCC







ATTCCAAGTT





Dynein
Cfa.4942.1.
0.001
PREDICTED: Canis
99.6016
ATACCTCAGAGGTCTCGTAG



A1_s_at


familiaris similar to


CTCGTGCCCTTGCCATCCAG





dynein; cytoplasmic;

AGCTGGGTGGNAGAGAGCT





heavy polypeptide 2;

GAGAAGCAGGCTCTTTTCTC





transcript variant 2

TGATACACTCGACCTGTCAG





(LOC479461); mRNA

AACTCTTCCACCCAGACACA







TTTCTCAATGCTCTTCGCCAG







GAAACAGCAAGGGTGATGGG







CTGCTCTGTGGATAGCCTTA







AGTTTGTAGCTTCGTGGAAA







GGTCGGCTGCAAGAAGCAAA







GCTGCAGATCAAGATGGGCG







GCTTGCTTCTGGAAGGCTGC







AGTTTTGACGGGAGCCGGCT







CTCTGAAAACCACCACGATT







CTCCAAGTGTGTCACCAGTT







CTCCCTTGCTGTGTTGGCTG







GATTCCCCAGGGTGCATATG







GTCCCTATTCTCCTGACGAG







TGCATATCTCTGCCCGTGTA







CACGAGCGCTGAGAGGGAT







CGTGTGGTAGCCAACATCGA







CGTCCCGTGTGGGGGCANC







CAAGACCAGTGGATTCAGTG







TGGAGCCGCTCTGTTTCTAA







AAAA





Dynactin
Cfa.1807.1.
0.01
PREDICTED: Canis
100
AGGACGACAAGGCTCAGGAC



S1_at


familiaris similar to


GCAAAGTGTGAAACTGCCTT





dynactin 3 isoform 2;

TGTAACAGGGCAGAAGCAGC





transcript variant 1

TCTGTATTGGATTCACAACCT





(LOC474750); mRNA

ACCTATCTGCATTCAGGTGG







GGCTCGGAGGTCAGAGGTCT







GGCTACTTGAGGTTTGCTGT







TTGCAC





Kinesin
Cfa.10496.
0.01
PREDICTED: Canis
99.73046
AGCCACAGCATTTCCTTTTAA



1.S1_s_at


familiaris similar to


CTTGGTTCAATTTTTGTAGCA





Kinesin-like protein

AGACTGAGCAGTTCTAAATC





KIF2 (Kinesin-2)

CTTTGCGTGCATGCATACCT





(HK2); transcript

CATCAGTGNACTGTACATAC





variant 5

CTTGCCCTCTCCCAGAGACA





(LOC478071); mRNA

GCTGTGCTCACCTCTTCCTG







CTTTGTGCCTTGACTAAGGC







TTTTGACCCTAAATTTCTGAA







GCACAGCCAAGATAAAGTAC







ATTCCTTAATTGTCAGTGTAA







ATTACCTTTATTGTGTGTACA







TTTTTACTGTACTTGAGACAT







TTTTTGTGTGTGACTAGTTAA







TTTTGCAGGATGTGCCATATC







ATTGAATGGAACTAAAGTCTG







TGACAGTGGACATAGCTGCT







GGACCATTCCATCTTACATGTA





Heat
CfaAffx.11022.
0.01
PREDICTED: Canis
100
GGTGCTACTGTTTGAAACAG


Shock
1.S1_s_at


familiaris similar to


CTCTACTCTCCTCCGGCTTCT


Protein 1


Heat shock protein

CACTGGAGGATCCCCAGACT


(HSP90)


HSP 90-beta (HSP

CACTCCAACCGCATTTACCG





84) (Tumor specific

CATGATAAAGCTAGGCCTGG





transplantation 84 kDa

GCATCGATGAAGATGAAGTG





antigen) (TSTA)

GCAGCGGAGGAACCCAGTG





(LOC611252); mRNA

CTGCTGTTCCTGATGAGATC







CCTCCACTTGAGGGTGATGA







GGATGCCTCTCGCATGGAAG







AAGTC





PPlase
CfaAffx.1740.
0.01
PREDICTED: Canis
100
GACATCACCAGTGGAGACGG



1.S1_at


familiaris similar to


CACCGGCGGTATAAGCATTT





Peptidyl-prolyl cis-

ATGGTGAGACGTTTCCAGAT





trans isomerase C

GAAAACTTCAAACTGAAGCAT





(PPlase) (Rotamase)

TATGGCATTGGTTGGGTCAG





(Cyclophilin C)

CATGGCCAACGCTGGGCCTG





(LOC481480); mRNA

ACACCAACGGCTCTCAGTTC







TTTATCACCTTGACCAAGCCC







ACTTGGTTGGATGGCAAACA







TGTGGTATTTGGAAAAGTCCT







TGATGGAATGACTGTGGTCC







ACTCCATAGAACTTCAGGCA







ACCGATGGGCACG





Calcinuerin
Cfa.19761.
0.001
PREDICTED: Canis
98.83382
GAATTAACAATCTGCTTGAGC



1.S1_at


familiaris similar to


CCCAAAACACTACTTATGCAC





protein phosphatase

TTCACTTGCCAAAAGATTTGN





3 (formerly 2B);

GCAAGGTTTTGTACCCTGGT





catalytic subunit; beta

AAATGATGCCAAAGTTTGTTT





isoform (calcineurin A

TCTGTGGTGTTTGTCAAATGT





beta); transcript

TCTATGTATAATTGACTGTCT





variant 5

GTAACATGCTGTTTNCTTCCT





(LOC479248); mRNA

CTGCAGATGTAGCTGCTTTC







CTAAATCTGTCTGTCTTTCTT







TAGGTTAGCTGTATGTCTGTA







AAAGTATGTTAAATTAAATTA







CTCTATCAGACGCTTGTCTGT







CTTTTGATGTAGAAGCAACTT







TGTAGCACCTTGTTTTGAGGT







NNGCTGCATTTGTTGCTGTA







CTTTGTGCAT





Protein
CfaAffx.408.
0.01
PREDICTED: Canis
99.64664
TTCAGTTCCTGTCTCATGGC


kinase C
1.S1_s_at


familiaris similar to


CGCTCCCGGGACCATGCCAT





myeloid-associated

CGCCGCCACTGCCTTCTCCT





differentiation marker

GCATCGCTTGTGTGGCTTAT





(LOC611521); mRNA

GCCACCGAAGTGGCCTGGA







CCCGGGCCCGTCCCGGAGA







GATCACCGGCTACATGGCCA







NTGTGCCGGGCCTGCTCAAG







GTGCTGGAGACCTTTGTGGC







CTGCATCATCTTCGCCTTCAT







CAGCAACCCCTCCCTGTACC







AGCACCAGCCGGCCCTGGA







GTGGTGTGTGGCCGTCTACT







CCATCTGTTTCATCCTGGCG







GCTGTGGCCATCCTACTGAA







CCTGGGGGACTGCACCAACA







TGCTGCCCATCTCCTTCCCC







AGTTTCCTGTCGGGCCTGGC







CCTGCTCTCCGTCCTGCTGT







ATGCCACGGCTCTGGNTCTC







TGGCCGCTCTACCAGTTCAA







CGAGAAGTATGGTGGCCAGC







CCCGTCGGTCGAGGGATGTT







AGCTGCGCCGACAGGCACA







CCTACTACGTGTGTACCTGG







GACCGCCGCCTGGCTGTGG







CCATCCTGACAGCCATCAAC







CTGCTGGCTTACGTGGCTGA







CCTGGTGTAC





Protein
Cfa.15485.
0.01
PREDICTED: Canis
100
GGAGCAGTCAGAACTAAGAC


Kinase C
1.A1_s_at


familiaris similar to


ATGGTCCGTTTTACTATATGA


Binding


protein kinase C

AGCAGCCACTCACCACAGAC


Protein


binding protein 1

CCTGTTGATGTTGTACCGCA





isoform b; transcript

GGATGGACGGAA





variant 11





(LOC477252); mRNA
















TABLE 10







Summary of genes affecting glucocorticoid receptors and nitric oxide


production










Gene




Expression



Compared


Gene
to Control
Role





Kinesin

Transport of organelles




from the (−) to (+) ends.




Binds microtubules.




ATPase activity


Capping Protein

Part of dynactin-dynein




hetero-complex


Calmodulin

Directly influences calcium




dependent dynein activity.




Binds to nitric oxide




synthase and up regulates




the production of nitric




oxide


Dynein

Transport of organelles




from the (+) to (−) ends.




Binds microtubules.




ATPase activity and force




production


Dynactin

Cytoplasmic dynein




activator. Binds




mirotubules and ↑average




length of dyein movements.


Heat Shock Protein 1 beta

Necessary for


(HSP90)

glucocorticoid receptor




binding and fast transport of




dynein complex to nucleus.




Calcinuerin activity.




