METHODS FOR INHIBITING MUSCLE ATROPHY

In one aspect, the invention relates methods for inhibiting or preventing muscle atrophy or increasing muscle mass by providing to a subject in need thereof an effective amount of ursolic acid, a derivative thereof, or an analog of the ursane scaffold. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 13/698,645 (having a 371(c) date of Apr. 22, 2013), which is a national phase application of International Application No. PCT/US11/37238, filed May 19, 2011, which claims the benefit of U.S. Provisional Application Nos. 61/346,813, filed on May 20, 2010, and 61/445,488, filed on Feb. 22, 2011. The entire contents of each of the prior applications are hereby incorporated by reference in their entereity herein.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

This invention was made with government support under grant VA Career Development Award-2 to Christopher M. Adams, and support from a VA Research Enhancement Award Program to Steven D. Kunkel. The United States government has certain rights in the invention.

BACKGROUND

Skeletal muscle atrophy is characteristic of starvation and a common effect of aging. It is also a nearly universal consequence of severe human illnesses, including cancer, chronic renal failure, congestive heart failure, chronic respiratory disease, insulin deficiency, acute critical illness, chronic infections such as HIV/AIDS, muscle denervation, and many other medical and surgical conditions that limit muscle use. However, medical therapies to prevent or reverse skeletal muscle atrophy in human patients do not exist. As a result, millions of individuals suffer sequelae of muscle atrophy, including weakness, falls, fractures, opportunistic respiratory infections, and loss of independence. The burden that skeletal muscle atrophy places on individuals, their families, and society in general, is tremendous.

The pathogenesis of skeletal muscle atrophy is not well understood. Nevertheless, important advances have been made. For example, it has been been described previously that insulin/IGF1 signaling promotes muscle hypertrophy and inhibits muscle atrophy, but is reduced by atrophy-inducing stresses such as fasting or muscle denervation (Bodine S C, et al. (2001) Nat Cell Biol 3(11):1014-1019; Sandri M, et al. (2004) Cell 117(3):399-4121; Stitt T N, et al. (2004) Mol Cell 14(3):395-403; Hu Z, et al. (2009) The Journal of clinical investigation 119(10):3059-3069; Dobrowolny G, et al. (2005) The Journal of cell biology 168(2):193-199; Kandarian S C & Jackman R W (2006) Muscle & nerve 33(2):155-165; Hirose M, et al. (2001) Metabolism: clinical and experimental 50(2):216-222; Pallafacchina G, et al. (2002) Proceedings of the National Academy of Sciences of the United States of America 99(14):9213-9218). The hypertrophic and anti-atrophic effects of insulin/IGF1 signaling are mediated at least in part through increased activity of phosphoinositide 3-kinase (PI3K) and its downstream effectors, including Akt and mammalian target of rapamycin complex 1 (mTORC1) Sandri M (2008) Physiology (Bethesda) 23:160-170; Glass D J (2005) The international journal of biochemistry & cell biology 37(10): 1974-1984).

Another important advance came from microarray studies of atrophying rodent muscle (Lecker S H, et al. (2004) Faseb J 18(1):39-51; Sacheck J M, et al. (2007) Faseb J 21(1):140-155; Jagoe R T, et al. Faseb J 16(13):1697-1712). Those studies showed that several seemingly disparate atrophy-inducing stresses (including fasting, muscle denervation and severe systemic illness) generated many common changes in skeletal muscle mRNA expression. Some of those atrophy-associated changes promote muscle atrophy in mice; these include induction of the mRNAs encoding atroginI/MAFbx and MuRF1 (two E3 ubiquitin ligases that catalyze proteolytic events), and repression of the mRNA encoding PGC-1 α (a transcriptional co-activator that inhibits muscle atrophy) (Sandri M, et al. (2006) Proceedings of the National Academy of Sciences of the United States of America 103(44):16260-16265; Wenz T, et al. Proceedings of the National Academy of Sciences of the United States of America 106(48):20405-20410; Bodine S C, et al. (2001) Science (New York, N.Y. 294(5547):1704-1708; Lagirand-Cantaloube J, et al. (2008) The EMBO journal 27(8):1266-1276; Cohen S, et al. (2009) The Journal of cell biology 185(6):1083-1095; Adams V, et al. (2008) Journal of molecular biology 384(1):48-59). However, the roles of many other mRNAs that are increased or decreased in atrophying rodent muscle are not yet defined. Data on the mechanisms of human muscle atrophy are even more limited, although atrogin-1 and MuRF1 are likely to be involved (Leger B, et al. (2006) Faseb J20(3):583-585; Doucet M, et al. (2007) American journal of respiratory and critical care medicine 176(3):261-269; Levine S, et al. (2008) The New England journal of medicine 358(13): 1327-1335).

Despite advances in understanding the physiology and pathophysiology of muscle atrophy, there is still a scarcity of compounds that are both potent, efficacious, and selective modulators of muscle growth and also effective in the treatment of muscle atrophy associated and diseases in which the muscle atrophy or the need to increase muscle mass is involved. These needs and other needs are satisfied by the present invention.

SUMMARY

In accordance with the purpose(s) of the invention, as embodied and broadly described herein, the invention, in one aspect, relates to compounds useful in methods to inhibit muscle atrophy and to increase muscle mass by providing to a subject in need thereof an effective amount of ursolic acid or a derivative thereof, and pharmaceutical compositions comprising compounds used in the methods.

In further aspects, the the purpose(s) of the invention, as embodied and broadly described herein, the invention, in one aspect, relates to compounds useful in methods to modulate muscle growth, methods to inhibit muscle atrophy and to increase muscle mass, methods to induce skeletal muscle hypertrophy, methods to enhance tissue growth, and pharmaceutical compositions comprising compounds used in the methods.

Disclosed are methods for preventing or treating muscle atrophy in an animal, the method comprising administering to the animal a compound of the formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl; or wherein R1a and R1b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R2a and R2b are independently selected from hydrogen and —OR11, provided that at least one of R2a and R2b is —OR11; or wherein R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl; or wherein R3a and R3b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12, and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein each R11 is independently selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount effective to prevent or treat muscle atrophy in the animal, wherein the amount is greater than 1000 per day when the compound is ursolic acid, boswellic acid, corosolic acid, betulinic acid, or UA0713.

Also disclosed are methods for increasing muscle mass and/or muscular strength in an animal, the method comprising administering to the animal a compound of the formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl; or wherein R1a and R1b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R2a and R2b are independently selected from hydrogen and —OR11, provided that at least one of R3a and R3b is —OR11; or wherein R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl; or wherein R3a and R3b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12, and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein each R11 is independently selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount effective to increase muscle mass and/or muscular strength in the animal, wherein the amount is greater than about 1000 mg per day when the compound is ursolic acid, boswellic acid, corosolic acid, betulinic acid, or UA0713.

Also disclosed are methods for enhancing tissue growth in vitro, the method comprising administering to the tissue a compound of the formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl; or wherein R1a and R1b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R2a and R2b are independently selected from hydrogen and —OR11, provided that at least one of R2a and R2b is —OR11; or wherein R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl; or wherein R3a and R3b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12, and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein each R11 is independently selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount effective to enhance growth of the tissue.

Also disclosed are methods for enhancing muscle formation in a mammal, the method comprising administering to the mammal a compound of the formula:

wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount greater than about 1000 mg per day and effective to enhance muscle formation in the mammal.

Also disclosed are methods for testing for performance enhancing use of a ursolic acid analog in an animal, the method comprising: (a) obtaining a biological test sample from the animal; and (b) measuring the amount of a compound of formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in the test sample to determine whether a superphysiological amount of the compound is present in the test sample; wherein the superphysiological amount of the compound in the test sample is indicative of performance enhancing use of the compound.

Also disclosed are pharmaceutical compositions comprising a pharmaceutically acceptable carrier and an effective amount of a compound of the formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl; or wherein R1a and R1b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R2a and R2b are independently selected from hydrogen and —OR11, provided that at least one of R2a and R2b is —OR11; or wherein R2a and R2b together comprise ═O; wherein each of r3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl; or wherein R3a and R3b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12, and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein each R11 is independently selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount effective to prevent or treat muscle atrophy in the animal, wherein the amount is greater than about 1000 mg per day when the compound is ursolic acid, boswellic acid, corosolic acid, betulinic acid, or UA0713.

Also disclosed are kits comprising at least one compound having a structure represented by a formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl; or wherein R1a and R1b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R2a and R2b are independently selected from hydrogen and —OR11, provided that at least one of R2a and R2b is —OR11; or wherein R2a and R2b together comprise ═O; wherein each of R2a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl; or wherein R3a and R3b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12, and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein each R11 is independently selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, and one or more of: (a) a protein supplement; (b) an anabolic agent; (c) a catabolic agent; (d) a dietary supplement; (e) at least one agent known to treat a disorder associated with muscle wasting; (f) instructions for treating a disorder associated with cholinergic activity; or (g) instructions for using the compound to increase muscle mass and/or muscular strength.

Also disclosed are methods for manufacturing a medicament associated with muscle atrophy or the need to increase muscle mass comprising combining at least one disclosed compound or at least one disclosed product with a pharmaceutically acceptable carrier or diluent.

Also disclosed are uses of a disclosed compound or a disclosed product in the manufacture of a medicament for the treatment of a disorder associated with muscle atrophy or the need to increase muscle mass.

While aspects of the present invention can be described and claimed in a particular statutory class, such as the system statutory class, this is for convenience only and one of skill in the art will understand that each aspect of the present invention can be described and claimed in any statutory class.

Unless otherwise expressly stated, it is in no way intended that any method or aspect set forth herein be construed as requiring that its steps be performed in a specific order. Accordingly, where a method claim does not specifically state in the claims or descriptions that the steps are to be limited to a specific order, it is no way intended that an order be inferred, in any respect. This holds for any possible non-express basis for interpretation, including matters of logic with respect to arrangement of steps or operational flow, plain meaning derived from grammatical organization or punctuation, or the number or type of aspects described in the specification.

BRIEF DESCRIPTION OF THE FIGURES

The accompanying figures, which are incorporated in and constitute a part of this specification, illustrate several aspects and together with the description serve to explain the principles of the invention.

FIG. 1 shows a schematic overview of the discovery process leading to a pharmacological compound that promotes skeletal muscle growth and inhibits skeletal muscle atrophy.

FIG. 2 shows representative data on the effect of fasting on skeletal muscle mRNA expression in healthy human adults.

FIG. 3 shows qPCR analysis of representative fasting-responsive mRNAs from human skeletal muscle.

FIG. 4 shows representative data on the identification of ursolic acid as an inhibitor of fasting-induced skeletal muscle atrophy.

FIG. 5 shows representative data on the identification of ursolic acid as an inhibitor of denervation-induced muscle atrophy.

FIG. 6 shows representative data on ursolic acid-mediated induction of muscle hypertrophy.

FIG. 7 shows representative data on the effect of ursolic acid on mouse skeletal muscle specific tetanic force.

FIG. 8 shows representative data on the effect of ursolic acid on muscle growth, atrophic gene expression, trophic gene expression, and skeletal muscle IGF-I signaling.

FIG. 9 shows representative data on the effect of ursolic acid on skeletal muscle expression of IGF1 gene exons, adipose IGF1 mRNA expression, and skeletal muscle insulin signaling.

FIG. 10 shows representative data on the effect of ursolic acid on adiposity.

FIG. 11 shows representative data on the effect of ursolic acid on food consumption, liver weight, kidney weight, and plasma ALT, bilirubin, and creatinine concentrations.

FIG. 12 shows representative data on the effect of ursolic acid on weight gain, white adipose tissue weight, skeletal muscle weight, brown adipose tissue weight and energy expenditure in a mouse model of obesity and metabolic syndrome.

FIG. 13 shows representative data on the effect of ursolic acid on obesity-related pre-diabetes, diabetes, fatty liver disease and hyperlipidemia in a mouse model of obesity and metabolic syndrome.

FIG. 14 shows representative data on the effect of oleanolic acid on skeletal muscle mass.

FIG. 15 shows representative data on the effect of targeted inhibition of PTP1B on skeletal muscle growth.

FIG. 16 shows representative data on the effect of ursolic acid serum concentration on muscle mass and adiposity.

Additional advantages of the invention will be set forth in part in the description which follows, and in part will be obvious from the description, or can be learned by practice of the invention. The advantages of the invention will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.

DESCRIPTION

The present invention can be understood more readily by reference to the following detailed description of the invention and the Examples included therein.

Before the present compounds, compositions, articles, systems, devices, and/or methods are disclosed and described, it is to be understood that they are not limited to specific synthetic methods unless otherwise specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and is not intended to be limiting. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, example methods and materials are now described.

All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided herein can be different from the actual publication dates, which can require independent confirmation.

A. DEFINITIONS

As used herein, nomenclature for compounds, including organic compounds, can be given using common names, IUPAC, IUBMB, or CAS recommendations for nomenclature. When one or more stereochemical features are present, Cahn-Ingold-Prelog rules for stereochemistry can be employed to designate stereochemical priority, E/Z specification, and the like. One of skill in the art can readily ascertain the structure of a compound if given a name, either by systemic reduction of the compound structure using naming conventions, or by commercially available software, such as CHEMDRAW™ (Cambridgesoft Corporation, U.S.A.).

As used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a functional group,” “an alkyl,” or “a residue” includes mixtures of two or more such functional groups, alkyls, or residues, and the like.

Ranges can be expressed herein as from “about” one particular value, and/or to “about” another particular value. When such a range is expressed, a further aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms a further aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10” is also disclosed. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.

References in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed. Thus, in a compound containing 2 parts by weight of component X and 5 parts by weight component Y, X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound.

A weight percent (wt. %) of a component, unless specifically stated to the contrary, is based on the total weight of the formulation or composition in which the component is included.

As used herein, the terms “optional” or “optionally” means that the subsequently described event or circumstance can or can not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.

As used herein, “ursolic acid” refers to ursolic acid, or extracts containing ursolic acid from plants such as apples, holy basil, bilberries, cranberries, elder flower, peppermint, lavender, oregano, thyme, sage, hawthorn, bearberry or prunes.

As used herein, “ursolic acid derivatives” refers to corosolic acid, betulinic acid, hederagenin, boswellic acids, UA0713, a substituted ursolic acid analog, an ursane compound or any other pentacyclic triterpene acids that prevents muscle atrophy, reduces muscle atrophy, increases muscle mass, increases muscle strength in an animal, including in humans, increases Akt phosphorylation, increases S6K phosphorylation, or stimulates biochemical events known to precede or follow Akt phosphorylation or S6K phosphorylation. For example, and not to be limiting, biochemical events known to precede or follow Akt phosphorylation or S6K phosphorylation can be events such as insulin receptor phosphorylation, IGF-I receptor phosphorylation, insulin receptor substrate (IRS) protein phosphorylation, phosphoinositide-3 kinase phosphorylation, phosphoinositide-3 kinase activation, phosphoinositide dependent kinase 1 activation, mammalian target of rapamycin complex 2 activation, adrenergic receptor activation, heterotrimeric G protein activation, adenylate cyclase activation, increased intracellular cyclic AMP, AMP kinase activation, protein kinase A activation, protein kinase C activation, CREB activation, mitogen activated protein kinase pathway activation, mammalian target of rapamycin complex 1 activation, 4E-BP1 phosphorylation, 4E-BP1 inactivation, GSK3β phosphorylation, GSK3β inactivation, increased protein synthesis, increased glucose uptake, Foxo transcription factor phosphorylation, Foxo transcription factor inactivation, Cdkn1a phosphorylation, Cdkn1a inactivation, reduced atrogin-1 mRNA, reduced MuRF1 mRNA, increased VEGFA mRNA, or increased IGF1 mRNA.

As used herein, the term “subject” refers to the target of administration, e.g. an animal. Thus the subject of the herein disclosed methods can be a vertebrate, such as a mammal, a fish, a bird, a reptile, or an amphibian. Alternatively, the subject of the herein disclosed methods can be a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, cat, guinea pig or rodent. The term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered. In one aspect, the subject is a mammal. A patient refers to a subject afflicted with a disease or disorder. The term “patient” includes human and veterinary subjects. In some aspects of the disclosed methods, the subject has been diagnosed with a need for treatment of one or more muscle disorders prior to the administering step. In some aspects of the disclosed method, the subject has been diagnosed with a need for increasing muscle mass prior to the administering step. In some aspects of the disclosed method, the subject has been diagnosed with a need for increasing muscle mass prior to the administering step.

As used herein, the term “treatment” refers to the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder. This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder. In various aspects, the term covers any treatment of a subject, including a mammal (e.g., a human), and includes: (i) preventing the disease from occurring in a subject that can be predisposed to the disease but has not yet been diagnosed as having it; (ii) inhibiting the disease, i.e., arresting its development; or (iii) relieving the disease, i.e., causing regression of the disease. In one aspect, the subject is a mammal such as a primate, and, in a further aspect, the subject is a human. The term “subject” also includes domesticated animals (e.g., cats, dogs, etc.), livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), and laboratory animals (e.g., mouse, rabbit, rat, guinea pig, fruit fly, etc.).

As used herein, the term “prevent” or “preventing” refers to precluding, averting, obviating, forestalling, stopping, or hindering something from happening, especially by advance action. It is understood that where reduce, inhibit or prevent are used herein, unless specifically indicated otherwise, the use of the other two words is also expressly disclosed.

As used herein, the term “diagnosed” means having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition that can be diagnosed or treated by the compounds, compositions, or methods disclosed herein. For example, “diagnosed with a muscle atrophy disorder” means having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition that can be diagnosed or treated by a compound or composition that can increase muscle mass. As a further example, “diagnosed with a need for increasing muscle mass” refers to having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition characterized by muscle atrophy or other disease wherein increasing muscle mass would be beneficial to the subject. Such a diagnosis can be in reference to a disorder, such as muscle atrophy, and the like, as discussed herein.

As used herein, the phrase “identified to be in need of treatment for a disorder,” or the like, refers to selection of a subject based upon need for treatment of the disorder. For example, a subject can be identified as having a need for treatment of a disorder (e.g., a disorder related to muscle atrophy) based upon an earlier diagnosis by a person of skill and thereafter subjected to treatment for the disorder. It is contemplated that the identification can, in one aspect, be performed by a person different from the person making the diagnosis. It is also contemplated, in a further aspect, that the administration can be performed by one who subsequently performed the administration.

As used herein, the terms “administering” and “administration” refer to any method of providing a pharmaceutical preparation to a subject. Such methods are well known to those skilled in the art and include, but are not limited to, oral administration, transdermal administration, administration by inhalation, nasal administration, topical administration, intravaginal administration, ophthalmic administration, intraaural administration, intracerebral administration, rectal administration, sublingual administration, buccal administration, and parenteral administration, including injectable such as intravenous administration, intra-arterial administration, intramuscular administration, and subcutaneous administration. Administration can be continuous or intermittent. In various aspects, a preparation can be administered therapeutically; that is, administered to treat an existing disease or condition. In further various aspects, a preparation can be administered prophylactically; that is, administered for prevention of a disease or condition.