Enhances the nitric oxide




production by binding to




nitric oxide synthase


PPIase

Necessary for




dynein/glucocorticoid




interaction and movement


Calcinuerin

Part of dynactin-dynein




hetero-complex. Catalyzes




the conversion of arginine




to citrulline and nitric oxide


Protein kinase C

Calcium-activated,




phospholipid-dependent,




serine- and threonine-




specific enzyme.


Protein Kinase C Binding

Associated with protein


Protein

kinase C










Effect of Nutrition on Genes Involved with DNA Damage/Protection and Neural Function


Eleven genes are identified that are related to DNA damage/protection and neural function. With regard to the latter, the genes identified are important for rebound potentiation; they are believed to have a potential role in motor learning. Interestingly, of these genes, all were down regulated except for of gamma-aminobutyric acid (GABA) A receptor, gamma 2 which was up regulated. The compositions disclosed herein may be part of a therapeutic regimen to treat animals suffering from diseases or disorders associated with or relating to DNA damage/protection and neural function. The identity of these genes and their putative role in DNA damage/protection and neural function are described in Tables 11 and 12 below.









TABLE 11







Genes involved in DNA damage/protection and neural function

















% match of








probe





Best current
sequence to
Probe Target


Gene
Probe
P-value
BLAST annotation
BLAST hit
Sequence
















Gamma-
CfaAffx.26362.1.S1_at
<0.01

Homo sapiens

100
CCTCTTCTTCGGATGTTT



aminobutyric


gamma-

TCCTTCAAGGCCCCTAC


acid (GABA) A


aminobutyric acid

CATTGAT


receptor,


(GABA) A receptor;


gamma 2


gamma 2





(GABRG2);





transcript variant 1;





mRNA





Calmodulin
Cfa.4168.1.S1_at
<0.01
PREDICTED:
52.54237
CCACCCATGGTGACGAT






Canis familiaris


GACACACATCCTGGTGG





similar to

CATGCGTGTGTTGGTTT





calmodulin 1;

AGCGTTGTCTGCGTTGT





transcript variant 3

ACTAGAGCGAAAATGGG





(LOC480416);

TGTCAGGCTTGTCACCA





mRNA

TTCACACAGAAATTTAAA







AAAAAAAAAAAAANNNN







GANAAAAAACCTTTACC







AAGGGAGCATCTTTGGA







CTCTCTGTTTTTAAAACC







TCCTGAACCATGACTTG







GAGCCAGCAGATTAGGC







TGTGGCTGTGGACTTCA







GCACAACCATCAACATT







GCTGATCAAGAAATTAC







AATATACGTCCATTCCAA







GTT





Calcinuerin
Cfa.19761.1.S1_at
<0.001
PREDICTED:
98.83382
GAATTAACAATCTGCTT






Canis familiaris


GAGCCCCAAAACACTAC





similar to protein

TTATGCACTTCACTTGC





phosphatase 3

CAAAAGATTTGNGCAAG





(formerly 2B);

GTTTTGTACCCTGGTAA





catalytic subunit;

ATGATGCCAAAGTTTGT





beta isoform

TTTCTGTGGTGTTTGTCA





(calcineurin A

AATGTTCTATGTATAATT





beta); transcript

GACTGTCTGTAACATGC





variant 5

TGTTTNCTTCCTCTGCA





(LOC479248);

GATGTAGCTGCTTTCCT





mRNA

AAATCTGTCTGTCTTTCT







TTAGGTTAGCTGTATGT







CTGTAAAAGTATGTTAAA







TTAAATTACTCTATCAGA







CGCTTGTCTGTCTTTTG







ATGTAGAAGCAACTTTG







TAGCACCTTGTTTTGAG







GTNNGCTGCATTTGTTG







CTGTACTTTGTGCAT





Calcium/calmodulin-
Cfa.3884.1.S1_at
<0.01

Homo sapiens

24.10714
GGTGCTGTTCACCACAG


dependent


PTEN induced

TAAGTGGCCTCTCAGTG


protein kinase


putative kinase 1

TTGCTGACCAAAGTGTG


II


(PINK1); mRNA

AAATCCTAGAGCTTCAG







GGGAGAGGACGTGGGG







GAAATCCGGGGCTTGAC







TTTATAATAGGATTATAG







AGATGAAAAGTACACCT







TGCTTTAGGCAACAGTT







GGGATTCCTAAGACGCA







TGTGTAAGAGCATATGT







GAAATCCCTTCCCCATT







GTTGATCTCTACTCACA







GAATTTTGTCTTTATTAT







GGTGTAAGAATCACTCT







TAAAGCCACATATTCAAT







TCAAAGCAAATACGTGT







TCTGCAGTTGCAAATGT







GTATTTAATTCTTCACAA







TTCCTGTAAG





Adenylate
CfaAffx.5462.1.S1_s_at
<0.01
PREDICTED:
100
GAAACTCGGTCTGGTGT


cyclase-



Canis familiaris


TCGATGACGTCGTGGGC


associated


similar to Adenylyl

ATTGTGGAGATAATCAA


protein 1


cyclase-associated

TAGTAGGGATGTCAAAG





protein 1 (CAP 1);

TTCAGGTAATGGGTAAA





transcript variant 1

GTGCCAACCATTTCCAT





(LOC475317);

CAACAAAACAGATGGCT





mRNA

GCCATGTTTACCTGAGC







AAGAATTCCCTGGATTG







CGAAATAGTCAGTGCCA







AATCTTCTGAGATGAAT







GTCCTCATTCCTACTGA







AGGCGGTGACTATAATG







AATTCCCAGTCCCTGAG







CAGTTCAAGACCCTATG







GAATGGGCAGAAGTTGG







TCACCACAGTGACAGAA







ATTGCTGGATAAGCGAA







GTGCCACTGGGTTCTTT







GCCCTCCCCCTCACACC







ATGGGATAAATCTATCA







GGACGGTTCTTTTCTAG







ATTTCCTTTACCTTTCTG







CTCTTAAACTGCTT





Protein
Cfa.6174.1.A1_at
<0.01
PREDICTED:
100
AAATCTTACGAAGCCCA


Phosphatase I



Canis familiaris


ATATGCAGGGAGTTAAC





similar to protein

TGAAAACTATCTTGGCA





phosphatase 1A

GTGAGGTTGGCACTGTT





isoform 1;

GATAAAGCTGGTCCCTT





transcript variant 2

CCTTTAACTGTCTTTTAG





(LOC480344);

GTTGTTCTTGCCTTGTT





mRNA

GCCAGGAGTATTGCAGG







TAATACAGTATATTCATA







AGAATATCAATCTTGGG







GCTAAAATGCCTTGATT







CTTTGCACCTCTTTTACA







AGTCCTTACGTTGAATTA







CTAATTGATAAGCAGCA







GCTTCCTACATATAGTA







GGAGACTGCCACGTTTT







TGCTATCATGATTGGCT







GGGCCTGCTGCTGTTCC







TAGTAAGGTAT





Diazepam
CfaAffx.14836.1.S1_s_at
<0.01
PREDICTED:
100
AATGGTGCCATCTTACT


binding



Canis familiaris


GAGGGATTTTGTAGGCT


inhibitor


similar to

GTTTTATAGATTTTCCTA





peroxisomal

AGCCTCTGGTTGCAGTG





D3; D2-enoyl-CoA

ATAAATGGTCCAGCCAT





isomerase isoform

AGGAATCTCCGTCACCA





1 (LOC478706);

TTCTCGGGCTATTCGAT





mRNA

CTTGTGTATGCTTCCGA







CAGGGCAACATTTCACA







CTCCTTTTACTCACCTG







GGCCAAAGTCCAGAAG







GATGTTCCTCCTATACTT







TTCCCAAGATAATGGGC







CAAGCCAAGGCAGCAG







AGATGCTCATGTTTGGA







AAGAAGTTAACAGCTAG







AGAAGCCTGTGCTCAAG







GACTTGTTACTGAAGTTT







TTCCCGATAGCACTTTT







CAGAAAGAAGTTTGGAC







CAGGCTGAAAGCATATT







CAAAACTCCCCCGAAAT







ACCTTGCATATTTCCAAA







CAGAGCATCAGAAATCT







TGAGAAAGAAAAGCTAC







ATGCTGTTAACGCAGAA







GAAAACAGCGTCCTCCA







GGAAAGGTGGCTGTCA







GACGAATGCATAAATGC







AGTCATGAGCTTCTTAT







CCCGGAAGGCCAA





Tumor protein
Cfa.1611.1.A1_s_at
<0.01
PREDICTED:
97.90874
ATGATAGTTGCCATGCC


p53 binding



Canis familiaris


AACCAGCTCCAGAATTA


protein


similar to tumor

CCGCAATTATTTGTTGC





protein p53 binding

CTGCAGGGTACAGCCTT





protein; 1;