The term “contacting” as used herein refers to bringing a disclosed compound and a cell, target receptor, or other biological entity together in such a manner that the compound can affect the activity of the target (e.g., receptor, transcription factor, cell, etc.), either directly; i.e., by interacting with the target itself, or indirectly; i.e., by interacting with another molecule, co-factor, factor, or protein on which the activity of the target is dependent.

As used herein, the terms “effective amount” and “amount effective” refer to an amount that is sufficient to achieve the desired result or to have an effect on an undesired condition. For example, a “therapeutically effective amount” refers to an amount that is sufficient to achieve the desired therapeutic result or to have an effect on undesired symptoms, but is generally insufficient to cause adverse side affects. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration; the route of administration; the rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of a compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. If desired, the effective daily dose can be divided into multiple doses for purposes of administration. Consequently, single dose compositions can contain such amounts or submultiples thereof to make up the daily dose. The dosage can be adjusted by the individual physician in the event of any contraindications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days. Guidance can be found in the literature for appropriate dosages for given classes of pharmaceutical products. In further various aspects, a preparation can be administered in a “prophylactically effective amount”; that is, an amount effective for prevention of a disease or condition.

As used herein, “EC50,” is intended to refer to the concentration or dose of a substance (e.g., a compound or a drug) that is required for 50% enhancement or activation of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc. EC50 also refers to the concentration or dose of a substance that is required for 50% enhancement or activation in vivo, as further defined elsewhere herein. Alternatively, EC50 can refer to the concentration or dose of compound that provokes a response halfway between the baseline and maximum response. The response can be measusred in a in vitro or in vivo system as is convenient and appropriate for the biological response of interest. For example, the response can be measured in vitro using cultured muscle cells or in an ex vivo organ culture system with isolated muscle fibers. Alternatively, the response can be measured in vivo using an appropriate research model such as rodent, including mice and rats. The mouse or rat can be an inbred strain with phenotypic characteristics of interest such as obesity or diabetes. As appropriate, the response can be measured in a transgenic or knockout mouse or rat wherein the a gene or genes has been introduced or knocked-out, as appropriate, to replicate a disease process.

As used herein, “IC50,” is intended to refer to the concentration or dose of a substance (e.g., a compound or a drug) that is required for 50% inhibition or diminuation of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc. IC50 also refers to the concentration or dose of a substance that is required for 50% inhibition or diminuation in vivo, as further defined elsewhere herein. Alternatively, IC50 also refers refers to the half maximal (50%) inhibitory concentration (IC) or inhibitory dose of a substance. The response can be measusred in a in vitro or in vivo system as is convenient and appropriate for the biological response of interest. For example, the response can be measured in vitro using cultured muscle cells or in an ex vivo organ culture system with isolated muscle fibers. Alternatively, the response can be measured in vivo using an appropriate research model such as rodent, including mice and rats. The mouse or rat can be an inbred strain with phenotypic characteristics of interest such as obesity or diabetes. As appropriate, the response can be measured in a transgenic or knockout mouse or rat wherein the a gene or genes has been introduced or knocked-out, as appropriate, to replicate a disease process.

The term “pharmaceutically acceptable” describes a material that is not biologically or otherwise undesirable, i.e., without causing an unacceptable level of undesirable biological effects or interacting in a deleterious manner.

As used herein, the term “derivative” refers to a compound having a structure derived from the structure of a parent compound (e.g., a compound disclosed herein) and whose structure is sufficiently similar to those disclosed herein and based upon that similarity, would be expected by one skilled in the art to exhibit the same or similar activities and utilities as the claimed compounds, or to induce, as a precursor, the same or similar activities and utilities as the claimed compounds. Exemplary derivatives include salts, esters, amides, salts of esters or amides, and N-oxides of a parent compound.

As used herein, the term “pharmaceutically acceptable carrier” refers to sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), carboxymethylcellulose and suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. These compositions can also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms can be ensured by the inclusion of various antibacterial and antifungal agents such as paraben, chlorobutanol, phenol, sorbic acid and the like. It can also be desirable to include isotonic agents such as sugars, sodium chloride and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the inclusion of agents, such as aluminum monostearate and gelatin, which delay absorption. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide, poly(orthoesters) and poly(anhydrides). Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues. The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable media just prior to use. Suitable inert carriers can include sugars such as lactose. Desirably, at least 95% by weight of the particles of the active ingredient have an effective particle size in the range of 0.01 to 10 micrometers.

A residue of a chemical species, as used in the specification and concluding claims, refers to the moiety that is the resulting product of the chemical species in a particular reaction scheme or subsequent formulation or chemical product, regardless of whether the moiety is actually obtained from the chemical species. Thus, an ethylene glycol residue in a polyester refers to one or more —OCH2CH2O— units in the polyester, regardless of whether ethylene glycol was used to prepare the polyester. Similarly, a sebacic acid residue in a polyester refers to one or more —CO(CH2)8CO— moieties in the polyester, regardless of whether the residue is obtained by reacting sebacic acid or an ester thereof to obtain the polyester.

As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described below. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms, such as nitrogen, can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This disclosure is not intended to be limited in any manner by the permissible substituents of organic compounds. Also, the terms “substitution” or “substituted with” include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. It is also contemplated that, in certain aspects, unless expressly indicated to the contrary, individual substituents can be further optionally substituted (i.e., further substituted or unsubstituted).

In defining various terms, “A1,” “A2,” “A3,” and “A4” are used herein as generic symbols to represent various specific substituents. These symbols can be any substituent, not limited to those disclosed herein, and when they are defined to be certain substituents in one instance, they can, in another instance, be defined as some other substituents.

The term “alkyl” as used herein is a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, s-butyl, t-butyl, n-pentyl, isopentyl, s-pentyl, neopentyl, hexyl, heptyl, octyl, nonyl, decyl, dode cyl, tetradecyl, hexadecyl, eicosyl, tetracosyl, and the like. The alkyl group can be cyclic or acyclic. The alkyl group can be branched or unbranched. The alkyl group can also be substituted or unsubstituted. For example, the alkyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, amino, ether, halide, hydroxy, nitro, silyl, sulfo-oxo, or thiol, as described herein. A “lower alkyl” group is an alkyl group containing from one to six (e.g., from one to four) carbon atoms.

Throughout the specification “alkyl” is generally used to refer to both unsubstituted alkyl groups and substituted alkyl groups; however, substituted alkyl groups are also specifically referred to herein by identifying the specific substituent(s) on the alkyl group. For example, the term “halogenated alkyl” or “haloalkyl” specifically refers to an alkyl group that is substituted with one or more halide, e.g., fluorine, chlorine, bromine, or iodine. The term “alkoxyalkyl” specifically refers to an alkyl group that is substituted with one or more alkoxy groups, as described below. The term “alkylamino” specifically refers to an alkyl group that is substituted with one or more amino groups, as described below, and the like. When “alkyl” is used in one instance and a specific term such as “alkylalcohol” is used in another, it is not meant to imply that the term “alkyl” does not also refer to specific terms such as “alkylalcohol” and the like.

This practice is also used for other groups described herein. That is, while a term such as “cycloalkyl” refers to both unsubstituted and substituted cycloalkyl moieties, the substituted moieties can, in addition, be specifically identified herein; for example, a particular substituted cycloalkyl can be referred to as, e.g., an “alkylcycloalkyl.” Similarly, a substituted alkoxy can be specifically referred to as, e.g., a “halogenated alkoxy,” a particular substituted alkenyl can be, e.g., an “alkenylalcohol,” and the like. Again, the practice of using a general term, such as “cycloalkyl,” and a specific term, such as “alkylcycloalkyl,” is not meant to imply that the general term does not also include the specific term.

The term “cycloalkyl” as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl, and the like. The term “heterocycloalkyl” is a type of cycloalkyl group as defined above, and is included within the meaning of the term “cycloalkyl,” where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted. The cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, amino, ether, halide, hydroxy, nitro, silyl, sulfo-oxo, or thiol as described herein.

The term “polyalkylene group” as used herein is a group having two or more CH2 groups linked to one another. The polyalkylene group can be represented by the formula —(CH2)a—, where “a” is an integer of from 2 to 500.

The terms “alkoxy” and “alkoxyl” as used herein to refer to an alkyl or cycloalkyl group bonded through an ether linkage; that is, an “alkoxy” group can be defined as —OA1 where A1 is alkyl or cycloalkyl as defined above. “Alkoxy” also includes polymers of alkoxy groups as just described; that is, an alkoxy can be a polyether such as —OA1-OA2 or —OA1-(OA2)a-OA3, where “a” is an integer of from 1 to 200 and A1, A2, and A3 are alkyl and/or cycloalkyl groups.

The term “alkenyl” as used herein is a hydrocarbon group of from 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon double bond.

Asymmetric structures such as (A1A2)C═C(A3A4) are intended to include both the E and Z isomers. This can be presumed in structural formulae herein wherein an asymmetric alkene is present, or it can be explicitly indicated by the bond symbol C═C. The alkenyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol, as described herein.

The term “cycloalkenyl” as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms and containing at least one carbon-carbon double bound, i.e., C═C. Examples of cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, norbornenyl, and the like. The term “heterocycloalkenyl” is a type of cycloalkenyl group as defined above, and is included within the meaning of the term “cycloalkenyl,” where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkenyl group and heterocycloalkenyl group can be substituted or unsubstituted. The cycloalkenyl group and heterocycloalkenyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein.

The term “alkynyl” as used herein is a hydrocarbon group of 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon triple bond. The alkynyl group can be unsubstituted or substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol, as described herein.

The term “cycloalkynyl” as used herein is a non-aromatic carbon-based ring composed of at least seven carbon atoms and containing at least one carbon-carbon triple bound. Examples of cycloalkynyl groups include, but are not limited to, cycloheptynyl, cyclooctynyl, cyclononynyl, and the like. The term “heterocycloalkynyl” is a type of cycloalkenyl group as defined above, and is included within the meaning of the term “cycloalkynyl,” where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkynyl group and heterocycloalkynyl group can be substituted or unsubstituted. The cycloalkynyl group and heterocycloalkynyl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein.

The term “aryl” as used herein is a group that contains any carbon-based aromatic group including, but not limited to, benzene, naphthalene, phenyl, biphenyl, phenoxybenzene, and the like. The term “aryl” also includes “heteroaryl,” which is defined as a group that contains an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorus. Likewise, the term “non-heteroaryl,” which is also included in the term “aryl,” defines a group that contains an aromatic group that does not contain a heteroatom. The aryl group can be substituted or unsubstituted. The aryl group can be substituted with one or more groups including, but not limited to, alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein. The term “biaryl” is a specific type of aryl group and is included in the definition of “aryl.” Biaryl refers to two aryl groups that are bound together via a fused ring structure, as in naphthalene, or are attached via one or more carbon-carbon bonds, as in biphenyl.

The term “aldehyde” as used herein is represented by the formula —C(O)H. Throughout this specification “C(O)” is a short hand notation for a carbonyl group, i.e., C═O.

The terms “amine” or “amino” as used herein are represented by the formula —NA1A2, where A1 and A2 can be, independently, hydrogen or alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein.

The term “alkylamino” as used herein is represented by the formula —NH(-alkyl) where alkyl is a described herein. Representative examples include, but are not limited to, methylamino group, ethylamino group, propylamino group, isopropylamino group, butylamino group, isobutylamino group, (sec-butyl)amino group, (tert-butyl)amino group, pentylamino group, isopentylamino group, (tert-pentyl)amino group, hexylamino group, and the like.

The term “dialkylamino” as used herein is represented by the formula —N(-alkyl)2 where alkyl is a described herein. Representative examples include, but are not limited to, dimethylamino group, diethylamino group, dipropylamino group, diisopropylamino group, dibutylamino group, diisobutylamino group, di(sec-butyl)amino group, di(tert-butyl)amino group, dipentylamino group, diisopentylamino group, di(tert-pentyl)amino group, dihexylamino group, N-ethyl-N-methylamino group, N-methyl-N-propylamino group, N-ethyl-N-propylamino group and the like.

The term “carboxylic acid” as used herein is represented by the formula —C(O)OH.

The term “ester” as used herein is represented by the formula —OC(O)A1 or —C(O)OA1, where A1 can be alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “polyester” as used herein is represented by the formula -(A1O(O)C-A2-C(O)O)a— or -(A1O(O)C-A2-OC(O))a—, where A1 and A2 can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein and “a” is an interger from 1 to 500. “Polyester” is as the term used to describe a group that is produced by the reaction between a compound having at least two carboxylic acid groups with a compound having at least two hydroxyl groups.

The term “ether” as used herein is represented by the formula A1OA2, where A1 and A2 can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein. The term “polyether” as used herein is represented by the formula —(A1O-A2O)a—, where A1 and A2 can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein and “a” is an integer of from 1 to 500. Examples of polyether groups include polyethylene oxide, polypropylene oxide, and polybutylene oxide.

The term “halide” as used herein refers to the halogens fluorine, chlorine, bromine, and iodine.

The term “heterocycle,” as used herein refers to single and multi-cyclic aromatic or non-aromatic ring systems in which at least one of the ring members is other than carbon. Heterocycle includes azetidine, dioxane, furan, imidazole, isothiazole, isoxazole, morpholine, oxazole, oxazole, including, 1,2,3-oxadiazole, 1,2,5-oxadiazole and 1,3,4-oxadiazole, piperazine, piperidine, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolidine, tetrahydrofuran, tetrahydropyran, tetrazine, including 1,2,4,5-tetrazine, tetrazole, including 1,2,3,4-tetrazole and 1,2,4,5-tetrazole, thiadiazole, including, 1,2,3-thiadiazole, 1,2,5-thiadiazole, and 1,3,4-thiadiazole, thiazole, thiophene, triazine, including 1,3,5-triazine and 1,2,4-triazine, triazole, including, 1,2,3-triazole, 1,3,4-triazole, and the like.

The term “hydroxyl” as used herein is represented by the formula —OH.

The term “ketone” as used herein is represented by the formula A1C(O)A2, where A1 and A2 can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein.

The term “azide” as used herein is represented by the formula —N3.

The term “nitro” as used herein is represented by the formula —NO2.

The term “nitrile” as used herein is represented by the formula —CN.

The term “silyl” as used herein is represented by the formula —SiA1A2A3, where A1, A2, and A3 can be, independently, hydrogen or an alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein.

The term “sulfo-oxo” as used herein is represented by the formulas —S(O)A1, —S(O)2A1, —OS(O)2A1, or —OS(O)2OA1, where A1 can be hydrogen or an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. Throughout this specification “S(O)” is a short hand notation for S═O. The term “sulfonyl” is used herein to refer to the sulfo-oxo group represented by the formula —S(O)2A1, where A1 can be hydrogen or an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “sulfone” as used herein is represented by the formula A1S(O)2A2, where A1 and A2 can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term “sulfoxide” as used herein is represented by the formula A1S(O)A2, where A1 and A2 can be, independently, an alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein.

The term “thiol” as used herein is represented by the formula —SH.

“R1,” “R2,” “R3,” “Rn,” where n is an integer, as used herein can, independently, possess one or more of the groups listed above. For example, if R1 is a straight chain alkyl group, one of the hydrogen atoms of the alkyl group can optionally be substituted with a hydroxyl group, an alkoxy group, an alkyl group, a halide, and the like. Depending upon the groups that are selected, a first group can be incorporated within second group or, alternatively, the first group can be pendant (i.e., attached) to the second group. For example, with the phrase “an alkyl group comprising an amino group,” the amino group can be incorporated within the backbone of the alkyl group. Alternatively, the amino group can be attached to the backbone of the alkyl group. The nature of the group(s) that is (are) selected will determine if the first group is embedded or attached to the second group.

As described herein, compounds of the invention may contain “optionally substituted” moieties. In general, the term “substituted,” whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. In is also contemplated that, in certain aspects, unless expressly indicated to the contrary, individual substituents can be further optionally substituted (i.e., further substituted or unsubstituted).

The term “stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain aspects, their recovery, purification, and use for one or more of the purposes disclosed herein.

Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted” group are independently halogen; —(CH2)0-4R; —(CH2)0-4OR; —O(CH2)0-4R, —O—(CH2)0-4C(O)OR; —(CH2)0-4CH(OR)2; —(CH2)0-4SR; —(CH2)0-4Ph, which may be substituted with R; —(CH2)0-4O(CH2)0-1Ph which may be substituted with R; —CH═CHPh, which may be substituted with R; —(CH2)0-4O(CH2)0-1-pyridyl which may be substituted with R; —NO2; —CN; —N3; —(CH2)0-4N(R)2; —(CH2)0-4N(R)C(O)R; —N(R)C(S)R; —(CH2)0- 4N(R)C(O)NR2; —N(R)C(S)NR2; —(CH2)0-4N(R)C(O)OR; —N(R)N(R)C(O)R; —N(R)N(R)C(O)NR2; —N(R)N(R)C(O)OR; —(CH2)0-4C(O)R; —C(S)R; —(CH2)0-4C(O)OR; —(CH2)0-4C(O)SR; —(CH2)0-4C(O)OSiR3; —(CH2)0-4OC(O)R; —OC(O)(CH2)0-4SR—, SC(S)SR; —(CH2)0-4SC(O)R; —(CH2)0-4C(O)NR2; —C(S)NR2; —C(S)SR; —SC(S)SR, —(CH2)0-4OC(O)NR2; —C(O)N(OR)R; —C(O)C(O)R; —C(O)CH2C(O)R; —C(NOR)R; —(CH2)0-4SSR; —(CH2)0-4S(O)2R; —(CH2)0-4S(O)2OR; —(CH2)0-4OS(O)2R; —S(O)2NR2; —(CH2)0-4S(O)R; —N(RS(O)2NR2; —N(R)S(O)2R; —N(OR)R; —C(NH)NR2; —P(O)2R; —P(O)R2; —OP(O)R2; —OP(O)(OR)2; SiR3; —(C1-4 straight or branched)alkylene)O—N(R)2; or —(C1-4 straight or branched alkylene)C(O)O—N(R)2, wherein each R may be substituted as defined below and is independently hydrogen, C1-6 aliphatic, —CH2Ph, —O(CH2)0-1Ph, —CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.

Suitable monovalent substituents on R (or the ring formed by taking two independent occurrences of R together with their intervening atoms), are independently halogen, —(CH2)0-2R, -(haloR), —(CH2)0-2OH, —(CH2)0-2OR, —(CH2)0-2CH(OR)2; —O(haloR), —CN, —N3, —(CH2)0-2C(O)R, —(CH2)0-2C(O)OH, —(CH2)0-2C(O)OR, —(CH2)0-2SR, —(CH2)0-2SH, —(CH2)0-2NH2, —(CH2)0-2NHR, —(CH2)0-2NR2, —NO2, —SiR3, —OSiR3, —C(O)SR, —(C1- 4 straight or branched alkylene)C(O)OR, or —SSR wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently selected from C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R include ═O and ═S.

Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: ═O, ═S, ═NNR*2, ═NNHC(O)R*, ═NNHC(O)OR*, ═NNHS(O)2R*, ═NR*, ═NOR*, —O(C(R*2))2-3O—, or —S(C(R*2))2-3S—, wherein each independent occurrence of R* is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: —O(CR*2)2-3O—, wherein each independent occurrence of R* is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on the aliphatic group of R* include halogen, —R, -(haloR), —OH, —OR, —O(haloR), —CN, —C(O)OH, —C(O)OR, —NH2, —NHR, —NR2, or —NO2, wherein each r is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include —R, —NR2, —C(O)R, —C(O)OR, —C(O)C(O)R, —C(O)CH2C(O)R, —S(O)2R, —S(O)2NR2, —C(S)NR2, —C(NH)NR2, or —N(R)S(O)2R; wherein each R is independently hydrogen, C1-6 aliphatic which may be substituted as defined below, unsubstituted —OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Suitable substituents on the aliphatic group of R are independently halogen, —R, -(haloR), —OH, —OR, —O(haloR), —CN, —C(O)OH, —C(O)OR, —NH2, —NHR, —NR2, or —NO2, wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

The term “leaving group” refers to an atom (or a group of atoms) with electron withdrawing ability that can be displaced as a stable species, taking with it the bonding electrons. Examples of suitable leaving groups include halides and sulfonate esters, including, but not limited to, triflate, mesylate, tosylate, brosylate, and halides.

The terms “hydrolysable group” and “hydrolysable moiety” refer to a functional group capable of undergoing hydrolysis, e.g., under basic or acidic conditions. Examples of hydrolysable residues include, without limitatation, acid halides, activated carboxylic acids, and various protecting groups known in the art (see, for example, “Protective Groups in Organic Synthesis,” T. W. Greene, P. G. M. Wuts, Wiley-Interscience, 1999).

The term “organic residue” defines a carbon containing residue, i.e., a residue comprising at least one carbon atom, and includes but is not limited to the carbon-containing groups, residues, or radicals defined hereinabove. Organic residues can contain various heteroatoms, or be bonded to another molecule through a heteroatom, including oxygen, nitrogen, sulfur, phosphorus, or the like. Examples of organic residues include but are not limited alkyl or substituted alkyls, alkoxy or substituted alkoxy, mono or di-substituted amino, amide groups, etc. Organic residues can preferably comprise 1 to 18 carbon atoms, 1 to 15, carbon atoms, 1 to 12 carbon atoms, 1 to 8 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms. In a further aspect, an organic residue can comprise 2 to 18 carbon atoms, 2 to 15, carbon atoms, 2 to 12 carbon atoms, 2 to 8 carbon atoms, 2 to 4 carbon atoms, or 2 to 4 carbon atoms.

A very close synonym of the term “residue” is the term “radical,” which as used in the specification and concluding claims, refers to a fragment, group, or substructure of a molecule described herein, regardless of how the molecule is prepared. For example, a 2,4-thiazolidinedione radical in a particular compound has the structure

regardless of whether thiazolidinedione is used to prepare the compound. In some embodiments the radical (for example an alkyl) can be further modified (i.e., substituted alkyl) by having bonded thereto one or more “substituent radicals.” The number of atoms in a given radical is not critical to the present invention unless it is indicated to the contrary elsewhere herein.

“Organic radicals,” as the term is defined and used herein, contain one or more carbon atoms. An organic radical can have, for example, 1-26 carbon atoms, 1-18 carbon atoms, 1-12 carbon atoms, 1-8 carbon atoms, 1-6 carbon atoms, or 1-4 carbon atoms. In a further aspect, an organic radical can have 2-26 carbon atoms, 2-18 carbon atoms, 2-12 carbon atoms, 2-8 carbon atoms, 2-6 carbon atoms, or 2-4 carbon atoms. Organic radicals often have hydrogen bound to at least some of the carbon atoms of the organic radical. One example, of an organic radical that comprises no inorganic atoms is a 5, 6, 7, 8-tetrahydro-2-naphthyl radical. In some embodiments, an organic radical can contain 1-10 inorganic heteroatoms bound thereto or therein, including halogens, oxygen, sulfur, nitrogen, phosphorus, and the like. Examples of organic radicals include but are not limited to an alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, mono-substituted amino, di-substituted amino, acyloxy, cyano, carboxy, carboalkoxy, alkylcarboxamide, substituted alkylcarboxamide, dialkylcarboxamide, substituted dialkylcarboxamide, alkylsulfonyl, alkylsulfinyl, thioalkyl, thiohaloalkyl, alkoxy, substituted alkoxy, haloalkyl, haloalkoxy, aryl, substituted aryl, heteroaryl, heterocyclic, or substituted heterocyclic radicals, wherein the terms are defined elsewhere herein. A few non-limiting examples of organic radicals that include heteroatoms include alkoxy radicals, trifluoromethoxy radicals, acetoxy radicals, dimethylamino radicals and the like.

“Inorganic radicals,” as the term is defined and used herein, contain no carbon atoms and therefore comprise only atoms other than carbon. Inorganic radicals comprise bonded combinations of atoms selected from hydrogen, nitrogen, oxygen, silicon, phosphorus, sulfur, selenium, and halogens such as fluorine, chlorine, bromine, and iodine, which can be present individually or bonded together in their chemically stable combinations. Inorganic radicals have 10 or fewer, or preferably one to six or one to four inorganic atoms as listed above bonded together. Examples of inorganic radicals include, but not limited to, amino, hydroxy, halogens, nitro, thiol, sulfate, phosphate, and like commonly known inorganic radicals. The inorganic radicals do not have bonded therein the metallic elements of the periodic table (such as the alkali metals, alkaline earth metals, transition metals, lanthanide metals, or actinide metals), although such metal ions can sometimes serve as a pharmaceutically acceptable cation for anionic inorganic radicals such as a sulfate, phosphate, or like anionic inorganic radical. Inorganic radicals do not comprise metalloids elements such as boron, aluminum, gallium, germanium, arsenic, tin, lead, or tellurium, or the noble gas elements, unless otherwise specifically indicated elsewhere herein.

Compounds described herein can contain one or more double bonds and, thus, potentially give rise to cis/trans (E/Z) isomers, as well as other conformational isomers. Unless stated to the contrary, the invention includes all such possible isomers, as well as mixtures of such isomers.

Unless stated to the contrary, a formula with chemical bonds shown only as solid lines and not as wedges or dashed lines contemplates each possible isomer, e.g., each enantiomer and diastereomer, and a mixture of isomers, such as a racemic or scalemic mixture. Compounds described herein can contain one or more asymmetric centers and, thus, potentially give rise to diastereomers and optical isomers. Unless stated to the contrary, the present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. Mixtures of stereoisomers, as well as isolated specific stereoisomers, are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.

Many organic compounds exist in optically active forms having the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L or R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and 1 or (+) and (−) are employed to designate the sign of rotation of plane-polarized light by the compound, with (−) or meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these compounds, called stereoisomers, are identical except that they are non-superimposable mirror images of one another. A specific stereoisomer can also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture. Many of the compounds described herein can have one or more chiral centers and therefore can exist in different enantiomeric forms. If desired, a chiral carbon can be designated with an asterisk (*). When bonds to the chiral carbon are depicted as straight lines in the disclosed formulas, it is understood that both the (R) and (S) configurations of the chiral carbon, and hence both enantiomers and mixtures thereof, are embraced within the formula. As is used in the art, when it is desired to specify the absolute configuration about a chiral carbon, one of the bonds to the chiral carbon can be depicted as a wedge (bonds to atoms above the plane) and the other can be depicted as a series or wedge of short parallel lines is (bonds to atoms below the plane). The Cahn-Inglod-Prelog system can be used to assign the (R) or (S) configuration to a chiral carbon.

Compounds described herein comprise atoms in both their natural isotopic abundance and in non-natural abundance. The disclosed compounds can be isotopically-labelled or isotopically-substituted compounds identical to those described, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 2H, 3H, 13C, 14C, 15N, 18O, 17O, 35S, 18F and 36Cl, respectively. Compounds further comprise prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labelled compounds of the present invention, for example those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability.

Further, substitution with heavier isotopes such as deuterium, i.e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labelled compounds of the present invention and prodrugs thereof can generally be prepared by carrying out the procedures below, by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.

The compounds described in the invention can be present as a solvate. In some cases, the solvent used to prepare the solvate is an aqueous solution, and the solvate is then often referred to as a hydrate. The compounds can be present as a hydrate, which can be obtained, for example, by crystallization from a solvent or from aqueous solution. In this connection, one, two, three or any arbitrary number of solvate or water molecules can combine with the compounds according to the invention to form solvates and hydrates. Unless stated to the contrary, the invention includes all such possible solvates.

The term “co-crystal” means a physical association of two or more molecules which owe their stability through non-covalent interaction. One or more components of this molecular complex provide a stable framework in the crystalline lattice. In certain instances, the guest molecules are incorporated in the crystalline lattice as anhydrates or solvates, see e.g. “Crystal Engineering of the Composition of Pharmaceutical Phases. Do Pharmaceutical Co-crystals Represent a New Path to Improved Medicines?” Almarasson, O., et. al., The Royal Society of Chemistry, 1889-1896, 2004. Examples of co-crystals include p-toluenesulfonic acid and benzenesulfonic acid.

It is also appreciated that certain compounds described herein can be present as an equilibrium of tautomers. For example, ketones with an a-hydrogen can exist in an equilibrium of the keto form and the enol form.

Likewise, amides with an N-hydrogen can exist in an equilibrium of the amide form and the imidic acid form. Unless stated to the contrary, the invention includes all such possible tautomers.

It is known that chemical substances form solids which are present in different states of order which are termed polymorphic forms or modifications. The different modifications of a polymorphic substance can differ greatly in their physical properties. The compounds according to the invention can be present in different polymorphic forms, with it being possible for particular modifications to be metastable. Unless stated to the contrary, the invention includes all such possible polymorphic forms.

In some aspects, a structure of a compound can be represented by a formula:

which is understood to be equivalent to a formula:

wherein n is typically an integer. That is, Rn is understood to represent five independent substituents, Rn(a), Rn(b), Rn(c), Rn(d), Rn(e). By “independent substituents,” it is meant that each R substituent can be independently defined. For example, if in one instance Rn(a) is halogen, then Rn(b) is not necessarily halogen in that instance.

Certain materials, compounds, compositions, and components disclosed herein can be obtained commercially or readily synthesized using techniques generally known to those of skill in the art. For example, the starting materials and reagents used in preparing the disclosed compounds and compositions are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Acros Organics (Morris Plains, N.J.), Fisher Scientific (Pittsburgh, Pa.), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991); March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition); and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989).

Unless otherwise expressly stated, it is in no way intended that any method set forth herein be construed as requiring that its steps be performed in a specific order. Accordingly, where a method claim does not actually recite an order to be followed by its steps or it is not otherwise specifically stated in the claims or descriptions that the steps are to be limited to a specific order, it is no way intended that an order be inferred, in any respect. This holds for any possible non-express basis for interpretation, including: matters of logic with respect to arrangement of steps or operational flow; plain meaning derived from grammatical organization or punctuation; and the number or type of embodiments described in the specification.

Disclosed are the components to be used to prepare the compositions of the invention as well as the compositions themselves to be used within the methods disclosed herein. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds can not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular compound is disclosed and discussed and a number of modifications that can be made to a number of molecules including the compounds are discussed, specifically contemplated is each and every combination and permutation of the compound and the modifications that are possible unless specifically indicated to the contrary. Thus, if a class of molecules A, B, and C are disclosed as well as a class of molecules D, E, and F and an example of a combination molecule, A-D is disclosed, then even if each is not individually recited each is individually and collectively contemplated meaning combinations, A-E, A-F, B-D, B-E, B-F, C-D, C-E, and C-F are considered disclosed. Likewise, any subset or combination of these is also disclosed. Thus, for example, the sub-group of A-E, B-F, and C-E would be considered disclosed. This concept applies to all aspects of this application including, but not limited to, steps in methods of making and using the compositions of the invention. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the methods of the invention.

It is understood that the compositions disclosed herein have certain functions. Disclosed herein are certain structural requirements for performing the disclosed functions, and it is understood that there are a variety of structures that can perform the same function that are related to the disclosed structures, and that these structures will typically achieve the same result.

B. COMPOUNDS

In one aspect, the invention relates to compounds useful in methods to inhibit muscle atrophy and to increase muscle mass by providing to a subject in need thereof an effective amount of ursolic acid or a derivative thereof, and pharmaceutical compositions comprising compounds used in the methods. In a further aspect, the invention relates to compounds useful in methods to modulate muscle growth, methods to inhibit muscle atrophy and to increase muscle mass, methods to induce skeletal muscle hypertrophy, methods to enhance tissue growth, and pharmaceutical compositions comprising compounds used in the methods.

In one aspect, the compounds of the invention are useful in the treatment of muscle disorders. In a further aspect, the muscle disorder can be skeletal muscle atrophy secondary to malnutrition, muscle disuse (secondary to voluntary or involuntary bedrest), neurologic disease (including multiple sclerosis, amyotrophic lateral sclerosis, spinal muscular atrophy, critical illness neuropathy, spinal cord injury or peripheral nerve injury), orthopedic injury, casting, and other post-surgical forms of limb immobilization, chronic disease (including cancer, congestive heart failure, chronic pulmonary disease, chronic renal failure, chronic liver disease, diabetes mellitus, Cushing syndrome and chronic infections such as HIV/AIDS or tuberculosis), burns, sepsis, other illnesses requiring mechanical ventiliation, drug-induced muscle disease (such as glucorticoid-induced myopathy and statin-induced myopathy), genetic diseases that primarily affect skeletal muscle (such as muscular dystrophy and myotonic dystrophy), autoimmune diseases that affect skeletal muscle (such as polymyositis and dermatomyositis), spaceflight, or age-related sarcopenia.

It is contemplated that each disclosed derivative can be optionally further substituted. It is also contemplated that any one or more derivative can be optionally omitted from the invention. It is understood that a disclosed compound can be provided by the disclosed methods. It is also understood that the disclosed compounds can be employed in the disclosed methods of using.

1. Ursolic Acid Derivatives

Ursolic acid is a highly water-insoluble pentacyclic triterpene acid that possesses a wide range of biological effects, including anti-cancer, anti-oxidant, anti-inflammatory, anti-allergic, hepatoprotective, gastroprotective, hypolipidemic, hypoglycemic, lipolytic anti-obesity, anti-atherogenic and immunomodulatory effects (Liu J (1995) Journal of ethnopharmacology 49(2):57-68; Liu J (2005) Journal of ethnopharmacology 100(1-2):92-94; Wang Z H, et al. (2010) European journal of pharmacology 628(1-3):255-260; Jang S M, et al. (2009) Int Immunopharmacol 9(1):113-119). However, its effects on skeletal muscle were not known previously. At the molecular level, ursolic acid inhibits the STAT3 activation pathway, reduces matrix metalloproteinase-9 expression via the glucocorticoid receptor, inhibits protein tyrosine phosphatases, acts as an insulin mimetic, activates PPARα, inhibits NF-kB transcription factors, translocates hormone-senstive lipase to stimulate lipolysis and inhibits the hepatic polyol pathway, among many other described effects. Its effects on skeletal muscle and IGF-I signaling were not previously known.

As medicine, ursolic acid is well tolerated and can be used topically and orally. Ursolic acid is present in many plants, including apples, basil, bilberries, cranberries, elder flower, peppermint, rosemary, lavender, oregano, thyme, hawthorn, prunes. Apple peels contain high quantity of ursolic acid and related compounds which are responsible for the anti-cancer activity of apple. Ursolic acid can also serve as a starting material for synthesis of more potent bioactive derivatives, such as anti-tumor agents.

Other names for ursolic acid include 3-β-hydroxy-urs-12-en-28-oic acid, urson, prunol, micromerol, and malol. The structure is shown below:

Other closely related pentacyclic triterpene acids with insulin sensitizing actions include oleanolic acid (Wang et at, 2010), corosolic acid (Sivakumar et at, 2009) and UA0713 (Zhang et at, 2006).

In one aspect, the invention relates to compounds of the formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl, or wherein R1a and R1b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen, or R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl, wherein R3a and R3b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12 and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof

In a further aspect, the invention relates to compounds of a formula:

wherein each of R1a and R1b is C1-C6 alkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen; wherein each of R4, R5, and R6 is independently C1-C6 alkyl; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein R9b is C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl.

In a further aspect, the invention relates to compounds of a formula:

wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl, or R1a and R1b are covalently bonded and, together with the intermediate carbon, comprise an optionally substituted 3- to 7-membered spirocycloalkyl; wherein R8 is C1-C6 alkyl; wherein R9a is C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl.

In a further aspect, the invention relates to compounds of a formula:

In a further aspect, the invention relates to compounds of a formula:

wherein R1a is —C(O)ZR10; wherein R1b is C1-C6 alkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein R9a is selected from hydrogen and C1-C6 alkyl; wherein Z is selected from —O— and —NR13—; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl.

In a further aspect, the invention relates to compounds of a formula:

wherein each of R1a and R1b is independently C1-C6 alkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen; wherein one of R3a and R3b is —OR11, and the other is hydrogen; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R8 is C1-C6 alkyl; wherein R9a is C1-C6 alkyl; wherein each R11 is independently selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl.

In a further aspect, the invention relates to compounds of a formula:

wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof.

In a further aspect, the compound is administered in an amount effective to prevent or treat muscle atrophy in the animal. In a still further aspect, the compound is administered in amount is greater than about 1000 mg per day when the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In a yet further aspect, the compound is administered in an amount greater than about 1000 mg per day and effective to enhance muscle formation in the mammal.

In a further aspect, the invention relates to compounds of a formula selected from:

a. R0 Groups and Optional Bonds

In one aspect, an optional covalent bond can be represented by —. Thus, in certain aspects, a particular bond is present, thereby providing a single covalent bond. In further aspects, a particular bond is present, thereby providing a double covalent bond. In further aspects, a particular bond is absent, thereby providing a double covalent bond.

In one aspect, R0 is optionally present. That is, in certain aspects, R0 is present. In further aspects, R0 is absent. In a further aspect, R0, when present, is hydrogen. It is understood that the presence and/or absence of R0 Groups and optional bonds serve to satisfy valence of the adjacent chemical moieties.

b. R1 Groups

In one aspect, R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl; or wherein R1a and R1b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl. In a further aspect, R1a is —CO2H. In a further aspect, R1b is methyl. In a further aspect, R1a and R1b are both methyl.

In one aspect, R1a is —C(O)ZR10. In a further aspect, R1a is selected from C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl. In a further aspect, R1b is selected from C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl.

In a further aspect, R1a and R1b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl.

c. R2 Groups

In one aspect, R2a and R2b are independently selected from hydrogen and —OR11, provided that at least one of R2a and R2b is —OR11; or wherein R2a and R2b together comprise ═O. In a further aspect, R2a is hydrogen, and R2b is —OR11. In a further aspect, R2a is —OR11, and R2b is hydrogen. In a further aspect, R2a and R2b together comprise ═O.