GAGGAGCAAAGAATTCT





transcript variant 4

GGATTGGCAACCCCGTG





(LOC478274);

AAAACCCTTTCCACAAT





mRNA

CTGAAGGTACTCTTGGT







GTCAGACCAACAGCAGA







ACTTCCTGGAGCTCTGG







TCTGAGATCCTCATGAC







CGGGGGGGCAGCCTCT







GTGAAGCAGCACCATTC







AAGTGCCCATAACAAAG







ATATTGCTTTAGGGGTA







TTTGACGTGGTGGTGAC







GGATCCCTCATGCCCAG







CCTCGGTGCTGAAGTGT







GCTGAAGCATTGCAGCT







GCCTGTGGTGTCACAAG







AGTGGGTGATCCAGTGC







CTCATTGTTGGGGAGAG







AATTGGATTCAAGCAGC







ATCCAAAATACAAACAT







GATTATGTTTCTCACTAA







TACTTGGTCTTAACTGAT







TTTATTCCCTGCTGTTGT







GGAGATTGTGNTTNNNC







CAGGTTTTAAATGTGTCT







TGTGTGTAACTGGATTC







CTTGCATGGATCT





Ubiquitin
CfaAffx.275.1.S1_s_at
<0.001
PREDICTED: Pan
97.19626
GATTTGGCCCGTGACCC


conjugating



troglodytes


TCCAGCACAATGTTCTG


enzyme E2D 3


LOC461941

CAGGTCCTGTTTGGGAT





(LOC461941);

GATATGTTTCATTGGCA





mRNA

AGCCACAATTATAGGAC







CTAATGACAGCCCATAT







CAAGG





NEDD8
Cfa.12556.1.A1_s_at
<0.001
PREDICTED:
99.12473
GGAATGGGCTACTCTAC


ultimate



Canis familiaris


TCATGCAGNCAAGCAGG


buster-1


similar to NEDD8

NCCTGCATCAGGCCAGT





ultimate buster-1

GGGAACCTGGACGAAG





(NY-REN-18

CCCTGAAGATTCTTCTC





antigen)

AGCAATCCTCAGATGTG





(LOC475542);

GTGGTTAAATGATTCAG





mRNA

ATCCTGAAACGANCAAC







CAGCAAGAAAGTCCTTC







CCAGGAAAACATTGACC







AACTGGTGTACATGGGC







TTCGACGCTGTGGTGGC







TGATGCTGCCTTGAGAG







TGTTCAGGGGAAACGTG







CAGCTGGCAGCTCAGN







CCCTCGCCCACAACGGA







GGAACTCTTCCTCCTGA







CCTGCAGCTCTTGGTGG







AAGACTCTTCATCAACG







CCATCCACGTCCCCTTC







CGACTCCGCAGGTACCT







CTAGTGCCTCAACAGAT







GAAGATATGGAAACCGA







AGCTGTCAATGAAATAC







TGGAAGATATTCCAGAA







CATGAAGAAGATTATCTT







GACTCAACACTGGAAG





BCL2-
CfaAffx.6742.1.S1_s_at
<0.01

Canis familiaris

100
GGCCCACCAGCTCTGA


associated X


BCL2-associated X

GCAGATCATGAAGACAG


protein (BAX)


protein (BAX);

GGGCCCTTTTGCTTCAG





mRNA

GGTTTCATCCAAGATCG







AGCAGGGCGAATGGGG







GGAGAGACACCTGAGCT







GCCCTTGGAGCAGGTG







CCCCAGGATGCATCCAC







CAAGAAGCTGAGCGAAT







GTCTCAAGCGCATCGGA







GATGAACTGGACAGTAA







CATGGAGTTGCAGAGGA







TGATCGCAGCTGTGGAC







ACAGACTCTCCCCGTGA







GGTCTTCTTCCGAGTGG







CAGCTGAGATGTTTTCT







GATGGCAACTTCAACTG







GGGCCGGGTTGTTGCC







CTCTTCTACTTTGCCAG







CAAACTGGTGCTCA
















TABLE 12







Summary of genes important for rebound potentiation


and DNA integrity










Gene




Expression



Compared


Gene
to Control
Role





Gamma-aminobutyric acid

Involved in single channel


(GABA) A receptor,

conductance (Cl-channel)


gamma 2


Calmodulin

Influx of calcium results in




calcium/calmodulin




complex which activates




CaMKII and calcineurin


Calcinuerin

Involved in the pathway for




RP suppression


Calcium/calmodulin-

Involved in induction and


dependent protein kinase II

suppression of RP


Adenylate cyclase-

Adenlyl cyclase is involved


associated protein 1

in suppression of RP


Protein Phosphatase I

Dephosphorylates




components in stress-




activated pathways. Active




PP-1 results in CaMKII




inhibition and RP




suppression


Diazepam binding inhibitor

Displaces benzodiazepine




Down regulates the effects




of GABA


Tumor protein p53 binding

Keep the cell from


protein

progressing through the cell




cycle if there is damage to




DNA present.


Ubiquitin conjugating

The regulated proteolysis of


enzyme E2D 3

proteins by proteasomes


(and NEDD8 ultimate

removes denatured,


buster-1)

damaged or improperly




translated proteins from




cells and regulates the level




of proteins like cyclins or




some transcription factors


BCL2-associated X protein

Accelerates programmed




cell death by binding to, and




antagonizing the apoptosis




repressor




BCL2










Effect of Nutrition on Genes Involved with Glucose Metabolism


Twenty four genes associated with glucose metabolism are down regulated in animals fed the super senior diet which would suggest that these animals are utilizing fat (fat oxidation) instead of glucose as a fuel source. The compositions disclosed herein may be part of a therapeutic regime in diabetic animals and/or for obesity prevention or treatment in an animal. These down regulated genes are identified and their putative role in glucose metabolism described in detail below in Tables 13 and 14.









TABLE 13







Genes involved in Glucose Metabolism

















% match of








probe





Best current
sequence to
Probe Target


Gene
Probe
P-value
BLAST annotation
BLAST hit
Seq.
















Phosphorylase
Cfa.10856.1.S1_at
<0.01
PREDICTED: Canis
99.3392
GAAAGTTCACCA



kinase



familiaris similar to


CTGCATGTTTTAT





phosphorylase kinase

GATCAGATAACT





beta; transcript variant

CATTGAAATGAG





2 (LOC478139);

TCTTTGCTCTTTA





mRNA

GACTAAATTCCC







ACCTAGTACTGC







CATTAAAATGAAT







TTGCCAGCTGGT







GTGCATACTGGA







AATGAAAAGATA







CTGAAAGAATGG







AACGAATGGTGA







GCTTAACTCAGT







GGCACTGTCATA







CTGGAAAAATAC







AGTAAAATCATAA







AAACAGATCTGC







CAGCTGATGTTT







TTATTCTCAGAAA







CAGCATTGTTGA







TAATATTTTAGTA







TACAGAGCTACT







GTACAATTTTTAC







CTTGNAAACATG







ACTGTGGTTTTG







TATTTGTGTTGAC







TTTAGGGGTTGG







GATAAAATNCAG







TATAATATATACC







TTATCAAACNTTT







TCTTTGAGCTCTT







ACTAAAAATATG







GCATGCATAAGA







TTGTTCAGAAGA







GTAGACTGTTAA







CCTAGTTTGTA





Phosphorylase
Cfa.10412.1.A1_s_at
<0.01
PREDICTED: Canis
99.36306
CTTCCAGAGCTG






familiaris


AAGCTGGCCATT





phosphorylase;

GATCNAAATTGA





glycogen; liver;

CAATGGCTTCTT





transcript variant 1

CTCTCCCAAGCA





(PYGL); mRNA

GCCTGNCCTCTT







CAAAGATTTAATC







AATATGCTATTTT







ATCATGACAGGT







TTAAAGTCTTCG







CAGACTATGAAG







CCTATGTCAAGT







GTCAAGAAAAAG







TCAGCCAGCTGT







ACATGAATCCAA







AGGCCTGGAACA







CAATGGTACTCA







AAAACATAGCTG







CCGCAGGGAAGT







TCTCTAGTGACC







GAACAATTAAGG







AATATGCCAGGG







ACATCTGGAACA







TGGAACCTTCAG







ATCTCAAGATTTC







CCTATCCAATG





Glycogen
Cfa.913.1.A1_s_at
<0.01
PREDICTED: Canis
99.49622
GACTCCACCGGA


synthase kinase 3



familiaris similar to


GGCAATTGCACT





Glycogen synthase

GTGTAGCCGTCT





kinase-3 beta (GSK-3

GCTGGAGTATAC





beta); transcript

ACCAACTGCCCG





variant 1

ATTGACACCACT





(LOC478575); mRNA

GGAAGCTTGTGC







ACATTCATTTTTT







GATGAATTAAGG







GACCCAAATGTC







AAACTACCAAAT







GGGCGAGACACA







CCTGCACTCTTC







AACTTCACCACT







CAAGAACTGTCA







AGTAATCCACCT







CTAGCTACCATC







CTTATTCCTCCTC







ATGCTCGGATTC







AAGCAGCTGCTT







CAACCCCTACAA







ATGCCACAGCAG







CCTCAGATGCTA







ATGCCGGAGACC







GTGGACAGACGA







ACAATGCCNCTT







CTGCATCAGCTT







CTAACTCCACCT







GAACAGTCCCGA







GCAGCCAGCTGC







ACAGGAAGAACC







ACCAGTTACTTG







AGTGTCACTCA





Calmodulin
Cfa.4168.1.S1_at
<0.01
PREDICTED: Canis
52.54237
CCACCCATGGTG






familiaris similar to


ACGATGACACAC





calmodulin 1;