In a further aspect, R2a is hydrogen. In a further aspect, R2a is —OR11; wherein R11 is selected from hydrogen, C1-C6 alkyl, and —C(O)R14; wherein R14 is C1-C6 alkyl. In a further aspect, R2b is —OR11; wherein R11 is selected from hydrogen, C1-C6 alkyl, and —C(O)R14; and wherein R14 is C1-C6 alkyl. In a further aspect, R2b is —OR11; wherein R11 is hydrogen.

In a further aspect, R2b is hydrogen. In a further aspect, R2a is —OR11; wherein R11 is selected from hydrogen, C1-C6 alkyl, and —C(O)R14; wherein R14 is C1-C6 alkyl. In a further aspect, R2a is —OR11; wherein R11 is hydrogen.

d. R3 Groups

In one aspect, each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl; or wherein R3a and R3b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl.

In a further aspect, R3a is hydrogen. In a further aspect, R3b is —OR11; wherein R11 is selected from hydrogen, C1-C6 alkyl, and —C(O)R14; wherein R14 is C1-C6 alkyl.

e. R4 Groups

In one aspect, R4 is independently selected from C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl. In a further aspect, R4 is methyl. In a further aspect, R4, R5, and R6 are all methyl.

f. R5 Groups

In one aspect, R5 is independently selected from C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl. In a further aspect, R5 is methyl.

g. R6 Groups

In one aspect, R6 is independently selected from C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl. In a further aspect, R6 is methyl.

h. R7 Groups

In one aspect, R7 is selected from C1-C6 alkyl, —CH2OR12, and —C(O)ZR12. In a further aspect, R7 is C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl. In a further aspect, R7 is —CH2OR12. In a further aspect, R7 is and —C(O)ZR12.

i. R8 Groups

In one aspect, R8 is selected from hydrogen and C1-C6 alkyl. In a further aspect, R8 is hydrogen. In a further aspect, R8 is C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl.

j. R9 Groups

In one aspect, each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl.

In a further aspect, R9a is hydrogen. In a further aspect, R9a is C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl. In a further aspect, R9b is hydrogen. In a further aspect, R9b is C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl. In a further aspect, R9b is selected from methyl, ethyl, vinyl, n-propyl, propen-2-yl, propyl, 2-propenyl, n-butyl, 1-buten-2-yl, 1-buten-3-yl, i-butyl, 1-buten-2-yl, 1-buten-3-yl, s-butyl, 2-buten-1-yl, 2-buten-2-yl, 2-buten-3-yl, and t-butyl.

In a further aspect, R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl.

k. R10 Groups

In one aspect, R10 is selected from hydrogen and C1-C6 alkyl. In a further aspect, R10 is hydrogen. In a further aspect, R10 is C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl.

l. R11 Groups

In one aspect, each R11 is independently selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl.

In a further aspect, R11 is hydrogen. In a further aspect, R11 is selected from C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14. In a further aspect, R11 is C1-C6 alkyl. In a further aspect, R11 is C1-C5 heteroalkyl. In a further aspect, R11 is C3-C6 cycloalkyl. In a further aspect, R11 is C4-C6 heterocycloalkyl. In a further aspect, R11 is phenyl. In a further aspect, R11 is heteroaryl. In a further aspect, R11 is —C(O)R14.

In a further aspect, R11 is unsubstituted. In a further aspect, R11, where permitted, is substituted with 0-2 groups. In a further aspect, R11, where permitted, is substituted with 1 group. In a further aspect, R11, where permitted, is substituted with 2 groups.

m. R12 Groups

In one aspect, R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons. In a further aspect, R12 is hydrogen. In a further aspect, R12 is optionally substituted organic residue having from 1 to 20 carbons. In a further aspect, R12 is optionally substituted organic residue having from 3 to 12 carbons.

In a further aspect, R12 is hydrogen. In a further aspect, R12 is alkyl. In a further aspect, R12 is heteroalkyl. In a further aspect, R12 is cycloalkyl. In a further aspect, R12 is heterocycloalkyl. In a further aspect, R12 is aryl. In a further aspect, R12 is heteroaryl. In a further aspect, R12 is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl. In a further aspect, R12 comprises a group having a formula:

wherein m is an integer from 0 to 10 (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10); and wherein AA represents an amino acid residue. In a further aspect, R12 is AA is a phenylalanine residue. In a further aspect, R12 comprises a group having a formula:

n. R13 Groups

In one aspect, R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, and —NCH3—.

In a further aspect, R13 is hydrogen. In a further aspect, R13 is C1-C4 alkyl, for example, methyl, ethyl, propyl, or butyl. In a further aspect, Z is N, and —NR12R13 comprises a moiety of the formula:

o. R14 Groups

In one aspect, R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl.

In a further aspect, R14 is C1-C6 alkyl, for example, methyl, ethyl, propyl, butyl, pentyl, or hexyl. In a further aspect, R14 is unsubstituted. In a further aspect, R14, where permitted, is substituted with 0-2 groups. In a further aspect, R14, where permitted, is substituted with 1 group. In a further aspect, R14, where permitted, is substituted with 2 groups.

p. AA Groups

In one aspect, AA represents an amino acid residue, for example, phenylalanine.

q. Y Groups

In one aspect, Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, and —NCH3—.

r. Z Groups

In one aspect, Z is selected from —O— and —NR13—. In a further aspect, Z is —O—. In a further aspect, Z is —NR13—; wherein R13 is hydrogen. In a further aspect, Z is —NR13—; wherein R13 is C1-C4 alkyl.

2. Example Compounds

In one aspect, a compound can be present as one or more of the following structures:

In a further aspect, a compound can be present as one or more of the following structures:

3. Inhibtion of Muscle Atrophy and Induction of Muscle Hypertrophy

In one aspect, the disclosed compounds inhibit muscle atrophy. In a further aspect, the disclosed compounds increase muscle mass. In a still further aspect, the disclosed compounds induce muscle hypertrophy. In a yet further aspect, the disclosed compounds inhibit of muscle atrophy and increase muscle mass. In an even further aspect, the disclosed compounds inhibit of muscle atrophy and induce muscle hypertrophy. In a further aspect, the inhibition of muscle atrophy is in an animal. In an even further aspect, the increase in muscle mass is in an animal. In a still further aspect, the animal is a mammal, In a yet further aspect, the mammal is a human. In a further aspect, the mammal is a mouse. In a yet further aspect, the mammal is a rodent.

In a further aspect, the disclosed compounds inhibit muscle atrophy when administered at an oral dose of greater than about 200 mg per day in a human. In a yet further aspect, the disclosed compounds inhibit muscle atrophy when administered at an oral dose of greater than about 300 mg per day in a human. In a still further aspect, the disclosed compounds inhibit muscle atrophy when administered at an oral dose of greater than about 400 mg per day in a human. In an even further aspect, the disclosed compounds inhibit muscle atrophy when administered at an oral dose of greater than about 500 mg per day in a human. In a further aspect, the disclosed compounds inhibit muscle atrophy when administered at an oral dose of greater than about 750 mg per day in a human. In a yet further aspect, the disclosed compounds inhibit muscle atrophy when administered at an oral dose of greater than about 1000 mg per day in a human. In a still further aspect, the disclosed compounds inhibit muscle atrophy when administered at an oral dose of greater than about mg per day in a human. In an even further aspect, the disclosed compounds inhibit muscle atrophy when administered at an oral dose of greater than about 2000 mg per day in a human.

In a further aspect, the disclosed compounds increase muscle mass when administered at an oral dose of greater than about 200 mg per day in a human. In a yet further aspect, the disclosed compounds increase muscle mass when administered at an oral dose of greater than about 300 mg per day in a human. In a still further aspect, the disclosed compounds increase muscle mass when administered at an oral dose of greater than about 400 mg per day in a human. In an even further aspect, the disclosed compounds increase muscle mass when administered at an oral dose of greater than about 500 mg per day in a human. In a further aspect, the disclosed compounds increase muscle mass when administered at an oral dose of greater than about 750 mg per day in a human. In a yet further aspect, the disclosed compounds increase muscle mass when administered at an oral dose of greater than about 1000 mg per day in a human. In a still further aspect, the disclosed compounds increase muscle mass when administered at an oral dose of greater than about mg per day in a human. In an even further aspect, the disclosed compounds increase muscle mass when administered at an oral dose of greater than about 2000 mg per day in a human.

It is contemplated that one or more compounds can optionally be omitted from the disclosed invention.

C. METHODS OF MAKING THE COMPOUNDS

In one aspect, the disclosed compounds comprise the products of the synthetic methods described herein. In a further aspect, the disclosed compounds comprise a compound produced by a synthetic method described herein. In a still further aspect, the invention comprises a pharmaceutical composition comprising a therapeutically effective amount of the product of the disclosed methods and a pharmaceutically acceptable carrier. In a still further aspect, the invention comprises a method for manufacturing a medicament comprising combining at least one compound of any of disclosed compounds or at least one product of the disclosed methods with a pharmaceutically acceptable carrier or diluent.

In one aspect, the invention relates to methods of making functionalized ursane compounds useful in methods of inhibiting muscle atrophy and increasing muscle mass. Such compounds can be useful in the treatment of various maladies associated with muscle wasting, useful for increasing muscle mass and/or muscle strength, as well as in enhancing muscle formation and/or muscular performance. The compounds of the invention can be prepared by employing reactions as shown in the following schemes, in addition to other standard manipulations that are known in the literature, exemplified in the experimental sections or clear to one skilled in the art. For clarity, examples having a single substituent are shown where multiple substituents are allowed under the definitions disclosed herein. The following examples are provided so that the invention might be more fully understood, are illustrative only, and should not be construed as limiting.

1. Route 1: Alkyl Etherification

In one aspect, functionalized ursane compounds of the present invention can be prepared generically as shown below.

Compounds are represented in generic form, with substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below.

In one aspect, Route 1 step 1 begins with a free acid. In an appropriate solvent, a base (e.g., K2CO3, NaOH) strong enough to deprotonate the carboxylic acid, but not the alcohol, is added, and the reaction is conducted at a temperature effective and for a time effective to insure carboxylic acid deprotonation. An appropriate alkyl halide or halide equivalent is added to the reaction mixture, and the reaction is conducted at a temperature effective and for a time effective to insure alkylation of the carboxyl group. In a further aspect, an alternate Route 1 step 1 also begins with the free carboxylic acid. Diazomethane is added, and the reaction is conducted at a temperature effective and for a time effective to insure reaction.

In a further aspect, Route 1 step 2 the alkyl ester is dissolved in an appropriate dry solvent under anhydrous reaction conditions. A base is added, and the reaction is conducted at a temperature effective and for a time effective to insure deprotonation. Then, an appropriate alkyl, heteroalkyl, cycloalkyl, or heterocycloalkyl halide or halide equivalent (i.e., R11X) is added to the reaction mixture. The reaction is conducted at a temperature effective and for a time effective insure complete reaction.

In a further aspect, in Route 1 step 3, the O-alkylated ursane compound alkyl ester is hydrolyzed with an appropriate base, such as LiOH, in an appropriate organic-aqueous mixed solvent system at a temperature effective and for a time effective to insure reaction. Then the reaction mixture can be acidified to a suitable pH with an appropriate aqueous acid of an sufficient concentration and at a temperature effective and for a time effective to insure reaction.

2. Route 2: Aryl Etherification

In one aspect, functionalized ursane compounds of the present invention can be prepared generically as shown below.

Compounds are represented in generic form, with substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below.

In one aspect, Route 2 step 1 begins with the ursane compound free carboxylic acid. In an appropriate solvent, a base (e.g., K2CO3, NaOH) strong enough to deprotonate the carboxylic acid, but not the alcohol group, is added, and the reaction is conducted at a temperature effective and for a time effective to insure deprotonation. Then, an appropriate alkyl halide or halide equivalent is added to the reaction mixture, and the reaction is conducted at a temperature effective and for a time effective to insure alkylation of the carboxyl group. In a further aspect, an alternate Route 2 step 1 begins with the ursane compound free carboxylic acid in an appropriate solvent. Diazomethane is added, and the reaction is conducted at a temperature effective and for a time effective to insure reaction.

In a further apect, Route 2 step 2, the ursane compound alkyl ester is dissolved in an appropriate, dry solvent, along with phenol, an aryl alcohol, or appropriate heteroaryl alcohol, under anhydrous reaction conditions, followed by the addition of triphenylphosphine. The reaction is conducted at a effective temperature and for an effective time period. Then, an appropriate coupling agent, such as DIAD or DEAD, is added, and the reaction is conducted at a temperature effective and for a time effective to insure reaction. In a further aspect, in Route 2 step 3, the O-arylated or heteroarylated ursane compound alkyl ester can be treated with an appropriate base, such as LiOH, in an appropriate organic-aqueous mixed solvent system at a temperature effective and for a time effective to insure complete reaction. The reaction mixture can then be acidified to a suitable pH.

3. Route 3: Acylation

In one aspect, functionalized ursane compounds of the present invention can be prepared generically as shown below.

Compounds are represented in generic form, with substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below.

In one aspect, Route 3 step 1 begins with the the ursane compound free carboxylic acid. In an appropriate solvent, a base (e.g., K2CO3, NaOH) strong enough to deprotonate the carboxylic acid, but not the alcohol group, is added, and the reaction is allowed to progress at a temperature effective and for a time effective to insure carboxylic acid deprotonation. Then, an appropriate benzyl halide or halide equivalent is added to the reaction mixture, and the reaction is conducted at a temperature effective and for a time effective to insure protection of the carboxyl group.

In Route 3 step 2, the ursane compound benzyl ester is dissolved in an appropriate, dry solvent under anhydrous reaction conditions, followed by the addition of an appropriate acid scavenger (weak base, e.g., K2CO3 or DIEA). The acyl halide (e.g., R14COX) or equivalent acylating reagent is then added. The reaction is conducted at a temperature effective and for a time effective to insure reaction. In a further aspect, in an alternate Route 3 step 2, the the ursane compound benzyl ester and a suitable carboxylic acid (e.g., R14CO2H) are dissolved in an appropriate, dry solvent under anhydrous reaction conditions. Ethyl-(N′,N′-dimethylamino)propylcarbodiimide hydrochloride (EDC), 1-hydroxybenzotriazole (HOBt), and a trialkylamine (R3N) are then added, and the reaction is conducted at a temperature effective and for a time effective to insure reaction.

In Route 3 step 3, the acylated ursane compound benzyl ester is reduced under standard conditions (e.g., hydrogenation with hydrogen gas in the presence of a suitable palladium catalyst), thereby liberating the ursane compound free carboxlic acid.

4. Route 4: Esterification

In one aspect, functionalized ursane compounds of the present invention can be prepared generically as shown below.

Compounds are represented in generic form, with substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below.

In one aspect, Route 4 step 1 begins with the ursane compound free carboxylic acid. An appropriate alcohol (e.g., R12OH) is added, and the reaction is conducted at a temperature effective and for a time effective to time to insure reaction.

In a further aspect in an alternate syntheis, Route 4 step 1 begins with the ursane compound free carboxylic acid in a dry solvent under dry reaction conditions.

Tetrahydropyran (THP) is added, along with an acid catalyst (e.g., pTsOH). The reaction is conducted at a temperature effective and for a time effective to insure protection of the hydroxyl group. A base (e.g., NaOH or NaH) is then added to the THP-protected ursane compound free carboxylic acid, in a dry solvent under anhydrous reaction conditions. The reaction is conducted at a temperature effective and for a time effective to insure carboxylic acid deprotonation. Then, an appropriate alkyl halide (i.e., R12X) or equivalent is added to the reaction mixture, and the reaction is conducted at a temperature effective and for a time effective to insure alkylation of the carboxyl group. Route 4 step 3 begins with the THP-protected ursane compound alkyl ester in an alcohol solvent. An acid catalyst (e.g., pTsOH) is added, and the reaction is conducted at a temperature effective and for a time effective to insure deprotection.

5. Route 5: Amide Formation

In one aspect, functionalized ursane compounds of the present invention can be prepared generically as shown below.

Compounds are represented in generic form, with substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below.

In one aspect, Route 5 step 1 begins with the ursane compound free carboxylic acid in a dry solvent. Under dry reaction conditions, tetrahydropyran (THP) and an acid catalyst (e.g., pTsOH) are added. The reaction is then conducted at a temperature effective and for a time effective to insure protection of the hydroxyl group. In Route 5 step 2, the THP-protected ursane compound free carboxylic acid is dissolved in an appropriate, dry solvent. Under anhydrous reaction conditions, a suitable amine (e.g., R12R13NH) is added, along with ethyl-(N′,N′-dimethylamino)propylcarbodiimide hydrochloride (EDC), 1-hydroxybenzotriazole (HOBt), and a trialkylamine (R3N), and the reaction is conducted at a temperature effective and for a time effective to time to insure complete reaction. In Route 5 step 3, the THP-protected ursane compound amide can then be deprotected by addition of an acid catalyst (e.g., pTsOH), and the reaction is conducted at a temperature effective and for a time effective to insure reaction.

6. Route 6: Reduction to Alcohol

In one aspect, functionalized ursane compounds of the present invention can be prepared generically as shown below.

Compounds are represented in generic form, with substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below.

In one aspect, the ursane compound free carboxylic acid, in a dry solvent, can be reacted with lithium aluminum hydride (LiAlH4) under dry reaction conditions to provide the corresponding primary alcohol. Alternatively, the ursane compound free carboxylic acid, in a dry solvent, can be reacted with diborane (B2H6) under dry reaction conditions to provide the corresponding primary alcohol. It is understood that protecting group chemistry, if needed, can also be used to protect sensitive remote functionality during these reaction steps.

7. Route 7: Hydroxyl Inversion

In one aspect, functionalized ursane compounds of the present invention can be prepared generically as shown below.

Compounds are represented in generic form, with substituents as noted in compound descriptions elsewhere herein. A more specific example is set forth below.

In one aspect, a hydroxyl functionality can be substituted with another group (e.g., alkoxyl, acyl, amino, etc.), while inverting the stereochemistry at the adjacent carbon, by reaction with an appropriate protic nucleophile in the presence of diethylazodicarboxylate (DEAD) and triphenylphosphine under Mitsunobu reaction conditions. While —OR11 is shown, it is understood that additional moieties (e.g., acetoxyl, amino, etc.) can be substituted at that position by appropriate selection of protic nucleophile (e.g., acetic acid, ammonia, etc.).

8. Plant Sources of Ursolic Acid Derivatives

Many pentacyclic acid triterpenes useful as synthetic precursors to the ursolic acid derivatives in the synthetic methods described above may be isolated and purified from a natural source such as plants or materials derived from plants. Alternatively, certain known synthetic precursors useful in the preparation of ursolic acid derivatives can often be obtained from commercial sources. Ursolic Acid is a useful known synthetic precursor to ursolic acid derivatives that can be used as a synthetic precursor to prepare certain disclosed compounds. For example, ursolic acid can be isolated from plants such as Holy Basil (Ocimum sanctum L.), peppermint leaves (Mentha piperita L.), lavender (Lavandula augustifolia Mill.), oregano (Origanum vulgare L.), thyme (Thymus vulgaris L.), hawthorn (Crataegus laevigata (Poir) DC), cherry laurel leaves (Prunus laurocerasus L.), loquat leaves (Eriobotrya japonica L.), glossy privet leaves (Ligustrum lucidum Ait. L.), bilberry (Vacciunum myrtillus L.), Devil's Claw (Harpagophytum procumbens DC), Elder Flowers (European var.; Sambucus nigra L.), and periwinkle (Vinca minor L.).

A variety of methods that are generally applicable to purifying ursolic acid and ursolic acid derivatives. For example, Nishimura, et al. (J. Nat. Prod. 1999, 62, 1061-1064) described the identification of 2,3-dihydroxy-24-nor-urs-4(23),12-dien-28-oic acid and 23-hydroxyursolic acid. Nishimura described procedures to isolate these compounds. Procedures described herein demonstrate these compounds will be contained in flash chromatography fraction 3 (FCF3) as described in the examples. Similar HPLC procedures described herein can be used to further purify these compounds including using a gradient with water with 0.05% TFA and acetonitrile with 0.05% TFA, mobile phase A and B respectively, with a C18 BetaMax Neutral column (250×8 mm; 5 um). The gradient may consist of 40% β isocratic for 5 min, then from approximately 40% to 70% B in 30 min. A skilled artisan would recognize the general applicability of the methods described in Nishimura et al to efficiently isolate either the ursolic acid, ursolic acid derivatives or structurally related pentacyclic acid triterpenes from various plants.

Other illustrative methods that are generally applicable to purifying ursolic acid and ursolic acid derivatives are also known. For example, Chaturvedula, et al. (J. Nat. Prod. 2004, 67, p. 899-901) described the isolation of 3-acetoxy-2-hydroxy ursolic acid, 3-(p-coumaroyl)ursolic acid, and 2,3-diacetoxyursolic acid. Adnyana, et al. (J. Nat. Prod. 2001, 64, p. 360-363) described the isolation of 2,3,6,19-tetrahydroxyoleanolic acid, 2,3,19-trihydroxyoleanolic acid, 2,3,19,23-tetrahydroxyursolic acid, and 2,3,23-trihydroxyoleanolic acid. Ikuta, et al. (J. Nat. Prod. 2003, 66, p. 1051-1054) described the isolation of 2,3-dihydroxyurs-12-en-11-on-28-oic acid and 2,3-dihydroxy-11-methoxyurs-12-en-28-oic acid. For example, similar HPLC procedures such as those described in U.S. Pat. No. 7,612,045 can be used to further purify these compounds including using a gradient with water with 0.05% TFA and acetonitrile with 0.05% TFA, mobile phase A and B respectively, with a C18 BetaMax Neutral column (250×8 mm; 5 um). The gradient may consist of 40% β isocratic for 5 min, then from approximately 40% to 70% B in 30 min.

Finally, another source of the known synthetic precursors useful in the synthetic methods described above to prepare ursolic acid derivatives are commercial sources or vendors. Purified forms of corosolic acid, ursolic acid, oleanolic acid, madecassic acid, asiatic acid, pygenic acid (A, B or C), caulophyllogenin and echinocystic acid may be obtained from a commercial source. For example, ursolic acid and oleanolic acid may be purchased from Sigma-Aldrich Chemical Company (St. Louis, Mo., USA) and corosolic acid, asiatic acid, madecassic acid, pygenic acid (A, B, or C), caulophyllogenin and echinocystic acid may be purchased from Chromadex (Santa Ana, Calif., USA). The compounds obtained from commercial sources may be furthered separated and purified as needed using methods such as column chromatography, high pressure liquid chromatography (HPLC), and/or recrystallization described herein. Additional methods of isolation of precursors are described in U.S. Pat. No. 7,612,045, U.S. patent application Ser. No. 10/355,201, and U.S. patent application Ser. No. 10/445,943.

It is further anticipated that the compounds of the invention can be obtained by direct synthesis. Direct synthesis may include either total synthesis or semi-synthesis. Exemplary synthetic methods for obtaining these compounds are described above. Additional synethetic procedures useful in the preparation of ursolic acid derivatives are described in U.S. Pat. No. 3,903,089, U.S. Pat. No. 7,612,045, and U.S. patent application Ser. No. 10/445,943, U.S. patent application Ser. No. 10/355,201. Further synthetic methods useful in the preparation of ursolic acid derivatives are Meng, Y., et al. (2010) Molecules 15:4033-4040; Gao, Y., et al. (2010) Molecules 15:4439-4449; Sporn, M. B., et al. (2011) Journal of Natural Products 74:537-545; Chadalapaka, G., et al. (2008) Biorganic and Medicinal Chemistry Letters 18(8):2633-2639; and, Sun, H., et al. (2006) Botanical Studies 47:339-368.

It is contemplated that each disclosed methods can further comprise additional steps, manipulations, and/or components. It is also contemplated that any one or more step, manipulation, and/or component can be optionally omitted from the invention. It is understood that a disclosed methods can be used to provide the disclosed compounds. It is also understood that the products of the disclosed methods can be employed in the disclosed methods of using.

D. PHARMACEUTICAL COMPOSITIONS

In one aspect, the invention relates to pharmaceutical compositions comprising the disclosed compounds. That is, a pharmaceutical composition can be provided comprising a therapeutically effective amount of at least one disclosed compound or at least one product of a disclosed method and a pharmaceutically acceptable carrier.

In one aspect, the invention relates to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of a compound having a structure represented by a formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl, or wherein R1a and R1b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen, or R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl, wherein R3a and R3b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12 and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein R19 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount effective to prevent or treat muscle atrophy in the animal, wherein the amount is greater than about 1000 mg per day when the compound is ursolic acid, boswellic acid, corosolic acid, betulinic acid, or UA0713.

In one aspect, the animal is an animal. In a further aspect, the animal is a mammal. In a yet further aspect, the mammal is a primate. In a still further aspect, the mammal is a human. In an even further aspect, the human is a patient.

In a further aspect, the animal is a domesticated animal. In a still further aspect, the domesticated animal is a domesticated fish, domesticated crustacean, or domesticated mollusk. In a yet further aspect, the domesticated animal is poultry. In an even further aspect, the poultry is selected from chicken, turkey, duck, and goose. In a still further aspect, the domesticated animal is livestock. In a yet further aspect, the livestock animal is selected from pig, cow, horse, goat, bison, and sheep.

In a further aspect, the effective amount is a therapeutically effective amount. In a still further aspect, the effective amount is a prophylactically effective amount. In a yet further aspect, the muscle disorder is muscle atrophy . In an even further aspect, the muscle disorder is a condition in need of increasing muscle mass. In an even further aspect, the effective amount is greater than about 1000 mg per day when the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713.

In a further aspect, the pharmaceutical composition is administered following identification of the mammal in need of treatment of muscle atrophy. In a still further aspect, the pharmaceutical composition is administered following identification of the mammal in need of prevention of muscle atrophy. In an even further aspect, the mammal has been diagnosed with a need for treatment of muscle atrophy prior to the administering step.

In a further aspect, the compound is not ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In a yet further aspect, the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713.

In certain aspects, the disclosed pharmaceutical compositions comprise the disclosed compounds (including pharmaceutically acceptable salt(s) thereof) as an active ingredient, a pharmaceutically acceptable carrier, and, optionally, other therapeutic ingredients or adjuvants. The instant compositions include those suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.

As used herein, the term “pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids. When the compound of the present invention is acidic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases. Salts derived from such inorganic bases include aluminum, ammonium, calcium, copper (-ic and -ous), ferric, ferrous, lithium, magnesium, manganese (-ic and -ous), potassium, sodium, zinc and the like salts. Particularly preferred are the ammonium, calcium, magnesium, potassium and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, as well as cyclic amines and substituted amines such as naturally occurring and synthesized substituted amines. Other pharmaceutically acceptable organic non-toxic bases from which salts can be formed include ion exchange resins such as, for example, arginine, betaine, caffeine, choline, N,N′-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.

As used herein, the term “pharmaceutically acceptable non-toxic acids”, includes inorganic acids, organic acids, and salts prepared therefrom, for example, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid and the like. Preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.

In practice, the compounds of the invention, or pharmaceutically acceptable salts thereof, of this invention can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). Thus, the pharmaceutical compositions of the present invention can be presented as discrete units suitable for oral administration such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient. Further, the compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non-aqueous liquid, as an oil-in-water emulsion or as a water-in-oil liquid emulsion. In addition to the common dosage forms set out above, the compounds of the invention, and/or pharmaceutically acceptable salt(s) thereof, can also be administered by controlled release means and/or delivery devices. The compositions can be prepared by any of the methods of pharmacy. In general, such methods include a step of bringing into association the active ingredient with the carrier that constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both. The product can then be conveniently shaped into the desired presentation.

Thus, the pharmaceutical compositions of this invention can include a pharmaceutically acceptable carrier and a compound or a pharmaceutically acceptable salt of the compounds of the invention. The compounds of the invention, or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds.

The pharmaceutical carrier employed can be, for example, a solid, liquid, or gas. Examples of solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Examples of liquid carriers are sugar syrup, peanut oil, olive oil, and water. Examples of gaseous carriers include carbon dioxide and nitrogen.

In preparing the compositions for oral dosage form, any convenient pharmaceutical media can be employed. For example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like can be used to form oral liquid preparations such as suspensions, elixirs and solutions; while carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like can be used to form oral solid preparations such as powders, capsules and tablets. Because of their ease of administration, tablets and capsules are the preferred oral dosage units whereby solid pharmaceutical carriers are employed. Optionally, tablets can be coated by standard aqueous or nonaqueous techniques

A tablet containing the composition of this invention can be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants. Compressed tablets can be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.

The pharmaceutical compositions of the present invention comprise a compound of the invention (or pharmaceutically acceptable salts thereof) as an active ingredient, a pharmaceutically acceptable carrier, and optionally one or more additional therapeutic agents or adjuvants. The instant compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.

Pharmaceutical compositions of the present invention suitable for parenteral administration can be prepared as solutions or suspensions of the active compounds in water. A suitable surfactant can be included such as, for example, hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.

Pharmaceutical compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability. The pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof.

Pharmaceutical compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, mouth washes, gargles, and the like. Further, the compositions can be in a form suitable for use in transdermal devices. These formulations can be prepared, utilizing a compound of the invention, or pharmaceutically acceptable salts thereof, via conventional processing methods. As an example, a cream or ointment is prepared by mixing hydrophilic material and water, together with about 5 wt % to about 10 wt % of the compound, to produce a cream or ointment having a desired consistency.

Pharmaceutical compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories can be conveniently formed by first admixing the composition with the softened or melted carrier(s) followed by chilling and shaping in moulds.

In addition to the aforementioned carrier ingredients, the pharmaceutical formulations described above can include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti-oxidants) and the like. Furthermore, other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient. Compositions containing a compound of the invention, and/or pharmaceutically acceptable salts thereof, can also be prepared in powder or liquid concentrate form.

In the treatment conditions which require modulation of cellular function related to muscle growth an appropriate dosage level will generally be about 0.01 to 500 mg per kg patient body weight per day and can be administered in single or multiple doses. Preferably, the dosage level will be about 0.1 to about 250 mg/kg per day; more preferably 0.5 to 100 mg/kg per day. A suitable dosage level can be about 0.01 to 250 mg/kg per day, about 0.05 to 100 mg/kg per day, or about 0.1 to 50 mg/kg per day. Within this range the dosage can be 0.05 to 0.5, 0.5 to 5.0 or 5.0 to 50 mg/kg per day. For oral administration, the compositions are preferably provided in the from of tablets containing 1.0 to 1000 miligrams of the active ingredient, particularly 1.0, 5.0, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900 and 1000 milligrams of the active ingredient for the symptomatic adjustment of the dosage of the patient to be treated. The compound can be administered on a regimen of 1 to 4 times per day, preferably once or twice per day. This dosing regimen can be adjusted to provide the optimal therapeutic response.

It is understood, however, that the specific dose level for any particular patient will depend upon a variety of factors. Such factors include the age, body weight, general health, sex, and diet of the patient. Other factors include the time and route of administration, rate of excretion, drug combination, and the type and severity of the particular disease undergoing therapy.

The present invention is further directed to a method for the manufacture of a medicament for modulating cellular activity related to muscle growth (e.g., treatment of one or more disorders associated with muscle dysfunction or atrophy) in mammals (e.g., humans) comprising combining one or more disclosed compounds, products, or compositions with a pharmaceutically acceptable carrier or diluent. Thus, in one aspect, the invention relates to a method for manufacturing a medicament comprising combining at least one disclosed compound or at least one disclosed product with a pharmaceutically acceptable carrier or diluent.

The disclosed pharmaceutical compositions can further comprise other therapeutically active compounds, which are usually applied in the treatment of the above mentioned pathological conditions.

It is understood that the disclosed compositions can be prepared from the disclosed compounds. It is also understood that the disclosed compositions can be employed in the disclosed methods of using.

E. METHODS OF USING THE COMPOUNDS AND COMPOSITIONS

1. Muscle Atrophy

Muscle atrophy is defined as a decrease in the mass of the muscle; it can be a partial or complete wasting away of muscle. When a muscle atrophies, this leads to muscle weakness, since the ability to exert force is related to mass. Muscle atrophy is a co-morbidity of several common diseases, and patients who have “cachexia” in these disease settings have a poor prognosis.

Muscle atrophy can also be skeletal muscle loss or weakness caused by malnutrition, aging, muscle disuse (such as voluntary and involuntary bedrest, neurologic disease (such as multiple sclerosis, amyotrophic lateral sclerosis, spinal muscular atrophy, critical illness neuropathy, spinal cord injury, peripheral neuropathy, or peripheral nerve injury), injury to the limbs or joints, casting, other post-surgical forms of limb immobilization, or spaceflight), chronic disease (such as cancer, congestive heart failure, chronic pulmonary disease, chronic renal failure, chronic liver disease, diabetes mellitus, glucocorticoid hypersecretion, and chronic infections such as HIV/AIDS or tuberculosis), burn injuries, sepsis, other illnesses requiring mechanical ventiliation, drug-induced muscle disease (such as glucocorticoid-induced myopathy and statin-induced myopathy), genetic diseases that primarily affect skeletal muscle (such as muscular dystrophy, myotonic dystrophy and inclusion body myositis), or autoimmune diseases that affect skeletal muscle (such as polymyositis and dermatomyositis).

There are many diseases and conditions which cause muscle atrophy, including malnutrition, muscle disuse (secondary to voluntary or involuntary bedrest, neurologic disease (including multiple sclerosis, amyotrophic lateral sclerosis, spinal muscular atrophy, critical illness neuropathy, spinal cord injury or peripheral nerve injury), orthopedic injury, casting, and other post-surgical forms of limb immobilization), chronic disease (including cancer, congestive heart failure, chronic pulmonary disease, chronic renal failure, chronic liver disease, diabetes mellitus, Cushing syndrome and chronic infections such as HIV/AIDS or tuberculosis), burns, sepsis, other illnesses requiring mechanical ventilation, drug-induced muscle disease (such as glucorticoid-induced myopathy and statin-induced myopathy), genetic diseases that primarily affect skeletal muscle (such as muscular dystrophy and myotonic dystrophy), autoimmune diseases that affect skeletal muscle (such as polymyositis and dermatomyositis), spaceflight, and aging.

Muscle atrophy occurs by a change in the normal balance between protein synthesis and protein degradation. During atrophy, there is a down-regulation of protein synthesis pathways, and an activation of protein breakdown pathways. The particular protein degradation pathway which seems to be responsible for much of the muscle loss seen in a muscle undergoing atrophy is the ATP-dependent, ubiquitin/proteasome pathway. In this system, particular proteins are targeted for destruction by the ligation of at least four copies of a small peptide called ubiquitin onto a substrate protein. When a substrate is thus “poly-ubiquitinated,” it is targeted for destruction by the proteasome. Particular enzymes in the ubiquitin/proteasome pathway allow ubiquitination to be directed to some proteins but not others—specificity is gained by coupling targeted proteins to an “E3 ubiquitin ligase.” Each E3 ubiquitin ligase binds to a particular set of substrates, causing their ubiquitination. For example, in skeletal muscle, the E3 ubiquitin ligases atrogin-1 and MuRF1 are known to play essential roles protein degradation and muscle atrophy.

Muscle atrophy can be opposed by the signaling pathways which induce muscle hypertrophy, or an increase in muscle size. Therefore one way in which exercise induces an increase in muscle mass is to downregulate the pathways which have the opposite effect. One important rehabilitation tool for muscle atrophy includes the use of functional electrical stimulation to stimulate the muscles which has had limited success in the rehabilitation of paraplegic patients.

Ursolic acid or ursolic acid derivatives can be used as a therapy for illness- and age-related muscle atrophy. It can be useful as a monotherapy or in combination with other strategies that have been considered, such as myostatin inhibition (Zhou, X., et al. (2010) Cell 142(4): 531-543). Given its capacity to reduce adiposity, fasting blood glucose and plasma lipid levels, ursolic acid or ursolic acid derivatives can also be used as a therapy for obesity, metabolic syndrome and type 2 diabetes.

The disclosed compounds can be used as single agents or in combination with one or more other drugs in the treatment, prevention, control, amelioration or reduction of risk of the aforementioned diseases, disorders and conditions for which compounds of formula I or the other drugs have utility, where the combination of drugs together are safer or more effective than either drug alone. The other drug(s) can be administered by a route and in an amount commonly used therefore, contemporaneously or sequentially with a disclosed compound. When a disclosed compound is used contemporaneously with one or more other drugs, a pharmaceutical composition in unit dosage form containing such drugs and the disclosed compound is preferred. However, the combination therapy can also be administered on overlapping schedules. It is also envisioned that the combination of one or more active ingredients and a disclosed compound will be more efficacious than either as a single agent.

Systemic administration of ursolic acid (by parenteral injection or by oral consumption) can be used to promote muscle growth and reduce muscle atrophy in all muscles, including those of the limbs and the diaphragm. Local administration of ursolic acid (by a topical route or localized injection) can be used to promote local muscle growth, as can be required following a localized injury or surgery.

In one aspect, the subject compounds can be coadministered with agents that stimulate insulin signaling, IGF1 signaling and/or muscle growth including insulin, insulin analogs, insulin-like growth factor 1, metformin, thiazoladinediones, sulfonylureas, meglitinides, leptin, dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 agonists, tyrosine-protein phosphatase non-receptor type 1 (PTPN1 a.k.a. PTP1B) inhibitors, myostatin signaling inhibitors, clenbuterol, and androgens including testosterone and 5-dehydroepiandrosterone. The derivative can be corosolic acid, UA0713, or other pentacyclic triterpene acids. The ursolic acid, derivative or salt thereof can be administered orally, intramuscularly, intravenously or intraarterially. The ursolic acid, derivative or salt thereof can be substantially pure. The ursolic acid, derivative or salt thereof can be administered at about 10 mg/day to 10 g/day.