ATCCTGGTGGCA





transcript variant 3

TGCGTGTGTTGG





(LOC480416); mRNA

TTTAGCGTTGTCT







GCGTTGTACTAG







AGCGAAAATGGG







TGTCAGGCTTGT







CACCATTCACAC







AGAAATTTAAAAA







AAAAAAAAAAAN







NNNGANAAAAAA







CCTTTACCAAGG







GAGCATCTTTGG







ACTCTCTGTTTTT







AAAACCTCCTGA







ACCATGACTTGG







AGCCAGCAGATT







AGGCTGTGGCTG







TGGACTTCAGCA







CAACCATCAACA







TTGCTGATCAAG







AAATTACAATATA







CGTCCATTCCAA







GTT





Protein Kinase C
CfaAffx.408.1.S1_s_at
<0.01
PREDICTED: Canis
99.64664
TTCAGTTCCTGT






familiaris similar to


CTCATGGCCGCT





myeloid-associated

CCCGGGACCATG





differentiation marker

CCATCGCCGCCA





(LOC611521); mRNA

CTGCCTTCTCCT







GCATCGCTTGTG







TGGCTTATGCCA







CCGAAGTGGCCT







GGACCCGGGCC







CGTCCCGGAGAG







ATCACCGGCTAC







ATGGCCANTGTG







CCGGGCCTGCTC







AAGGTGCTGGAG







ACCTTTGTGGCC







TGCATCATCTTC







GCCTTCATCAGC







AACCCCTCCCTG







TACCAGCACCAG







CCGGCCCTGGA







GTGGTGTGTGGC







CGTCTACTCCAT







CTGTTTCATCCT







GGCGGCTGTGG







CCATCCTACTGA







ACCTGGGGGACT







GCACCAACATGC







TGCCCATCTCCT







TCCCCAGTTTCC







TGTCGGGCCTGG







CCCTGCTCTCCG







TCCTGCTGTATG







CCACGGCTCTGG







NTCTCTGGCCGC







TCTACCAGTTCA







ACGAGAAGTATG







GTGGCCAGCCCC







GTCGGTCGAGG







GATGTTAGCTGC







GCCGACAGGCAC







ACCTACTACGTG







TGTACCTGGGAC







CGCCGCCTGGCT







GTGGCCATCCTG







ACAGCCATCAAC







CTGCTGGCTTAC







GTGGCTGACCTG







GTGTAC





Protein Kinase C
Cfa.15485.1.A1_s_at
<0.01
PREDICTED: Canis
100
GGAGCAGTCAGA


Binding Protein



familiaris similar to


ACTAAGACATGG





protein kinase C

TCCGTTTTACTAT





binding protein 1

ATGAAGCAGCCA





isoform b; transcript

CTCACCACAGAC





variant 11

CCTGTTGATGTT





(LOC477252); mRNA

GTACCGCAGGAT







GGACGGAA





Hexokinase 3
Cfa.19125.2.S1_at
<0.01

Macaca fascicularis

76.70683
TAATGACTGCCA





testis cDNA; clone:

ACTCACTGTTTGT





QtsA-14856; similar to

TGGAGTTATATG





human receptor

CAGAAATAAAGN





associated protein 80

CCAAGTCTTCAG





(RAP80); mRNA;

AAACAGGCTTCA





RefSeq:

GGATGCCCTCAC





NM_016290.3

CAGGGATGGAAG







AGGCAGGCTGCA







GCAAAGAGATGC







AGAGTTCCCTTG







CACATCTCGACT







TAAATGAGTCTC







CCATCAAGTCTTT







TGTTTCCATTTCA







GAAGCCACAGAT







TGCTTAGTGGAC







TTTAAAAAGCAAC







TTAACGTTCGGC







AAGGTAGTCGGA







CACGGACCAAAG







CAGGCAGAGGAA







GAAGGAGAAAAC







CCTGAATTTCTA







GGGTCCAGACAC







CCGACAAAACCA







TTAGCAATAGGG







GTGGGCCGTGTC







ATTAAGTCTTAGT







GGCTTCTGTTTC







ATTGTTGAACAA







GTTTTTTGGCCC







NGCAGTTTTCAC







CACCAGCACCAA







CTCAGCATTCTT







GTTTTGATGTTTT







CTATAAGCTATAC







AGACAATTGTGT







ATAGTATTCTGTT







TTATAACAGTCTG







GATTCACTT





Fructose 1,6
CfaAffx.26135.1.
<0.01
PREDICTED: Canis
100
AGTGGCGCTGTG


bisphosphatase
S1_s_at


familiaris aldolase A;


TGCTGAAAATTG





transcript variant 1

GGGAACACACTC





(LOC479787); mRNA

CCTCAGCCCTTG







CGATCATGGAAA







ATGCCAACGTTC







TGGCCCGTTAT





Glyceraldehyde
AFFX-
<0.01

Canis familiaris

100
AGCTCACTGGCA


3-phosphate
Cf_Gapdh_3_at

glyceraldehyde-3-

TGGCCTTCCGTG


dehydrogenase


phosphate

TCCCCACCCCCA





dehydrogenase

ATGTATCAGTTGT





(GAPDH); mRNA

GGATCTGACCTG







CCGCCTGGAGAA







AGCTGCCAAATA







TGACGACATCAA







GAAGGTAGTGAA







GCAGGCATCGGA







GGGACCCCTCAA







AGGCATCCTGGG







CTACACTGAGGA







CCAGGTGGTCTC







CTGTGACTTCAA







CAGTGACACCCA







CTCTTCCACCTT







CGACGCCGGGG







CTGGCATTGCCC







TCAATGACCACT







TTGTCAAGCTCA







TTTCCTGGTATG







ACAATGAATTTG







GCTACAGCAACC







GGGTGGTGGAC







CTCATGGTCTAC







ATGG





Glucose 6-
Cfa.19351.1.S1_at
<0.01

Homo sapiens cDNA

15.11194
GAATGTGTTGGC


phosphate


FLJ30869 fis; clone

AGACTGAGGCCC


dehydrogenase


FEBRA2004224

CCCATGTTTTTAA







TGCGCACTGGGG







ACAACCATCTAA







GGTCTAGAAACT







TTTGGACCATAG







GAAAGATAGGTT







TATGGTCCTCTT







CCAGATGCAGCC







CTAGGAGAGCAT







TCCCATGGGGTC







TCTGGATCCCTT







TCNTTGCTCTGT







GAGGCTCTGTGA







CCACCTTTTGNN







NTGNNGGGGGC







AGGGGGNCTTCC







TCAGCTCCGCCT







CCAGTGCCCCCA







GGTCCCCCACGG







CTCACAGTCCNT







GAAAATTCAGAG







CTGCCCTGTAAG







GATTTTGTCCACT







GGGCAATTCAGA







TATACTTCGATAT







CCCTGAGAAAGA







AGAGGCAGCAGC







AAACACTCCCNA







GGGCATCTGTCT







CAGNANTCTCTC







NTTGNATGAGAC







AGAAGCCTACTT







TTCAGAAANCTTA







TCANGGNTACTT







TATAAGAAACTTT







TTTTTTTTTNCTA







AAATCAGACAAA







AGGTGGCTTNTG







CATATTCTTNATT







AATAACTGTGTCT







TTGTCTCCTCTG







CTTAACTTTAGGA





Enolase
CfaAffx.30133.1.
<0.01
PREDICTED: Canis
97.72257
GGTACATCACGC



S1_s_at


familiaris similar to


CTGATCAGCTGG





T21B10.2b; transcript

CTGACCTCTACA





variant 1

AGTCCTTCATCA





(LOC479597); mRNA

GGGACTACCCAG







TGGTGTCTATCG







AAGACCCCTTCG







ACCAGGATGACT







GGGAAGCTTGGC







AGAAATTCACTG







CCAGCGCTGGAA







TCCAGGTGGNGG







GGGANGATCTCA







CCGTGACCAACC







CAAAGCGGATTT







CCAAGGCTGTGG







GCGAGAAATNGT







GCAACTGCCTCC







TGCTTAAAGTGA







ACCAGATTGGCT







CTGTGACCGAGT







CTCTTCAGGCGT







GCAAGCTGGCCC







AGTCCAATGGGT







GGGGCGTCATG







GTGTCGCATCGC







TCCGGGGAGACC







GAAGATACCTTC







ATCGCTGACCTG







GTGGTGGGANTC







TGCACTGGGCAG







ATCAAGACGGGT







GCACCATGCAGA







TCTGAGCGCTTG







GCCAAGTACAAC







CAGATCCTCAGA







ATTGAAGAGGAA







CTGGGTAGCAAG







GCCAAGTTCGCC







GGCAGAAGCTTC







AGAA





Lactate
Cfa.300.1.S1_at
<0.01
PREDICTED: Canis
97.99427
ATCTGACCTGTT


dehydrogenase



familiaris similar to L-


ACTCAAGTCGTA





lactate

ATATTAAAATGGC





dehydrogenase A

CTAAGAAAAAAA





chain (LDH-A) (LDH

CATCAGTTTCCTA





muscle subunit)

AAGTTACACATA





(LDH-M)

GGAATGGTTCAC





(Proliferation-inducing

AAAACCCTGCAG





gene 19 protein);

CTATGTCCTGAT





transcript variant 1

GCTGGATGAGAC





(LOC476882); mRNA

CTGTCTTGTGTA







GTCCTAAATTGG







TTAACGTAATATC







GGAGGCACCACT







GCCAATGTCATA







TATGCTGCAGCT







ACTCCTTAAACC







AGATGTGTATTTA







CTGTGTTTTGTAA







CTTCTGATTCCTT







CATCCCAACATC







CAACATGCCTAG







GCCATCTTTTCTT







CTTCAGTCACAT







CCTGGGATCCAA







TGTATAAATTCAA







TATTGCATGTATT







GTGCATAACTCT







TCTA





Citrate lyase
Cfa.10361.2.S1_at
<0.01
PREDICTED: Canis
98.49624
AGTATGCCAGAT






familiaris similar to


CGGAACCTTTTT





citrate lyase beta like

CCCATTTACAGTT





(LOC476974); mRNA

CATGTTAATCCAA







TTTTTTTTATTAT







CTCACTGGCCAG







TTATTCCTTTAAA







AATGAACTTCCTT







CTTTTTGATTCCA







AGCTTATGATTTT







ACTGCTCATTAAT







GTGTTACAAATAT







GCACTTAATGATT







TCACAGGGAGAT







AAAATAGTGAAG







AGAGATGGGCTG







AGGGGCTGTTAG







GACTTTAATGAAA







CAGATCTTTCCC







GAATATTTCTCCC







TTCACATTTCTCA







CATTAGATGTTTC







CCACATTGTTCTA







CTCCACACTATA







AATAATTTTAAGG







CCAATCTTAAAAA







ATGGTAGTTAAG







TGAAGGGGTTGT







GTTTATTTCACTA







GAAATCTGATAA







AACGAGAGATGA







CATAGAAAAAGT







TATCATTTTTGTT







CATACAGATGGC







TTCTAAAAATAAA







TCTTCAAAACTGA







TTACTTTTAACCT







CCACCTCCCAAA







ATGAAACATCCC







TACATTTGAACTG







CTAGGTGAGAAC







TCTGAAAGCCCT







CATCC





Glycerol kinase
CfaAffx.21204.1.
<0.01
PREDICTED: Canis
100
GGGTACATCCTA



S1_s_at


familiaris similar to


TGGCTGCTATTT





glycerol kinase

CGTCCCCGCGTT





isoform 2; transcript

TTCAGGGTTATAT





variant 8

GCACCTTACTGG





(LOC480872); mRNA

GAGCCCAGTGCA







AGAGGGATCATC







TGTGGGCTCACT







CAATTCACCAATA







AATGCCATATTG







CTTTTGCTGCATT







AGAAGCTGTTTG







TTTCCAAACCCG







GGAGATTTTGGA







TGCCATGAACCG







AGACTGCGGAAT







TCCACTCAGTCA







TTTGCAGGTAGA







TGGAGGAATGAC







CAACAACAAAATT







CTTATGCAACTA







CAAGCAGACATT







CTATATATCCCA







GTAGTGAAGCCC







TCGATGCCAGAA







ACAACTGCCCTG







GGAGCTGCCATG







GCAGCCGGGGC







TGCGGAGGGAGT







TGGTGTTTGGAG







TCTTGAACCCGA







GGATCTGTCAGC







AGTCACGATGGA







GCGATTTGAACC







CCAGATCAATGC







TGAGGAAAGTGA







AATTCGTTACTCT







ACATGGAAGAAG







GCTGTGATGAAG







TCAGTGGGCTGG







GTTACAACTCA





Transketolase
CfaAffx.13684.1.
<0.01

Homo sapiens

86.53846
GAAGATCTGGCC



S1_s_at

transketolase

ATGTTTCGGTCC





(Wernicke-Korsakoff

ATCCCCACTGCT





syndrome); mRNA

ACGATCTTTTACC





(cDNA clone

CAAGTGACGGGG





MGC: 15349

TGTCAACAGAGA





IMAGE: 4310396);

AGGCGGTGGAAT





complete cds

TAGCAGCCAATA







CAAAGGGCATCT







GCTTCATCCGGA







CCAGCCGCCCAG







AAAACGCCATCA







TCTATAACAACAA







TGAGGATTTCCA







AATCAAACAAGC







CAAGGTGGTCCT







GAAGAGCAAGGA







TGACCAGGTGAC







TGTGATTGGGGC







CGGAGTGACCCT







ACATGAGGCCTT







GGCTGCTGCTGA







ACTGCTGAAGAA







AGAGAAGATCAA







CATTCGTGTGTT







GGACCCCTTCAC







CATCAAGCCCCT







GGACAGAAATCT







CATTCTCGAAAG







CGCCCGTGCGAC







CAAGGGCAGGAT







CGTCACCGTGGA







GGACCATTACTA







TGAAGGTGGCAT







AGGTGAGGCAGT







GTCCTCTGCCTT







GGTGGGTGAGC







CTGGCATCACCG







TCTCCCGCCTTG







CAGTTGGTGAGG







TACCAAGAAGCG







GGAAGCCAGCTG







AGCTGCTGAAGA







TGTTTGGCATTG







ACAGGGACGCCA







TCGCACAAGCTG







TGAGGGACCTTG







TCGCCAA





Ribulose
Cfa.13084.1.A1_s_at
<0.01

Homo sapiens SLIT-

57.79468
CCCCAAGGAGAT


phosphate 3-


ROBO Rho GTPase

GAGGAGCGATGA


epimerase


activating protein 2

CCCCAGCAACAG





(SRGAP2); mRNA

GAANAACAGCCC







ACTGAAGGGCTG







GTGTGTGTGTNC







TTCACGTGCCAG







AAGAGAAGTTTA







GATCCTCCCAGG







GGAATCGCAATG







TTGTGGCGTCCT







GACTTGTATGTC







ACGTTTTGTGTAA







AAATGGTATATTC







TTTAAAATAGTGT







TGATAACTGGAA







TATTGTATGTATG







CTTGGAGATGCT







TTGTGTGAACCT







AAGACTGTCACT







CAACAGATGTTG







GATTGGG





Ribose 5-
Cfa.335.2.S1_at
<0.01
PREDICTED: Canis
100
AGCCTTTCTACT


phosphate



familiaris similar to


GACCCTGCAAGA


isomerase


ribose 5-phosphate

GTGGAGCGTGTT





isomerase A (ribose

CACCTTGAACCC





5-phosphate

CCAGCGTGCAGC





epimerase)

TGAGGTAGACAT





(LOC475755); partial

GCCTCTCCAGGA





mRNA

GCCTTTGCCTTA







ATGCATCTGTGC







CAGACAGACGGC







TGG





Cytochrome c
CfaAffx.4942.1.S1_s_at
<0.01
PREDICTED: Canis
100
GGCAGTTTGAAA


oxidase



familiaris similar to


ATAAAGTTCCAG


polypeptide VIIa-


cytochrome c

AGAAACAAAAGC


liver/heart,


oxidase; subunit 7a 3

TATTTCAGGAGG


mitochondrial


(LOC611134); mRNA

ATAATGGAATTC


precursor




CAGTGCATCTAA







AGGGTGGAGTAG







CTGATGCCCTCC







TGTATAGAGCCA







CTATGATGCTTA







CAGTTGGTGGAA







CAGCATATGCCA







TGTATCAGCTAG







CTGTGGCTTCTT







TTCCCAAGAAGCA





Cytochrome c
Cfa.15065.1.S1_at
<0.01
PREDICTED: Canis
99.75961
GGTCCGCAGTCG


oxidase subunit



familiaris similar to


TTCTGTGCGGTC


VIII liver form


Cytochrome c oxidase

ATGTCTGTGCTG





polypeptide VIII-liver;