In another aspect, the subject compounds can be administered in combination with agents that stimulate insulin signaling, IGF1 signaling and/or muscle growth including insulin, insulin analogs, insulin-like growth factor 1, metformin, thiazoladinediones, sulfonylureas, meglitinides, leptin, dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 agonists, tyrosine-protein phosphatase non-receptor type 1 (PTPN1, which is also commonly referred to as PTP1B) inhibitors, myostatin signaling inhibitors, clenbuterol, and androgens including testosterone and 5-dehydroepiandrosterone. The derivative can be corosolic acid, UA0713, or other pentacyclic triterpene acids. The ursolic acid, derivative or salt thereof can be administered orally, intramuscularly, intravenously or intraarterially. The ursolic acid, derivative or salt thereof can be substantially pure. The ursolic acid, derivative or salt thereof can be administered at about 10 mg/day to 10 g/day.

The pharmaceutical compositions and methods of the present invention can further comprise other therapeutically active compounds as noted herein which are usually applied in the treatment of the above mentioned pathological conditions.

2. Treatment Methods

The compounds disclosed herein are useful for treating, preventing, ameliorating, controlling or reducing the risk of a variety of muscle disorders. Examples of such muscle disorders include, but are not limited to, skeletal muscle atrophy secondary to malnutrition, muscle disuse (secondary to voluntary or involuntary bedrest), neurologic disease (including multiple sclerosis, amyotrophic lateral sclerosis, spinal muscular atrophy, critical illness neuropathy, spinal cord injury or peripheral nerve injury), orthopedic injury, casting, and other post-surgical forms of limb immobilization, chronic disease (including cancer, congestive heart failure, chronic pulmonary disease, chronic renal failure, chronic liver disease, diabetes mellitus, Cushing syndrome and chronic infections such as HIV/AIDS or tuberculosis), burns, sepsis, other illnesses requiring mechanical ventiliation, drug-induced muscle disease (such as glucorticoid-induced myopathy and statin-induced myopathy), genetic diseases that primarily affect skeletal muscle (such as muscular dystrophy and myotonic dystrophy), autoimmune diseases that affect skeletal muscle (such as polymyositis and dermatomyositis), spaceflight, or age-related sarcopenia.

Thus, provided is a method for treating or preventing muscle atrophy, comprising: administering to a subject at least one disclosed compound; at least one disclosed pharmaceutical composition; and/or at least one disclosed product in a dosage and amount effective to treat the disorder in the subject.

Also provided is a method for increasing muscle mass, comprising: administering to a subject at least one disclosed compound; at least one disclosed pharmaceutical composition; and/or at least one disclosed product in a dosage and amount effective to treat the disorder in the subject.

a. Preventing or Treating Muscle Atrophy

In one aspect, the invention relates to a method for preventing or treating muscle atrophy in an animal, the method comprising administering to the animal a compound of the formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl, or wherein R1a and R1b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen, or R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl, wherein R3 and R3b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12 and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount effective to prevent or treat muscle atrophy in the animal, wherein the amount is greater than about 1000 mg per day when the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713.

In a further aspect, the compound administered is a disclosed compound or a product of a disclosed method of making a compound.

In a further aspect, the animal is a mammal. In a yet further aspect, the mammal is a primate. In a still further aspect, the mammal is a human. In an even further aspect, the human is a patient.

In a further aspect. the animal is a domesticated animal. In a still further aspect, the domesticated animal is a domesticated fish, domesticated crustacean, or domesticated mollusk. In a yet further aspect, the domesticated animal is poultry. In an even further aspect, the poultry is selected from chicken, turkey, duck, and goose. In a still further aspect, the domesticated animal is livestock. In a yet further aspect, the livestock animal is selected from pig, cow, horse, goat, bison, and sheep.

In a further aspect, the effective amount is a therapeutically effective amount. In a still further aspect, the effective amount is a prophylactically effective amount. In a yet further aspect, muscle atrophy is prevented by administration of the compound. In an even further aspect, muscle atrophy is treated by administration of the compound. In a still further aspect, the method further comprises the step of identifying the mammal in need of treatment of muscle atrophy. In a yet further aspect, the method further comprises the step of identifying the mammal in a need of prevention of muscle atrophy. In an even further aspect, the mammal has been diagnosed with a need for treatment of muscle atrophy prior to the administering step.

In a further aspect, the compound is not ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In a still further aspect, the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In yet further aspect, the compound is not administered as a foodstuff.

b. Increasing Muscle Mass and/or Strength

In one aspect, the invention relates to a method for increasing muscle mass and/or muscular strength in an animal, the method comprising administering to the animal a compound of the formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl, or wherein R1a and R1b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen, or R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl, wherein R3a and R3b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12 and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount effective to prevent or treat muscle atrophy in the animal, wherein the amount is greater than about 1000 mg per day when the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In a further aspect, the compound administered is a disclosed compound or a product of a disclosed method of making a compound.

In a further aspect, the compound administered is a disclosed compound or a product of a disclosed method of making a compound.

In a further aspect, the animal is a mammal. In a yet further aspect, the mammal is a primate. In a still further aspect, the mammal is a human. In an even further aspect, the human is a patient.

In a further aspect. the animal is a domesticated animal. In a still further aspect, the domesticated animal is a domesticated fish, domesticated crustacean, or domesticated mollusk. In a yet further aspect, the domesticated animal is poultry. In an even further aspect, the poultry is selected from chicken, turkey, duck, and goose. In a still further aspect, the domesticated animal is livestock. In a yet further aspect, the livestock animal is selected from pig, cow, horse, goat, bison, and sheep.

In a further aspect, the effective amount is a therapeutically effective amount. In a still further aspect, the effective amount is a prophylactically effective amount. In a yet further aspect, muscle atrophy is prevented by administration of the compound. In an even further aspect, muscle atrophy is treated by administration of the compound. In a still further aspect, the method further comprises the step of identifying the mammal in need of treatment of muscle atrophy. In a yet further aspect, the method further comprises the step of identifying the mammal in need of prevention of muscle atrophy. In an even further aspect, the mammal has been diagnosed with a need for treatment of muscle atrophy prior to the administering step.

In a further aspect, the compound is not ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In a still further aspect, the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In yet further aspect, the compound is not administered as a foodstuff.

c. Enhancing Muscle Formation

In one aspect, the invention relates to a method of enhancing muscle formation in a mammal, the method comprising administering to the mammal a compound of the formula:

wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, R12 and R13, when present, are covalently bonded and —NR12R13 comprises a moiety represented by the formula:

wherein X is selected from O, S, SO, SO2, NH and NCH3; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount of at least about 200 mg/kg and effective to enhance muscle formation in the mammal.

In a further aspect, the compound administered is a disclosed compound or a product of a disclosed method of making a compound.

In a further aspect, the mammal is a human. In a still further aspect, the human is a patient. In a yet further aspect, administration of the compound prevents muscle atrophy in the mammal. In an even further aspect, administration of the compound treats muscle atrophy in the mammal. In a still further aspect, administration of of the compound increases muscle mass in the mammal. In a yet further aspect, administration of the compound increases muscular strength in the mammal.

In a further aspect, the compound is administered in an effective amount. In a yet further aspect, the effective amount is a therapeutically effective amount. In a still further aspect, the effective amount is a prophylactically effective amount. In a still further aspect, the method further comprises the step of identifying the mammal in need of treatment of muscle atrophy. In a yet further aspect, the method further comprises the step of identifying the mammal in need of prevention of muscle atrophy. In an even further aspect, the mammal has been diagnosed with a need for treatment of muscle atrophy prior to the administering step.

In a further aspect. the mammal is a domesticated animal. In a yet further aspect, domesticated animal is livestock. In a yet further aspect, the livestock animal is selected from pig, cow, horse, goat, bison, and sheep.

In a further aspect, the compound is not ursolic acid. In a still further aspect, the compound is ursolic acid. In a further aspect, the compound is not ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In a still further aspect, the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In yet further aspect, the compound is not administered as a foodstuff.

3. Enhancing Tissue Growth In Vitro

In one aspect, the invention relates to a method of enhancing tissue growth in vitro, the method comprising administering to the tissue a compound of the formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl, or wherein R1a and R1b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen, or R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl, wherein R3a and R3b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12 and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in an amount effective to enhance growth of the tissue.

In a further aspect, the compound administered is a disclosed compound or a product of a disclosed method of making a compound.

In a further aspect, the tissue comprises animal cells. In a still further aspect, the animal cells are muscle cells. In a yet further aspect, the muscle cells are myosatellite cells. In an even further aspect, the myosatellite cells are grown on a scaffold.

4. Manufacture of a Medicament

In one aspect, the invention relates to a method for the manufacture of a medicament for inhibiting muscle atrophy and for increasing muscle mass in a mammal comprising combining a therapeutically effective amount of a disclosed compound or product of a disclosed method with a pharmaceutically acceptable carrier or diluent.

In a further aspect, the medicament is modulates muscle growth. In a still further aspect, the medicament inhibits muscle atrophy. In a yet further aspect, the medicament increases muscle mass. In an even further aspect, the medicament induces skeletal muscle hypertrophy.

5. Methods of Testing for Performance Enhancing Use

In one aspect, the invention relates to a method of testing for performance enhancing use of a ursolic acid analog in an animal, the method comprising: (a) obtaining a biological test sample from the animal; and (b) measuring the amount of a compound of formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl, or wherein R1a and R1b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein one of R1a and R2b is —OR11, and the other is hydrogen, or R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl, wherein R3a and R3b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12 and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, in the test sample to determine whether a superphysiological amount of the compound is present in the biological test sample; wherein the superphysiological amount of the compound in the biological test sample is indicative of performance enhancing use of the compound.

In a further aspect, the superphysiological amount is greater than the peak concentration from administration at a level of about 1000 mg per day. In a still further aspect, the superphysiological amount is the amount that results from administration of the compound at a level greater than 200 mg per day. In a still further aspect, the superphysiological amount is the amount resulting from administration of the compound at a level greater than 200 mg per day. In an even further aspect, the biological test sample is obtained about 12 hours to about 96 hours following administration of the compound.

In a further aspect, the animal is a mammal. In a yet further aspect, the animal is a domesticated animal. In a still further aspect, the mammal is a human.

In a further aspect, the biological sample is blood, urine, saliva, hair, muscle, skin, fat, or breath.

6. Use of Compounds

In one aspect, the invention relates to the use of a compound for increasing muscle mass in a mammal, the compound having a structure represented by a formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl, or wherein R1a and R1b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein one of R2a and R2b is —OR11, and the other is hydrogen, or R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl, wherein R3a and R3b are optionally covalently bonded and, together with the intermediate carbon, comprise an optionally substituted C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12 and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise C3-C5 cycloalkyl or C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein R11 is selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, R12 and R13, when present, are covalently bonded and —NR12R13 comprises a moiety represented by the formula:

wherein X is selected from O, S, SO, SO2, NH and NCH3; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof.

In a further aspect, a use is the treatment of a mammal. In a yet further aspect, the mammal is a human. In a still further aspect, the human is a patient. In a yet further aspect, a use is administration of the compound to a mammal to prevent muscle atrophy. In a yet further aspect, a use is administration of the compound to increase muscular strength in the mammal. In a further aspect. the mammal is a domesticated animal. In a yet further aspect, domesticated animal is livestock. In a yet further aspect, the livestock animal is selected from pig, cow, horse, goat, bison, and sheep.

In a further aspect, a use is administration of the compound in an effective amount. In a yet further aspect, the effective amount is a therapeutically effective amount. In a still further aspect, the effective amount is a prophylactically effective amount. In a still further aspect, prior to use the mammal in need of treatment of muscle atrophy is identified. In a yet further aspect, prior to use the mammal in need of prevention of muscle atrophy is identified. In an even further aspect, the mammal has been diagnosed with a need for treatment of muscle atrophy prior to the administering step.

In a further aspect, the compound is not ursolic acid. In a still further aspect, the compound is ursolic acid. In a further aspect, the compound is not ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In a still further aspect, the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713. In yet further aspect, the compound is not used as a foodstuff. In an even further aspect, the compound is used in an amount is greater than about 1000 mg per day when the compound is ursolic acid, beta-boswellic acid, corosolic acid, betulinic acid, or UA0713.

7. Kits

In one aspect, the invention relates to a kit comprising at least one compound having a structure represented by a formula:

wherein each is an optional covalent bond, and R0 is optionally present; wherein n is 0 or 1; wherein R0, when present, is hydrogen; wherein R1a is selected from C1-C6 alkyl and —C(O)ZR10; wherein R1b is selected from C1-C6 alkyl; or wherein R1a and R1b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R2a and R2b are independently selected from hydrogen and —OR11, provided that at least one of R2a and R2b is —OR11; or wherein R2a and R2b together comprise ═O; wherein each of R3a and R3b is independently selected from hydrogen, hydroxyl, C1-C6 alkyl, and C1-C6 alkoxyl, provided that R3a and R3b are not simultaneously hydroxyl; or wherein R3a and R3b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein each of R4, R5, and R6 is independently selected from C1-C6 alkyl; wherein R7 is selected from C1-C6 alkyl, —CH2OR12, and —C(O)ZR12; wherein R8 is selected from hydrogen and C1-C6 alkyl; wherein each of R9a and R9b is independently selected from hydrogen and C1-C6 alkyl, provided that R9a and R9b are not simultaneously hydrogen; or wherein R9a and R9b are covalently bonded and, along with the intermediate carbon, together comprise optionally substituted C3-C5 cycloalkyl or optionally substituted C2-C5 heterocycloalkyl; wherein R10 is selected from hydrogen and C1-C6 alkyl; wherein each R11 is independently selected from hydrogen, C1-C6 alkyl, C1-C5 heteroalkyl, C3-C6 cycloalkyl, C4-C6 heterocycloalkyl, phenyl, heteroaryl, and —C(O)R14; wherein R11, where permitted, is substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; wherein R12 is selected from hydrogen and optionally substituted organic residue having from 1 to 20 carbons; wherein Z is selected from —O— and —NR13—; wherein R13 is selected from hydrogen and C1-C4 alkyl; or, wherein Z is N, R12 and R13 are covalently bonded and —NR12R13 comprises a moiety of the formula:

wherein Y is selected from —O—, —S—, —SO—, —SO2—, —NH—, —NCH3—; and wherein R14 is C1-C6 alkyl and substituted with 0-2 groups selected from cyano, acyl, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, pentyl, hexyl, hydroxyl, acetoxyl, methoxyl, ethoxyl, propoxyl, and butoxyl; or a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof, and one or more of: (a) a protein supplement; (b) an anabolic agent; (c) a catabolic agent; (d) a dietary supplement; (e) at least one agent known to treat a disorder associated with muscle wasting; (f) instructions for treating a disorder associated with cholinergic activity; or (g) instructions for using the compound to increase muscle mass and/or muscular strength.

In a further aspect, the kit ocomprises a disclosed compound or a product of a disclosed method.

In a further aspect, the at least one compound and the at least one agent are co-formulated. In a still further aspect, the at least one compound and the at least one agent are co-packaged.

The kits can also comprise compounds and/or products co-packaged, co-formulated, and/or co-delivered with other components. For example, a drug manufacturer, a drug reseller, a physician, a compounding shop, or a pharmacist can provide a kit comprising a disclosed compound and/or product and another component for delivery to a patient.

It is contemplated that the disclosed kits can be used in connection with the disclosed methods of making, the disclosed methods of using, and/or the disclosed compositions.

8. Non-Medical Uses

Also provided are the uses of the disclosed compounds and products as pharmacological tools in the development and standardization of in vitro and in vivo test systems for the evaluation of the effects of modulators of muscle hypertrophy or inhibitors of muscle atrophy related activity in laboratory animals such as cats, dogs, rabbits, monkeys, rats and mice, as part of the search for new therapeutic agents of increase muscle mass and/or inhibit muscle hypertrophy.

F. EXPERIMENTAL

The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the compounds, compositions, articles, devices and/or methods claimed herein are made and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.

Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in ° C. or is at ambient temperature, and pressure is at or near atmospheric.

Certain materials, reagents and kits were obtained from specific vendors as indicated below, and as appropriate the vendor catalog, part or other number specifying the item are indicated. Vendors indicated below are as follows: “Ambion” is Ambion, a division of Life Technologies Corporation, Austin, Tex., USA; “Applied Biosystems” is Applied Biosystems, a division of Life Technologies Corporation, Carlsbad, Calif., USA; “Boehringer Mannheim” is Boehringer Mannheim Corporatin, Indiapolis, Ind., USA; “CardinalHealth” is Cardinal Health, Inc., Dublin, Ohio, USA; “Cell Signaling” is Cell Signaling Technology, Inc., Beverly, Mass., USA; “Columbus Inst” is Columbus Instruments International, Columbus, Ohio, USA; “Harlan” is Harlan Laboratories, Indianapolis, Ind., USA; “Instrumedics” is Instrumedics, Inc., Richmond, Ill., USA; “Invitrogen” is Invitrogen Corporation, Carlsbad, Calif., USA; “Microm” is the Microm division (Walldorf, Germany) of Thermo Fisher Scientific Inc., Rockford, Ill., USA; “Millipore” is Millipore Corporation, Billerica, Mass., USA; a division of Merck KGaA, Darmstadt, Germany; “Ortho” is Ortho Clinical Diagnostics, Rochester, N.Y., USA; “Pierce” is Pierce Biotechnology, Inc., Milwaukee, Wis., USA, a division of Thermo Fisher Scientific, Inc.; “R&D Systems” is R&D Systems Inc., Minneapolis, Minn., USA; “Roche Diagnostics” is Roche Diagnostics Corporation, Indianapolis, Ind., USA; “Sakura” is Sakura Finetek USA, Inc., Torrance, Calif., USA; “Santa Cruz” is Santa Cruz Biotechnology, Inc., Santa Cruz, Calif., USA; and, “Sigma” is Sigma-Aldrich Corporation, Saint Louis, Mo., USA.

1. General Methods

a. Human Subject Protocol.