GTGCCGCAGCTG





mitochondrial

CTGAGGGGCCTA





precursor

ACAGGCCTCACC





(Cytochrome c

CGGCGGCTCCC





oxidase subunit 8-2)

GGTGCATCGTGC





(LOC476040); mRNA

CCAGATCCATTC







CAAGCCGCCGC







GGGAGCAGCTC







GGGACCATGGAT







GTTGCCGTTGGG







CTCACCTNCTGC







TTCCTGTGTTTCC







TCCTGCCATCGG







GCTGGGTCCTGT







CACACCTGGAGA







GCTACAAGAAGC







GGGAGTGAAGG







GGGCTGTCCTGT







CCCTCACCCTGT







GACCTGACCACC







CCTGGCCTGTCC







TGATCATGTCTG







CTGCATTCCTGG







CCGGCCTTCCAT







GGATCATGTCCT







TCAATTACAGTG







ACCTCTTCTACA







GTCATGACCTCT







TGATTTCTCCATG







GTGACATCCTGG







GACCAAACATAT







TGGTTTATAA





Ubiquinolucytochrome c
Cfa.1425.2.A1_at
<0.01
PREDICTED: Canis
27.18053
CTTATGCATTCCT


reductase



familiaris similar to


TCCAAAATTGGA





Ubiquinol-

TCATTTAGGTCAA





cytochrome-c

ATTATTTGATGTT





reductase complex

AAATCATAAGTTT





core protein 2;

TCATTTGCTTACA





mitochondrial

TTTACGATATCAG





precursor (Complex III

CGTCAGCTACGG





subunit II); transcript

AATCAATCTGCT





variant 1

GAAGGACCGTGG





(LOC479815); mRNA

CTGGCGGCGTGT







ACGATCCAGCAA







CCAGCGCCTGG







GACCCGACTTCA







TCCAGGAACCCC







TCAGAAGACTCC







ACTGACATTAGG







AAGACTCATAAG







AACCTTACAAGA







AAAAGTATCAAC







CCCATCAAAACG







GCAGAAAAGAAA







CATATCTTGTTAT







TAGTAGCTGAAA







TTCCATTTTCTAC







ATGTTGCCATAC







CTTATAAAAACTA







CACTAAGCTACG







CTTAAGGAAATA







CATTTTCTTAAAT







AAATTAGAATTGA







AACCAATTTTTAA







GTAAATCTAGGG







NTTCAATTTATTC







TCATTGNGTNTT







GTTTCTGGTGCA







ATCATGAANAAC







AGCATNCTATTAA







CCAACCTTGGTC







CCATGTACATAA





ATP synthase
CfaAffx.3186.1.S1_s_at
<0.01
PREDICTED: Canis
98.57651
AATTGGGACTGT






familiaris similar to


GTTTGGGAGCCT





ATP synthase; H+

CATCATTGGTTAT





transporting;

NCCAGGAATCCC





mitochondrial F0

TCTCTGAAGCAA





complex; subunit c

CAGCTCTTCTCC





isoform 2a precursor

TACGCCATTCTG





(LOC477595); mRNA

GGCTTTGCCCTC







NCGGAGGCCATG







GGGCTTTTTTGC







CTGATNGTGGCC







TTTCTCATCCTCT







TNGCCATGTGAA







GGAGTCGTCTCC







ACCTCCCATAGG







TCTTTCTCCCATG







TCTTGTCTGCCC







TGTATGCCCTGT







ATGTTCCTTTTCC







TATACCTCCCCA







GGCAGCCTGGG







GAAAGTGGTTGG







CTCAGGGTTTGA







CA





NADH-
Cfa.4415.1.S1_at
<0.01
PREDICTED: Canis
98.20789
GGTGACTTTGGA


ubiquinone



familiaris similar to


CGTCCGTTCCTG


oxidoreductase


NADH-ubiquinone

CTCTGTGGAGGC





oxidoreductase

NNTGCTTCGTTC





MLRQ subunit

CGGGCCTTGCG





(Complex I-MLRQ)

GCAACTCGGTNT





(CI-MLRQ)

TTCCTTCCCCTG





(LOC477682); mRNA

CGCGGGAGACCT







CTGCCACAACCA







TGTTACGCCAGA







TCATCGGTCAGG







CCAAGAAGCATC







CGAGCTTGATCC







CCCTCTTCATATT







TATTGGGGCAGG







AGGTACTGGAGC







AGCGCTGTATGT







ATTGCGCTTGGC







ATTGTTCAATCCA







GATGTTAGTTGG







GATAGGAAGAAT







AACCCAGAACCT







TGGAACAAACTG







GGTCCCAATGAT







CAATACAAGTTCT







ACTCAGTGAATG







TAGATTACAGCA







AACTGAAGAAAG







AAGGTCCAGACT







TCTAAATGAAATG







TTTCACTATAAAG







CTGCTTAGAATG







AAGGTCTTCCAG







AAGCCATCCGCA







CAATTTTCCACTT







ATCCAGGAAATA







TTTCCCCTCTAAA







TGCACGAAATCA







TGTTGGTGTATT







GTGTTGGGGTTT







ACACTNNANNAN







TAAATATCTGAAA







CTTGANANGTGT







CACTATTTAATGC







TGAAAATTTGCTC







TGAACTTTA





Facilitated
Cfa.1370.1.A1_at
<0.01

Homo sapiens cDNA

23.95833
TTGGAAGGATGG


glucose


FLJ44038 fis; clone

ATGCTTGCCCCA


transporter/


TESTI4028880; highly

GGTCATGGACAC


Glucose


similar to Glucose

CTCCACAAATCA


transporter-like


transporter type 3;

TCTAGTTTCCCA


protein III


brain

GTATTTTTATAAA


(GLUT3)




TGGAGATTGGGC







TCCATGACACTTT







ACTTGGTCTTCC







TTCTTACATAGGT







TTTTTGATTACCC







TTTCTCTCCTTGG







TGCTTATATACTT







AAGACCCTTTAG







CCAAACCCTTGC







CAATGACAGTAT







TTCAGTCACTAG







TTCTCACTGTTTC







CTCTGATCATTG







AGCCTTTGGAAA







AAAAATCTCACA







GAGCTTATATGT







AATGGGGCTTGG







TTGAACAGATGA







CTTCCTGTAACT







GCACCTCTACTT







TTGGCTTCTCAA







AAACAGTGGGTT







GGCAGTAATGCA







GCGTGGAAGTTT







TCCCATTTCTCA







GTGAC
















TABLE 14







Summary of Genes involved in Glucose Metabolism










Gene




Expression



Compared to


Gene
Control
Role





Phosphorylase kinase

Necessary for activation of




glycogen synthase which




stores glucose as glycogen


Phosphorylase

Necessary for glycogen




conversion to glucose 1-




phosphate which feeds into




glycolysis


Glycogen synthase kinase 3

Necessary for activation of




glycogen synthase which




stores glucose as glycogen


Calmodulin

Necessary for activation of




glycogen synthase which




stores glucose as glycogen


Protein Kinase C

Necessary for activation of




glycogen synthase which




stores glucose as glycogen


Protein Kinase C Binding

Necessary for activation of


Protein

glycogen synthase which




stores glucose as glycogen


Hexokinase 3

Necessary for glucose




conversion to pyruvate to




enter the TCA cycle


Fructose 1,6

Necessary for glucose


bisphosphatase

conversion to pyruvate to




enter the TCA cycle


Glyceraldehyde 3-

Necessary for glucose


phosphate dehydrogenase

conversion to pyruvate to




enter the TCA cycle


Glucose 6-phosphate

Involved in pentose


dehydrogenase

phosphate pathway


Enolase

Necessary for glucose




conversion to pyruvate to




enter the TCA cycle


Lactate dehydrogenase

Involved in converting




private to lactate


Citrate lyase

Necessary for citrate




conversion to oxaloacetate




which feeds acetyl-CoA




into the fatty acid synthesis




pathway


Glycerol kinase

Necessary for changing




glycerol into DHAP which




feeds into glycolysis


Transketolase

Involved in pentose




phosphate pathway


Ribulose phosphate 3-

Involved in pentose


epimerase

phosphate pathway


Ribose 5-phosphate

Involved in pentose


isomerase

phosphate pathway


Cytochrome c oxidase

Associated with the


polypeptide VIIa-

production of ATP (energy


liver/heart, mitochondrial

source) in the electron


precursor

transport chain which is




associated with the TCA




cycle


Cytochrome c oxidase

Associated with the


subunit VIII liver form

production of ATP (energy




source) in the electron




transport chain which is




associated with the TCA




cycle


Ubiquinol--cytochrome c

Associated with the


reductase

production of ATP (energy




source) in the electron




transport chain which is




associated with the TCA




cycle


ATP synthase

Associated with the




production of ATP (energy




source) in the electron




transport chain which is




associated with the TCA




cycle


NADH-ubiquinone

Associated with the


oxidoreductase

production of ATP (energy




source) in the electron




transport chain which is




associated with the TCA




cycle


Facilitated glucose

Involved in glucose uptake


transporter/Glucose


transporter-like protein-III


(GLUT3)









Example 5
Comparison of Gene Expression Profiles of Genes Associated with the Aging Process: Healthy Adult Dogs Versus Senior Dogs in Comparison to Control Diet Versus Super Senior Diet

A dog's gene expression profile changes as the dog ages from being an adult dog to becoming a geriatric (senior) dog. This is true for genes associated with numerous biological pathways such as, e.g., glucose metabolism, blood clotting and bone and muscle integrity but also with regard to genes that have been associated with the aging process, or senescence, in general. With regard to this class of “aging” associated genes, we have found that, by feeding senior dogs a super senior diet according to the present invention, the gene expression profile of certain of these genes in lymphocytes tends to move towards the profile of an adult dog from that of a geriatric dog. Thus, geriatric dogs fed a super senior diet according to the present invention can have their genetic profile altered to resemble more closely the genetic profile of a healthy adult dog.


The results displayed below in Tables 15-20, show that genes normally altered with the aging process can be regulated through nutritional strategies targeted at common aging changes. Specifically, the results show that, when fed a super senior diet, generally the expression levels of the genes in lymphocytes move in the opposite direction as that of the expression level in a healthy adult animal compared to the expression level in a geriatric animal. That is, when the expression level in a healthy adult animal is high compared to a geriatric animal (i.e., “down regulated” in the geriatric animal), the super senior fed geriatric animals generally also have higher expression level (altered to be “up regulated”) as compared to a geriatric animal fed the control diet. Similarly, when the expression level in a healthy adult animal is low compared to a geriatric animal (“up regulated” in the geriatric animal), the super senior fed geriatric animals generally also have lower expression level (altered to be “down regulated”) as compared to a control diet fed geriatric animal. Thus, expression levels of aging related genes in geriatric dogs may be beneficially altered when the geriatric dog is fed a super senior diet of the present invention and thus the dogs may therefore lead lives of improved quality.









TABLE 15







Aging Genes Associated With Inflammation









Direction of



Expression













Super





Senior




Adult v.
v. Control


Annotation
Probe ID.
Geriatric
Diet





C1q and tumor
CfaAffx.26423.1.S1_at
up
down


necrosis factor


related protein 2


TNFRSF1A-
CfaAffx.31209.1.S1_s_at
down
down


associated via


death domain


isoform 1


T-cell surface
CfaAffx.15424.1.S1_at
down
down


antigen CD2


precursor


Delta-
Cfa.17192.1.S1_s_at
down
down


aminolevulinic


acid dehydratase


Attractin precursor
CfaAffx.10508.1.S1_at
up
up


Cytochrome C
Cfa.16058.1.A1_x_at
up
up


oxidase subunit III


NEDD4-like
Cfa.8453.1.A1_at
up
up


ubiquitin-protein


ligase 1


Ubiquitin specific
CfaAffx.23104.1.S1_at
down
down


peptidase 11


Cartilage
Cfa/15775.1.A1_at
up
up


intermediate layer


protein
















TABLE 16







Genes Associated With DNA Repair/Cell Survival









Direction of



Expression













Super Senior




Adult v.
v. Control


Annotation
Probe
Geriatric
Diet





Gemin 7
CfaAffx.7805.1.S1_s_at
down
down


Iroquis-class
CfaAffx..16317.1.S1_at
up
down


homeodomain


protein IRX-4


Chromobox
CfaAffx.9308.1.S1_at
down
down


homolog 4


Ubiquitin specific
CfaAffx.23104,1,S1_at
down
down


peptidase 11


ADP-ribosylation
Cfa.13803.1.S1_s_at
down
down


factor-like 10B


Eukaryotic
CfaAffx.4294.1.S1_at
up
up


translation initiation


factor 5B


General
CfaAffx.1649.1.S1_s_at
down
down


transcription factoR


11 H. polypeptide 4


NEDD4-like
Cfa,8453.1.A1_at
up
up


ubiquitin-protein


ligase 1


Poly(ADP-
Cfa.5341.1.A1_at
up
down


ribose)polymerase


family, member 8
















TABLE 17







Aging Genes Associated With Fat/Cholesterol Metabolism









Direction of



Expression













Super Senior




Adult v.
v. Control


Annotation
Probe
Geriatric
Diet





Phophomevanonate
Cfa.1406.1.S1_s_at
down
down


kinase


5-AMP-activated
CfaAffx.13848.1.S1_at
down
down


protein kinase,


gamma-1 subunit


Apoliprotein A-II
Cfa.8770.1.S1_at
down
down


precursor


C1q and tumor
CfaAffx.26423.1.S1_at
up
down


necrosis factor


related protein 2


Attractin precursor
CfaAffx.10508.1.S1_at
up
up
















TABLE 18







Aging Genes Associated With Protein Synthesis









Direction of



Expression













Super Senior




Adult v.
v. Control


Annotation
Probe
Geriatric
Diet





Branched chain
Cfa.17186.1_s_at
down
down


keto acid


dehydrogenase E1,


alpha polypeptide


Seryl-tRNA
CfaAffx.9380.1.S1_s_at
down
down


synthesis


Mitochondrial 28S
CfaAffx.200.1.S1_at
down
down


ribosomal protein


S33


Ribosomal protein
CfaAffx.416.1.S1_x_at
down
up


S3a


60S ribosomal
CfaAffx.802.1.S1_at
down
up


protein L21
















TABLE 19







Aging Genes Associated With Cell Growth/Death









Direction of



Expression













Super




Adult v.
Senior v.


Annotation
Probe
Geriatric
Control Diet





Sorting nexin-9
Cfa.1874.1.S1_at
up
up


Cell growth
Cfa.3200.1.S1_s_at
down
down


regulator with


RING finger


domain 1


Solute carrier
CfaAffx.14864.1.S1_at
up
up


family 39 (zinc


transporter)