The study referred to herein was approved by the Institutional Review Board at the University of Iowa, and involved seven healthy adults who gave their informed consent before participating. One week prior to the fasting study, subjects made one visit to the Clinical Research Unit (“CRU”) for anthropometric measurements, a dietary interview that established each subject's routine food intake and food preferences, and baseline determinations of blood hemoglobin (“Hb”) Al c turbidimetric immunoinhibition using the BM/Hitachi 911 analyzer (Boehringer Mannheim); plasma triglycerides and plasma free T4 and TSH by electrochemiluminescence immunoassay using the Elecsys® System (Roche Diagnostics); plasma CRP by immuno-turbidimetric assay using the Roche Cobas Integra® high-sensitivity assay (Roche Diagnostics); and, plasma TNF-α levels using the Quantikine® Kit (R&D Systems). To ensure that subjects were eating their routine diet prior to the fasting study, subjects ate only meals prepared by the CRU dietician (based on the dietary interview) for 48 hours before the fasting study. The fasting study began at t =0 hours, when subjects were admitted to the CRU and began fasting. While fasting, subjects remained in the CRU and were encouraged to maintain their routine physical activities. Water was allowed ad libitum, but caloric intake was not permitted. At about 40 hours, a percutaneous biopsy was taken from the vastus lateralis muscle using a Temno® Biopsy Needle (CardinalHealth; Cat #T1420) under ultrasound guidance. Subjects then ate a CRU-prepared mixed meal, and at t=46 hours, a muscle biopsy was taken from the contralateral vastus lateralis muscle. Plasma glucose and insulin levels were measured at t=36, 40, 42 and 46 hours; the Elecsys® system was used to quantitate plasma insulin. Our study protocol of humans with spinal cord injury was described previously (Adams C M, et al. (2011) Muscle Nerve. 43(1):65-75).

b. Microarray Analysis of Human Skeletal Muscle Mrna Levels.

Following harvest, skeletal muscle samples were immediately placed in RNAlater (Ambion) and stored at −80° C. until further use. Total RNA was extracted using TRIzol solution (Invitrogen), and microarray hybridizations were performed at the University of Iowa DNA Facility, as described previously (Lamb J, et al. (2006) Science (New York, N.Y. 313(5795):1929-1935). The loge hybridization signals as shown herein reflect the mean signal intensity of all exon probes specific for an individual mRNA. To determine which human skeletal muscle mRNAs were significantly altered by fasting (P≤0.02), paired t-tests were used to compare fasted and fed loge signals. To determine which mouse skeletal muscle mRNAs were significantly altered by ursolic acid (P≤0.005), unpaired t-tests were used to compare loge signals in mice fed control diet or diet supplemented with ursolic acid. Highly expressed mRNAs were defined as those significantly altered mRNAs that were repressed from or induced to a loge signal>8. These raw microarray data from humans and mice have been deposited in NCBI's Gene Expression Omnibus (“GEO”) and are accessible through GEO Series accession numbers GSE28016 and GSE28017, respectively. Exon array studies of the effects of fasting on mouse skeletal muscle, and the effects of spinal cord injury on human skeletal muscle were described previously (Adams C M, et al. (2011) Muscle & nerve 43(1):65-75; Ebert S M, et al. (2010) Molecular Endocrinology 24(4):790-799).

c. Quantitative Real-Time RT-PCR (QPCR).

TRIzol-extracted mRNA was treated with DNase I using the Turbo DNA free kit (Ambion). qPCR analysis of human mRNA and mouse IGF-I mRNA was performed using TaqMan Gene Expression Assays (Applied Biosystems). First strand cDNA was synthesized from 2 μg of RNA using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Part No. 4368814). The real time PCR contained, in a final volume of 20 μl, 20 ng of reverse transcribed RNA, 1 μl of 20× TaqMan Gene Expression Assay, and 10 μl of TaqMan Fast Universal PCR Master Mix (Applied Biosystems; Part No. 4352042). qPCR was carried out using a 7500 Fast Real-Time PCR System (Applied Biosystems) in 9600 emulation mode. qPCR analysis of mouse atrogin-1 and MuRF1 mRNA levels was performed as previously described (Ebert S M, et al. (2010) Molecular Endocrinology 24(4):790-799). All qPCR reactions were performed in triplicate and the cycle threshold (Ct) values were averaged to give the final results. To analyze the data, the ΔCt method was used, with the level of 36B4 mRNA serving as the invariant control.

d. Mouse Protocols.

Male C57BL/6 mice, ages 6-8 weeks, were obtained from NCI, housed in colony cages with 12 h light/12 h dark cycles, and used for experiments within 3 weeks of their arrival. Unless otherwise indicated, mice were maintained on standard chow (Harlan; Teklad Diet, Formula 7013, NIH-31 Modified Open Formula Mouse/Rat Sterilizable Diet). Metformin (Sigma) was dissolved in 0.9% NaCl at a concentration of 250 mg/ml. Ursolic acid (Enzo Life Sciences) was dissolved in corn oil at a concentration of 200 mg/ml (for i.p. injections); alternatively, the ursolic acid was added directly to standard chow (Harlan; Teklad Diet, Formula 7013) or standard high fat diet (Harlan; Teklad Diet, Formula TD. 93075) as a customized chow. Oleanolic acid (Sigma) was dissolved in corn oil at a concentration of 200 mg/ml. Mice were fasted by removing food, but not water, for 24 hours. Fasting blood glucose levels were obtained from the tail vein with an ACCU-CHEK® Aviva glucose meter (Roche Diagnostics). Unilateral hindlimb muscle denervation was performed by transsecting the sciatic nerve under anesthesia, and was followed by administration of ursolic acid (200 mg/kg) or vehicle alone (corn oil) via i.p injection twice daily for 7 days. Forelimb grip strength was determined using a grip strength meter equipped with a triangular pull bar (Columbus Inst). Each mouse was subjected to 5 consecutive tests to obtain the peak value. Plasma IGF-I and leptin levels were measured by RIA at the Vanderbilt University Hormone Assay Core Facility. Plasma cholesterol, triglyceride, creatinine, bilirubin and ALT were measured using the VITROS® 350 Chemistry System (Ortho). All animal procedures were approved by the Institutional Animal Care and Use Committee of the University of Iowa.

e. Histological Analysis.

Following harvest, tissues were immediately placed in isopentane that had been chilled to −160° C. with liquid N2. Muscles were embedded in tissue freezing medium, and 10 μm sections from the mid-belly were prepared using a Microm HM 505 E cryostat equipped with a CryoJane sectioning system (Instrumedics). Adipose tissue was fixed in 10% neutral buffered formalin, embedded in paraffin, and then 4 μm sections were prepared using a Microm HM355 S motorized microtome (Microm). Hematoxylin and eosin stains were performed using a DRS-601 automatic slide stainer (Sakura), and examined on an Olympus IX-71 microscope equipped with a DP-70 camera. Image analysis was performed using ImageJ software (public domain, available from the National Institutes of Health, USA). Muscle fiber diameter was measured using the lesser diameter method, as described elsewhere (Dubowitz V, et al. (2007) Muscle biopsy: a practical approach (Saunders Elsevier, Philadelphia) 3rd Ed pp XIII, 611 s).

f. Analysis of IGF-I and Insulin-Mediated Protein Phosphorylation.

Mouse quadriceps muscles were snap frozen in liquid N2, and Triton-X 100 soluble protein extracts were prepared as described previously (Ebert S M, et al. (2010) Molecular endocrinology 24(4):790-799). Mouse C2C12 myoblasts were obtained from American Type Culture Collection (“ATCC”), and maintained in Dulbecco's modified Eagle's medium (DMEM; ATCC #30-2002) containing antibiotics (100 units/ml penicillin, 100 μg/ml streptomycin sulfate) and 10% (v/v) fetal bovine serum (FBS). On day 0, myotubes were set-up in 6-well plates at a density of 2.5×105 cells/well. On day 2, differentiation into myotubes was induced by replacing 10% FBS with 2% horse serum. On day 7, myotubes were serum-starved by washing 2 times with phosphate buffered saline, and then adding fresh serum-free media. After 16 hours of serum-starvation, 10 μM ursolic acid (from a 10 mM stock prepared in DMSO), or an equal volume of DMSO, with or without 10 nM mouse IGF-I (Sigma; Cat. No. I8779) or 10 nM bovine insulin (Sigma; Cat. No. I6634) was directly added to the media. For analysis of Akt, S6K, ERK and FoxO phosphorylation, myotubes were incubated in the presence or absence of ursolic acid, IGF-I and/or insulin for 20 min, and then harvested into SDS lysis buffer (10 mM Tris-HCl, pH 7.6, 100 mM NaCl, 1% (w/v) SDS, 1 μg/ml pepstatin A, 2 μg/ml aprotonin, 10 μg/ml leupeptin, 200 μM phenylmethylsulfonyl fluoride and a 1:100 dilution of phosphatase inhibitor cocktail 3 (Sigma). An aliquot of each muscle extract or cell lysate was mixed with 0.25 volume of sample buffer (250 mM Tris-HCl, pH 6.8, 10% SDS, 25% glycerol, 0.2% (w/v) bromophenol blue, and 5% (w/v) 2-mercaptoethanol) and heated for 5 min at 95° C., whereas a separate aliquot was used to determine protein concentration by the BCA kit (Pierce). Samples (25 μg) were subjected to 8% SDS-PAGE, then transferred to Hybond-C extra nitrocellulose filters (Millipore). Immunoblots were performed at 4° C. for 16 h using a 1:2000 dilution of antibodies detecting total Akt, phospho-Akt(Ser473), total S6K, phospho-S6K(T421/S424), total ERK1/2, phospho-ERK(T202/Y204), FoxO3a, or phospho-FoxO1(T24)/FoxO3a(T32) (Cell Signaling). For analysis of IGF-1 receptor or insulin receptor phosphorylation, myotubes were incubated in the presence or absence of ursolic acid, IGF-I and/or insulin for 2 min, and then harvested into RIPA buffer (10 mM Tris-HCL, pH 7.4, 150 mM NaCl, 0.1% (w/v) SDS, 1% (w/v) Triton X-100, 1% Na deoxycholate, 5 mM EDTA, 1 mM NaF, 1 mM Na orthovanadate, 1 μg/ml pepstatin A, 2 μg/ml aprotonin, 10 μug/ml leupeptin, 200 μM phenylmethylsulfonyl fluoride, 1:100 dilution of phosphatase inhibitor cocktail 2 (Sigma) and a 1:100 dilution of phosphatase inhibitor cocktail 3 (Sigma). The protein concentration was measured using the BCA kit, after which the extract was diluted to a concentration of 1 mg/ml in RIPA buffer (final volume 500 μl). Then 2 μg anti-IGF-1 receptor β antibody (Cell Signaling) or 2 μg anti-insulin receptor β antibody (Santa Cruz) was added with 50 μl protein G plus Sepharose beads (Santa Cruz), and then the samples were rotated at 4° C. for 16 h. Immunoprecipitates were washed three times for 20 min with 1 ml RIPA buffer and then mixed with 100 μl sample buffer (50 mM Tris-HCl (pH 6.8), 2% SDS, 5% glycerol, 0.04% (w/v) bromophenol blue and 5% (w/v) 2-mercaptoethanol), then boiled for 5 min. Immunoprecipitates were subjected to 8% SDS-PAGE. For analysis of total IGF-1 receptor, phospho-insulin receptor and total insulin receptor, proteins were transferred to Hybond-C extra nitrocellulose filters (Millipore). For analysis of phospho-IGF-1 receptor, proteins were transferred to PVDF membranes (Bio-Rad). Immunoblots were performed at room temperature using a 1:2000 dilution of anti-IGF-1 receptor β antibody, 1:5000 dilution of mouse anti-phospho-tyrosine 4G10 monoclonal antibody (Millipore), a 1:2000 dilution of anti-insulin receptor β, or 1:2000 dilution of anti-phospho-insulin receptor β (Y1162/1163) (Santa Cruz).

g. PTP1B Inhibition Via RNA Interference.

The plasmidspCMV-miR-PTP1B #1 and pCMV-miR-PTP1B #2 were generated by ligating PTPN1-specific oligonucleotide duplexes (Invitrogen) into the pcDNA6.2GW/EmGFP miR plasmid (Invitrogen), which contains a CMV promoter driving co-cistronic expression of engineered pre-miRNAs and EmGFP. pCMV-miR-control encodes a non-targeting pre-miRNA hairpin sequence (miR-neg control; Invitrogen) in pcDNA6.2GW/EmGFP miR plasmid. Male C57BL/6 mice were obtained from NCI at ages 6-8 weeks, and used for experiments within 3 weeks of their arrival. Electroporation of mouse tibialis anterior muscles and isolation of skeletal muscle RNA was performed as described previously (Ebert S M, et al. (2010) Molecular endocrinology 24(4):790-799). First strand cDNA was synthesized in a 20 μl reaction that contained 2 μg of RNA, random hexamer primers and components of the High Capacity cDNA reverse transcription kit (Applied Biosystems). qPCR analysis of PTPN1 mRNA levels was performed using a Taqman expression assay as described previously (Ebert S M, et al. (2010) Molecular endocrinology 24(4):790-799). qPCR was carried out using a 7500 Fast Real-Time PCR System (Applied Biosystems). All qPCR reactions were performed in triplicate and the cycle threshold (Ct) values were averaged to give the final results. Fold changes were determined by the ΔCt method, with level of 36B4 mRNA serving as the invariant control. Skeletal muscle sections were prepared and transfected (EmGFP-positive) muscle fibers were identified and measured as described previously (Ebert S M, et al. (2010) Molecular endocrinology 24(4):790-799).

h. Measurement of Serum Ursolic Acid Levels.

Ursolic acid is extracted from serum using a 10:1 mixture of hexane:propanol (recovery>90%), and then conjugated via its carboxylic acid group to 2-(2,3-naphthalimino)ethyl trifluoromethanesulfonate (Invitrogen; Ne-OTf), a moiety that enhances TUV and fluorescence detection. Derivatized samples are then analyzed on a Waters Acquity UPLC equipped with a 100×2.1 mm C18 HSS column with 1.8 μm beads (Waters Part No. 186003533) and a TUV detector.

2. Identification of Therapeutics to Treat Muscle Atrophy

Skeletal muscle atrophy is common and debilitating condition that lacks a pharmacologic therapy. To identify and develop new therapeutic approaches to this pathophysiological condition (FIG. 1), an approach using gene expression signatures to connect small molecules, genes, and disease was used. Briefly, 63 mRNAs were identified that were regulated by fasting in both human and mouse muscle, and 29 mRNAs that were regulated by both fasting and spinal cord injury in human muscle. These two unbiased mRNA expression signatures of muscle atrophy were used to query the Connectivity Map, an algorithm that allows gene signature datasets to be used to find relationships between small molecules, genes, and disease.

Three complimentary studies to characterize global atrophy-associated changes in skeletal muscle mRNA levels in humans and mice were carried. These three studies determined the effects of: A) fasting on human skeletal muscle mRNA levels, B) spinal cord injury (“SCI”) on human skeletal mRNA levels (Adams C M, et al. (2011) Muscle & nerve 43(1):65-75) and C) fasting on mouse skeletal muscle mRNA levels (Ebert S M, et al. (2010) Molecular endocrinology 24(4):790-799). In each study, exon expression arrays were used that quantitated levels of >16,000 mRNAs. Although there were many significant changes in each study, analysis focused on mRNAs whose levels were similarly altered in at least two atrophy models. Thus, by comparing the effects of fasting on human and mouse skeletal muscle, there were two sets of mRNAs identified: a) 31 mRNAs that were increased by fasting in both species, and b) 32 mRNAs that were decreased by fasting in both species. These evolutionarily conserved, fasting-regulated skeletal muscle mRNAs were termed “atrophy signature-1.” Next, the effects of fasting and SCI on human skeletal muscle were determined and two sets of mRNAs were identitied: a) 18 mRNAs that were increased by fasting and SCI, and b) 17 mRNAs that were decreased by fasting and SCI. This second group of mRNAs was termed “atrophy signature-2.” Almost all of the mRNAs in atrophy signatures-1 and -2 have previously uncharacterized roles in normal or atrophied skeletal muscle. It was next hypothesized that pharmacologic compounds whose effects on cellular mRNA levels were opposite to atrophy signatures-1 and -2 might inhibit skeletal muscle atrophy. To identify candidate compounds, the Connectivity Map (Lamb J, et al. (2006) Science (New York, N.Y. 313(5795):1929-1935) was used to compare atrophy signatures-1 and -2 to mRNA expression signatures of >1300 bioactive small molecules. These results identified several predicted inhibitors of human skeletal muscle atrophy, including ursolic acid. As a proof-of-concept of the utility of atrophy signatures-1 and -2 described herein, the effects of ursolic acid were assessed in mice, and surprisingly it was discovered ursolic acid inhibited muscle atrophy and promoted muscle hypertrophy.

3. Effects of Fasting on Skeletal Muscle mRNA Expression in Humans.

Prolonged fasting induces muscle atrophy, but its effects on global mRNA expression in human skeletal muscle were not known heretofore. In order to determine the relationship between global mRNA expression and human skeletal muscle status, seven healthy adult human volunteers (3 male and 4 female) with ages ranging from 25 to 69 years (mean=46 years) were studied. The overall study design is shown in FIG. 2A. The mean body mass index of these subjects (±SEM) was 25±1. Their mean weight was 69.4±4.8 kg. Baseline circulating levels of hemoglobin A1c (HbA1c), triglycerides (TG), thyroid-stimulating hormone (TSH), free thyroxine (free T4), C-reactive protein (CRP) and tumor necrosis factor-α (TNF-α) were within normal limits (FIG. 2A). The table (FIG. 2A, insert) shows baseline circulating metabolic and inflammatory markers. The graph shows plasma glucose and insulin levels (FIG. 2A). Data are means±SEM from the seven study subjects. In some cases, the error bars are too small to see. While staying in the University of Iowa Clinical Research Unit, the subjects fasted for 40 h by forgoing food but not water. The mean weight loss during the fast was 1.7±0.1 kg (3±0% of the initial body weight).

After the 40 h fast, a muscle biopsy was obtained from the subjects' vastus lateralis (VL) muscle. Immediately after the muscle biopsy, the subjects ate a mixed meal. Five hours later (six hours after the first biopsy), a second muscle biopsy from their contralateral VL muscle. Thus, each subject had a muscle biopsy under fasting and nonfasting conditions. As expected, plasma glucose and insulin levels were low at the end of the 40 h fast, rose after the meal, and returned to baseline by the time of the second biopsy (FIG. 2A). These data indicate comparable levels of plasma glucose and insulin at the times of the first (fasting) and second (nonfasting) muscle biopsies.

To determine the effect of fasting on skeletal muscle mRNA expression, RNA was isolated from the paired muscle biopsies and then analyzed it with exon expression arrays. Using P≤0.02 (by paired t-test) as criteria for statistical significance, it was found that 281 mRNAs were higher in the fasting state and 277 were lower (out of >17,000 mRNAs measured; see FIG. 2B). A complete list of these fasting-responsive mRNAs is shown below in Table 1 (“Change” is the mean loge change or difference between fasting and fed states). The data in Table 1 is for all mRNAs in this study whose levels were increased or decreased by fasting (P≤0.02 by paired t-test).