Choline kinase
Cfa.8353.1.A1_at
down
down


alpha isoform a


Kv Channel
Cfa.3460.1.S1_at
up
down


interacting protein 2


Ribosomal
CfaAffx.416.1.S1_x_at
down
up


protein S3a


Ubiquin specific
CfaAffx.23104.1.S1_at
down
down


peptidase 11
















TABLE 20







Genes Altered With Age and Super Senior Diet with


Unknown Functions









Direction of



Expression













Super Senior




Adult v.
v. Control


Annotation
Probe
Geriatric
Diet





Protein
Cfa.13958.1.A1_at
up
up


KIAA0406


Hypothetical
CfaAffx.12332.1.S1_at
up
up


LOC477905


Nitilase 1
CfaAffx.19716.1.S1_s_at
down
down


CG5645-PA
CfaAffx.27184.1.S1_at
down
down


Transcribed locus
Cfa.12738.1.A1_at
up
up


Transcribed locus
Cfa.18839.1.S1_at
up
down








Claims
  • 1. A method for modulating biological functions associated with the aging process of a senior or super senior companion animal comprising feeding the animal a composition comprising: at least about 9% by weight protein;at least about 5% by weight fat; andat least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid.
  • 2. The method of claim 1, wherein the biological functions associated with the aging process comprises inflammation, DNA repair or cell survival, fat or cholesterol metabolism, protein synthesis, cell growth and cell death.
  • 3. The method of claim 1 wherein the animal is chosen from a cat, a dog, and a horse.
  • 4. A method for modulating biological functions associated with the aging process of a senior or super senior animal comprising feeding the animal a composition comprising: at least one omega-3 polyunsaturated fatty acid chosen from docosahexaenoic acid and eicosapentaenoic acid;at least one antioxidant; andat least one nutrient chosen from choline, manganese, methionine, cysteine, L-carnitine, lysine, and mixtures thereof.
  • 5. The method of claim 4 wherein the omega-3 polyunsaturated fatty acid in the composition is DHA and wherein the composition comprises at least about 0.02% by weight DHA as measured on a dry matter basis.
  • 6. The method of claim 4 wherein the omega-3 polyunsaturated fatty acid in the composition is DHA and wherein the composition comprises about 0.02% to about 0.40% by weight DHA as measured on a dry matter basis.
  • 7. The method of claim 4 wherein the omega-3 polyunsaturated fatty acid in the composition comprises EPA and wherein the composition comprises at least about 0.1% by weight EPA as measured on a dry matter basis.
  • 8. The method of claim 4 wherein the omega-3 polyunsaturated fatty acid in the composition comprises EPA, and wherein the composition comprises about 0.1% by weight to about 1% by weight EPA as measured on a dry matter basis.
  • 9. The method of claim 4 wherein the omega-3 polyunsaturated fatty acid in the composition comprises a mixture of DHA and EPA, and wherein the composition comprises at least about 0.02% by weight DHA and at least about 0.1% by weight EPA on a dry matter basis.
  • 10. The method of claim 4 wherein the composition comprises one or more antioxidants chosen from vitamin E, vitamin C, taurine, beta-carotene, carnitine, lipoic acid, and cystine.
  • 11. The method of claim 4 wherein the composition comprises at least about 500 IU/kg vitamin E, at least about 50 ppm vitamin C and at least about 600 ppm taurine.
  • 12. The method of claim 4 wherein the composition further comprises at least about 1000 ppm choline.
  • 13. The method of claim 4 wherein the composition fed to the animal is an animal treat or an animal toy.
  • 14. The method of claim 4 wherein the composition fed to the animal as a nutritional supplement.
  • 15. A method for modulating biological functions associated with the aging process of a senior or super senior small or regular breed canine comprising feeding the animal a composition comprising: about 60% to about 70% by weight carbohydrate;about 15% to about 25% by weight protein chosen from animal protein and vegetable protein;about 5% to about 7% by weight fat chosen from animal fat and vegetable fat;about 2.5% to about 4% by weight of at least one omega-3 polyunsaturated fatty acids;about 1% to about 2% by weight fiber;about 1% to about 2% by weight minerals; andabout 0.5 to about 1.5% by weight vitamins.
  • 16. A method for modulating biological functions associated with the aging process of a senior or super senior large breed dog, wherein the method comprises feeding the animal a composition comprising: about 60% to about 70% by weight carbohydrate; about 15% to about 25% by weight protein chosen from animal protein and vegetable protein;about 5% to about 7% by weight fat chosen from animal fat and vegetable fat;about 3% to about 5% by weight of at least one omega-3 polyunsaturated fatty acids;about 1% to about 1.5% by weight fiber;about 0.5% to about 1% by weight minerals; andabout 0.75 to about 1.25% by weight vitamins.
  • 17. A method for modulating biological functions associated with the aging process of a senior or super senior cat, wherein the method comprises feeding the animal a composition comprising: about 30% to about 35% by weight carbohydrate;about 40% to about 50% by weight protein chosen from animal protein and vegetable protein;about 12% to about 15% by weight fat chosen from animal fat and vegetable fat;about 1% to about 2% by weight of at least one omega-3 polyunsaturated fatty acids;about 3% to about 5% by weight fiber;about 1% to about 2% by weight minerals; andabout 1% to about 2% by weight vitamins.
  • 18. The method of claim 1 wherein the method comprises feeding the animal the composition in an amount effective to modulating biological functions associated with the aging process, wherein modulation of biological functions associated with the aging process is evidenced by improvement in one or more biological pathways chosen from blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport.
  • 19. The method of claim 1 wherein the method comprises feeding the animal the composition in an amount effective to modulating biological functions associated with the aging process, wherein modulation of biological functions associated with the aging process is evidenced by a beneficial change in expression of one or more genes which encode proteins associated with or related to biological pathways chosen from blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport.
  • 20. A method to treat an animal suffering from a disorder or disease associated with or related to aging chosen from blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport comprising administering to said animal an effective amount of a composition comprising at least about 9% by weight protein, at least about 5% by weight fat, and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid.
  • 21. The method of claim 20 wherein said composition further comprises at least one omega-3 polyunsaturated fatty acid chosen from docosahexaenoic acid (“DHA”) and eicosapentaenoic acid (“EPA”).
  • 22. The method of claim 20 wherein said composition further comprises at least one antioxidant and at least one nutrient chosen from choline, manganese, methionine, cysteine, L-carnitine, lysine, and mixtures thereof.
  • 23. A method to treat an animal suffering from a disorder or disease associated with or related to a biological pathway chosen from blood clotting and platelet activation and aggregation, bone and muscle integrity, inflammatory responses, cartilage degradation and pain response, DNA damage and repair pathways, neural function, glycogen synthesis and degradation, glycolysis, gluconeogenesis, the pentose phosphate pathway, the aging process, and electron transport comprising administering to said animal an effective amount of a composition comprising the components disclosed in Table 1 or Table 1A.
  • 24. The method of claim 1 wherein the method further comprises measuring the enhancement in the quality of life of said animal comprising quantitating the gene expression levels of one or more genes chosen from those disclosed in Tables 5-14 in said animal prior to and after feeding said composition and comparing said levels in the animal wherein an enhancement in the quality of life of said animal is reflected by a beneficial change in gene expression levels in said animal.
  • 25. The method of claim 1 wherein the method comprises feeding the animal the composition in an amount effective to enhance the animal's quality of life, wherein enhanced quality of life is evidenced by a beneficial change in expression of one or more aging genes which encode proteins associated with or related to biological pathways chosen from inflammation, DNA repair, cell survival, fat or cholesterol metabolism, protein synthesis, cell growth and cell death.
  • 26. The method of claim 31 wherein the change in expression is in one or more genes listed on Tables 15-19 and wherein the change in expression is towards the expression level in a healthy adult companion animal as compared to the expression level in a geriatric animal.
  • 27. The method of claim 32 wherein said animal is a dog.
  • 28. The method of claim 1 wherein the method comprises feeding the animal the composition in an amount effective to enhance the animal's quality of life, wherein enhanced quality of life is evidenced by a change in expression of one or more genes listed on Table 20 and wherein the change in expression is towards the expression level in a healthy adult animal as compared to the expression level in a geriatric animal.
  • 29. The method of claim 34 wherein said animal is a dog.
  • 30. A methods of altering the expression of at least one peptide in a mammal, the method comprising administering to the mammal a composition comprising: at least about 9% by weight protein;at least about 5% by weight fat; andat least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid,wherein the at least one peptide is selected from the group consisting of X, Y and Z.
  • 31. The method of claim 30, wherein the mammal is a dog, cat or horse.
  • 32. The method of claim 30, wherein the mammal is a geriatric mammal.
  • 33. The method of claim 30, wherein the composition further comprises at least one of an antioxidant, choline, manganese, methionine, cysteine, L-carnitine, lysine or a combination thereof.
  • 34. The method of claim 30, wherein the expression of the at least one gene is increased.
  • 35. The method of claim 36, wherein the expression of the at least one gene is decreased.
  • 36. A method for screening one or more test compounds for its ability to alter the expression of at least one gene of interest in a mammal, the method comprising a) administering a control composition to a control group of mammals and determining the levels of expression of the at least one gene of interest,b) administering the one or more test compositions to an experimental group of mammals and determining the levels of expression of the least one gene of interest, wherein the test composition comprises at least about 9% by weight protein; at least about 5% by weight fat; and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid, andc) determining the differences in expression levels in the at least one gene of interest between the control and experimental groups of mammals after each group has been administered their respective compositions,wherein a difference in the expression levels of the at least one gene of interest indicates that the test composition is capable of altering the expression of the at least one gene of interest.
  • 37. The method of claim 36, where the difference indicates that levels of expression of the at least one gene of interest is increased in the experimental group compared to the control group.
  • 38. The method of claim 36, where the difference indicates that levels of expression of the at least one gene of interest is decreased in the experimental group compared to the control group.
  • 39. The method of claim 36, wherein the at least one gene of interest is selected from the group consisting of X, Y and Z.
  • 40. The method of claim 36, wherein the levels of expression of more than one gene of interest are determined.
  • 41. A method for screening one or more test compounds for its ability to alter the expression of at least one gene of interest in a mammal, the method comprising a) administering a control composition to a control group of mammals and determining the levels of expression of the at least one gene of interest, wherein the control composition comprises at least about 9% by weight protein; at least about 5% by weight fat; and at least about 0.05% by weight of at least one omega-3 polyunsaturated fatty acid,b) administering the one or more test compositions to an experimental group of mammals and determining the levels of expression of the least one gene of interest, andc) determining the differences in expression levels in the at least one gene of interest between the control and experimental groups of mammals after each group has been administered their respective compositions,wherein a difference in the expression levels of the at least one gene of interest indicates that the test composition is capable of altering the expression of the at least one gene of interest.
  • 42. The method of claim 41, where the difference indicates that levels of expression of the at least one gene of interest is increased in the experimental group compared to the control group.
  • 43. The method of claim 41, where the difference indicates that levels of expression of the at least one gene of interest is decreased in the experimental group compared to the control group.
  • 44. The method of claim 41, wherein the at least one gene of interest is selected from the group consisting of X, Y and Z.
  • 45. The method of claim 44, wherein the levels of expression of more than one gene of interest are determined.
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US2009/051155 7/20/2009 WO 00 4/8/2011
Provisional Applications (1)
Number Date Country
61082183 Jul 2008 US