Representative fasting-responsive human skeletal muscle mRNAs, and the effect of fasting on their log2 hybridization signals, as assessed by Affymetrix Human Exon 1.0 ST arrays are shown in FIG. 2B. In each subject, the fasting signal was normalized to the nonfasting signal from the same subject. Data are means±SEM from 7 subjects. P≤0.02 by paired t-test for all mRNAs shown. The complete set of 458 fasting-responsive mRNAs is shown in Table 1. Most of the differentially expressed mRNAs identified as altered by fasting surprisingly did not have previously known roles in muscle atrophy. However, fasting increased several mRNAs that encode proteins with known roles in catabolic processes such as fat oxidation, reverse cholesterol transport, thermogenesis, inhibition of protein synthesis, autophagy, ubiquitin-mediated proteolysis, glutamine transport and heme catabolism (FIG. 2B). Of these, atrogin-1, MuRF1 and ZFAND5 mRNAs encode proteins known to be required for skeletal muscle atrophy in mice (Bodine S C, et al. (2001) Science (New York, N.Y. 294(5547):1704-1708; Hishiya A, et al. (2006) The EMBO journal 25(3):554-564). Conversely, fasting significantly decreased several mRNAs encoding proteins with known roles in anabolic processes such as glycogen synthesis, lipid synthesis and uptake, polyamine synthesis, iron uptake, angiogenesis, and mitochondrial biogenesis (FIG. 2B). Of these, PGC-1α mRNA encodes a protein that inhibits atrophy-associated gene expression and skeletal muscle atrophy in mice (Sandri M, et al. (2006) Proceedings of the National Academy of Sciences of the United States of America 103(44):16260-16265).

The results were further validated using qPCR to to analyze RNA from paired fed and fasted skeletal muscle biopsy samples obtained from seven healthy human subjects (see FIG. 3; data are means±SEM; *P≤0.01 by paired t-test.). In each subject, the fasting mRNA level was normalized to the nonfasting level, which was set at 1. The mRNA encoding myostatin (MSTN) is a control transcript whose level was not altered by fasting, as assessed by exon expression arrays. Taken together, these data established an mRNA expression signature of fasting in human skeletal muscle.

TABLE 1 FASTING-RESPONSIVE HUMAN MRNAS. Change Affymetrix Gene Accession (Fasting − ID mRNA Assignment No. Fed) SEM P 3062082 PDK4 NM_002612 // NM_002612 2.15 0.34 0.000 PDK4 // pyruvate dehydrogenase kinase, isozyme 4 // 7q21.3 // 5166 2319340 SLC25A33 NM_032315 // NM_032315 1.42 0.41 0.007 SLC25A33 // solute carrier family 25, member 33 // 1p36.22 // 84275 3165957 IFNK NM_020124 // NM_020124 0.96 0.28 0.007 IFNK // interferon, kappa // — // 56832 /// ENST00000276943 // IF 3424158 MYF6 NM_002469 // NM_002469 0.95 0.12 0.000 MYF6 // myogenic factor 6 (herculin) // 12q21 // 4618 /// ENST00000 3422144 LGR5 NM_003667 // NM_003667 0.88 0.12 0.000 LGR5 // leucine- rich repeat- containing G protein-coupled receptor 5 2356115 TXNIP NM_006472 // NM_006472 0.85 0.22 0.004 TXNIP // thioredoxin interacting protein // 1q21.1 // 10628 /// ENS 3233605 PFKFB3 NM_004566 // NM_004566 0.84 0.18 0.002 PFKFB3 // 6- phosphofructo-2- kinase/fructose-2,6- biphosphatase 3 // 3151607 FBXO32 NM_058229 // NM_058229 0.82 0.19 0.002 FBXO32 // F-box protein 32 // 8q24.13 // 114907 /// NM_148177 // FB 2745547 GAB1 NM_207123 // NM_207123 0.71 0.08 0.000 GAB1 // GRB2- associated binding protein 1 // 4q31.21 // 2549 /// NM 3173479 FOXD4L3 NM_199135 // NM_199135 0.68 0.25 0.017 FOXD4L3 // forkhead box D4- like 3 // 9q13 // 286380 /// NM_012184/ 3199500 CER1 NM_005454 // NM_005454 0.64 0.24 0.019 CER1 // cerberus 1, cysteine knot superfamily, homolog (Xenopus lae 3444309 TAS2R9 NM_023917 // NM_023917 0.63 0.22 0.015 TAS2R9 // taste receptor, type 2, member 9 // 12p13 // 50835 /// EN 3452323 SLC38A2 NM_018976 // NM_018976 0.62 0.13 0.001 SLC38A2 // solute carrier family 38, member 2 // 12q // 54407 /// E 3381843 UCP3 NM_003356 // NM_003356 0.59 0.04 0.000 UCP3 // uncoupling protein 3 (mitochondrial, proton carrier) // 11q 3147508 KLF10 NM_005655 // NM_005655 0.58 0.11 0.001 KLF10 // Kruppel- like factor 10 // 8q22.2 // 7071 /// NM_001032282 3982534 LPAR4 NM_005296 // NM_005296 0.57 0.17 0.008 LPAR4 // lysophosphatidic acid receptor 4 // Xq13-q21.1 // 2846 /// 3384321 RAB30 NM_014488 // NM_014488 0.56 0.21 0.019 RAB30 // RAB30, member RAS oncogene family // 11q12-q14 // 27314 // 3256192 C10orf116 NM_006829 // NM_006829 0.55 0.19 0.013 C10orf116 // chromosome 10 open reading frame 116 // 10q23.2 // 109 2705690 GHSR NM_198407 // NM_198407 0.54 0.20 0.016 GHSR // growth hormone secretagogue receptor // 3q26.31 // 2693 /// 3326938 LOC100130104 AF274942 // AF274942 0.53 0.16 0.009 LOC100130104 // PNAS-17 // 11p13 // 100130104 2318656 PER3 NM_016831 // NM_016831 0.52 0.16 0.009 PER3 // period homolog 3 (Drosophila) // 1p36.23 // 8863 /// ENST00 3209623 ZFAND5 NM_001102420 // NM_001102420 0.51 0.13 0.005 ZFAND5 // zinc finger, AN1-type domain 5 // 9q13- q21 // 7763 /// 3741300 OR1D4 NM_003552 // NM_003552 0.50 0.19 0.019 OR1D4 // olfactory receptor, family 1, subfamily D, member 4 // 17p 2899176 HIST1H2BD NM_138720 // NM_138720 0.49 0.16 0.010 HIST1H2BD // histone cluster 1, H2bd // 6p21.3 // 3017 /// NM_02106 3439256 RPS11 ENST00000270625 // ENST00000270625 0.49 0.11 0.002 RPS11 // ribosomal protein S11 // 19q13.3 // 6205 /// BC10002 2973232 KIAA0408 NM_014702 // NM_014702 0.49 0.14 0.006 KIAA0408 // KIAA0408 // 6q22.33 // 9729 /// NM_001012279 // C6orf17 3291151 RHOBTB1 NM_014836 // NM_014836 0.48 0.09 0.001 RHOBTB1 // Rho- related BTB domain containing 1 // 10q21.2 // 9886/ 2358136 C1orf51 BC027999 // BC027999 0.48 0.17 0.016 C1orf51 // chromosome 1 open reading frame 51 // 1q21.2 // 148523 // 3948936 0.47 0.18 0.020 3944129 HMOX1 NM_002133 // NM_002133 0.46 0.13 0.006 HMOX1 // heme oxygenase (decycling) 1 // 22q12|22q13.1 // 3162 /// 2968652 SESN1 NM_014454 // NM_014454 0.46 0.12 0.004 SESN1 // sestrin 1 // 6q21 // 27244 /// ENST00000302071 // SESN1 // 2951881 PXT1 NM_152990 // NM_152990 0.45 0.14 0.008 PXT1 // peroxisomal, testis specific 1 // 6p21.31 // 222659 /// ENS 2819747 POLR3G NM_006467 // NM_006467 0.45 0.13 0.007 POLR3G // polymerase (RNA) III (DNA directed) polypeptide G (32 kD) 2957384 GSTA2 NM_000846 // NM_000846 0.44 0.10 0.002 GSTA2 // glutathione S- transferase A2 // 6p12.1 // 2939 /// NM_1536 4014387 RPSA NM_002295 // NM_002295 0.44 0.16 0.018 RPSA // ribosomal protein SA // 3p22.2 // 3921 /// NM_001012321 // 3021158 C7orf58 NM_024913 // NM_024913 0.44 0.07 0.000 C7orf58 // chromosome 7 open reading frame 58 // 7q31.31 // 79974/ 2976155 OLIG3 NM_175747 // NM_175747 0.44 0.12 0.006 OLIG3 // oligodendrocyte transcription factor 3 // 6q23.3 // 167826 3261886 C10orf26 NM_017787 // NM_017787 0.44 0.17 0.019 C10orf26 // chromosome 10 open reading frame 26 // 10q24.32 // 5483 2489169 0.42 0.12 0.006 2790062 TMEM154 NM_152680 // NM_152680 0.42 0.14 0.012 TMEM154 // transmembrane protein 154 // 4q31.3 // 201799 /// ENST00 3792656 CCDC102B NM_024781 // NM_024781 0.42 0.12 0.007 CCDC102B // coiled-coil domain containing 102B // 18q22.1 // 79839 3554282 INF2 NM_022489 // NM_022489 0.41 0.14 0.012 INF2 // inverted formin, FH2 and WH2 domain containing // 14q32.33 2614142 NR1D2 NM_005126 // NM_005126 0.39 0.15 0.019 NR1D2 // nuclear receptor subfamily 1, group D, member 2 // 3p24.2 3404636 GABARAPL1 NM_031412 // NM_031412 0.39 0.10 0.004 GABARAPL1 // GABA(A) receptor- associated protein like 1 // 12p13.2 3063856 tcag7.1177 ENST00000292369 // ENST00000292369 0.39 0.09 0.003 tcag7.1177 // opposite strand transcription unit to STAG3 // 3461981 TSPAN8 NM_004616 // NM_004616 0.39 0.14 0.015 TSPAN8 // tetraspanin 8 // 12q14.1-q21.1 // 7103 /// ENST0000039333 2908154 C6orf206 BC029519 // BC029519 0.39 0.09 0.003 C6orf206 // chromosome 6 open reading frame 206 // 6p21.1 // 221421 3415046 FLJ33996 AK091315 // AK091315 0.39 0.15 0.019 FLJ33996 // hypothetical protein FLJ33996 // 12q13.13 // 283401 /// 3326400 CAT NM_001752 // NM_001752 0.39 0.09 0.003 CAT //catalase // 11p13 // 847 /// ENST00000241052 // CAT // catal 2390322 OR2M5 NM_001004690 // NM_001004690 0.38 0.12 0.011 OR2M5 // olfactory receptor, family 2, subfamily M, member 5 // 2402536 TRIM63 NM_032588 // NM_032588 0.38 0.12 0.009 TRIM63 // tripartite motif-containing 63 // 1p34-p33 // 84676 /// E 2976768 CITED2 NM_006079 // NM_006079 0.37 0.10 0.005 CITED2 // Cbp/p300- interacting transactivator, with Glu/Asp-rich ca 3218528 ABCA1 NM_005502 // NM_005502 0.37 0.14 0.016 ABCA1 // ATP- binding cassette, sub-family A (ABC1), member 1 // 9q3 3377861 DKFZp761E198 NM_138368 // NM_138368 0.37 0.06 0.000 DKFZp761E198 // DKFZp761E198 protein // 11q13.1 // 91056 /// BC1091 2961347 FILIP1 NM_015687 // NM_015687 0.37 0.10 0.005 FILIP1 // filamin A interacting protein 1 // 6q14.1 // 27145 /// EN 3097580 C8orf22 NM_001007176 // NM_001007176 0.37 0.08 0.002 C8orf22 // chromosome 8 open reading frame 22 // 8q11 // 492307 3755655 FBXL20 NM_032875 // NM_032875 0.35 0.08 0.002 FBXL20 // F-box and leucine-rich repeat protein 20 // 17q12 // 8496 3057505 CCL26 NM_006072 // NM_006072 0.35 0.12 0.012 CCL26 // chemokine (C-C motif) ligand 26 // 7q11.23 // 10344 /// EN 3307795 C10orf118 NM_018017 // NM_018017 0.35 0.13 0.020 C10orf118 // chromosome 10 open reading frame 118 // 10q25.3 // 550 3654699 NUPR1 NM_001042483 // NM_001042483 0.35 0.10 0.007 NUPR1 // nuclear protein 1 // 16p11.2 // 26471 /// NM_012385 // 3778252 ANKRD12 NM_015208 // NM_015208 0.34 0.08 0.002 ANKRD12 // ankyrin repeat domain 12 // 18p11.22 // 23253 /// NM_001 2662560 C3orf24 NM_173472 // NM_173472 0.34 0.08 0.002 C3orf24 // chromosome 3 open reading frame 24 // 3p25.3 // 115795/ 3896370 RP5-1022P6.2 NM_019593 // NM_019593 0.34 0.10 0.007 RP5-1022P6.2 // hypothetical protein KIAA1434 // 20p12.3 // 56261/ 3389566 KBTBD3 NM_198439 // NM_198439 0.34 0.08 0.003 KBTBD3 // kelch repeat and BTB (POZ) domain containing 3 // 11q22.3 3247818 FAM133B NM_152789 // NM_152789 0.34 0.11 0.010 FAM133B // family with sequence similarity 133, member B // 7q21.2 2457988 ZNF706 AF275802 // AF275802 0.34 0.12 0.016 ZNF706 // zinc finger protein 706 // 8q22.3 // 51123 /// BC015925 // 3525234 IRS2 NM_003749 // NM_003749 0.34 0.09 0.004 IRS2 // insulin receptor substrate 2 // 13q34 // 8660 /// ENST00000 2730281 ODAM NM_017855 // NM_017855 0.34 0.12 0.016 ODAM // odontogenic, ameloblast asssociated // 4q13.3 // 54959 /// 3768969 ABCA5 NM_018672 // NM_018672 0.33 0.10 0.008 ABCA5 // ATP- binding cassette, sub-family A (ABC1), member 5 // 17q 3687494 MAPK3 NM_001040056 // NM_001040056 0.33 0.09 0.004 MAPK3 // mitogen- activated protein kinase 3 // 16p11.2 // 5595/ 3405396 CREBL2 NM_001310 // NM_001310 0.33 0.07 0.002 CREBL2 // cAMP responsive element binding protein-like 2 // 12p13/ 3647504 PMM2 NM_000303 // NM_000303 0.33 0.10 0.008 PMM2 // phosphomannomutase 2 // 16p13.3- p13.2 // 5373 /// ENST00000 3392840 BUD13 NM_032725 // NM_032725 0.33 0.07 0.002 BUD13 // BUD13 homolog (S. cerevisiae) // 11q23.3 // 84811 /// ENST 3453837 TUBA1A NM_006009 // NM_006009 0.33 0.07 0.002 TUBA1A // tubulin, alpha 1a // 12q12- q14.3 // 7846 /// ENST00000301 2409310 ELOVL1 NM_022821 // NM_022821 0.32 0.09 0.005 ELOVL1 // elongation of very long chain fatty acids (FEN1/Elo2, SUR 3837707 ZNF114 NM_153608 // NM_153608 0.31 0.09 0.007 ZNF114 // zinc finger protein 114 // 19q13.32 // 163071 /// ENST000 3504434 XPO4 NM_022459 // NM_022459 0.31 0.10 0.009 XPO4 // exportin 4 // 13q11 // 64328 /// ENST00000255305 // XPO4 // 2431877 0.31 0.11 0.017 3837836 PSCD2 NM_017457 // NM_017457 0.31 0.05 0.000 PSCD2 // pleckstrin homology, Sec7 and coiled-coil domains 2 (cytoh 3869396 ZNF432 NM_014650 // NM_014650 0.31 0.09 0.006 ZNF432 // zinc finger protein 432 // 19q13.33 // 9668 /// ENST00000 3981120 OGT NM_181672 // NM_181672 0.31 0.10 0.013 OGT // O-linked N- acetylglucosamine (GlcNAc) transferase (UDP- N-ace 2622607 SLC38A3 NM_006841 // NM_006841 0.30 0.11 0.016 SLC38A3 // solute carrier family 38, member 3 // 3p21.3 // 10991 // 3978812 FOXR2 NM_198451 // NM_198451 0.30 0.09 0.008 FOXR2 // forkhead box R2 // Xp11.21 // 139628 /// ENST00000339140/ 3571904 NPC2 NM_006432 // NM_006432 0.30 0.10 0.011 NPC2 // Niemann- Pick disease, type C2 // 14q24.3 // 10577 /// NM_00 2417945 PTGER3 NM_198715 // NM_198715 0.30 0.11 0.017 PTGER3 // prostaglandin E receptor 3 (subtype EP3) // 1p31.2 // 573 3059393 SEMA3E NM_012431 // NM_012431 0.30 0.09 0.009 SEMA3E // sema domain, immunoglobulin domain (Ig), short basic doma 2336456 MGC52498 NM_001042693 // NM_001042693 0.30 0.10 0.011 MGC52498 // hypothetical protein MGC52498 // 1p32.3 // 348378 // 3726772 CROP NM_016424 // NM_016424 0.30 0.11 0.016 CROP // cisplatin resistance- associated overexpressed protein // 17 2784265 IL2 NM_000586 // IL2 // NM_000586 0.29 0.11 0.019 interleukin 2 // 4q26-q27 // 3558 /// ENST00000226730 // IL2 2495782 LIPT1 NM_145197 // NM_145197 0.29 0.10 0.012 LIPT1 // lipoyltransferase 1 // 2q11.2 // 51601 /// NM_145198 // LI 2377094 PFKFB2 NM_006212 // NM_006212 0.29 0.10 0.012 PFKFB2 // 6- phosphofructo-2- kinase/fructose-2,6- biphosphatase 2 // 2469213 KLF11 NM_003597 // NM_003597 0.29 0.10 0.011 KLF11 // Kruppel- like factor 11 // 2p25 // 8462 /// ENST00000305883 3662387 HERPUD1 NM_014685 // NM_014685 0.29 0.07 0.003 HERPUD1 // homocysteine- inducible, endoplasmic reticulum stress-ind 3771215 ACOX1 NM_004035 // NM_004035 0.29 0.10 0.013 ACOX1 // acyl- Coenzyme A oxidase 1, palmitoyl // 17q24- q25|17q25.1 3203135 TOPORS NM_005802 // NM_005802 0.28 0.11 0.018 TOPORS // topoisomerase I binding, arginine/serine- rich // 9p21 // 2805482 0.28 0.09 0.008 3247757 UBE2D1 NM_003338 // NM_003338 0.28 0.08 0.007 UBE2D1 // ubiquitin- conjugating enzyme E2D 1 (UBC4/5 homolog, yeast 3444147 KLRC1 NM_002259 // NM_002259 0.28 0.10 0.015 KLRC1 // killer cell lectin-like receptor subfamily C, member 1 // 3348891 C11orf57 NM_018195 // NM_018195 0.28 0.09 0.011 C11orf57 // chromosome 11 open reading frame 57 // 11q23.1 // 55216 3906942 SERINC3 NM_006811 // NM_006811 0.28 0.07 0.003 SERINC3 // serine incorporator 3 // 20q13.1-q13.3 // 10955 /// NM_1 2930418 UST NM_005715 // UST // NM_005715 0.28 0.06 0.002 uronyl-2- sulfotransferase // 6q25.1 // 10090 /// ENST0000036 3188200 OR1L1 NM_001005236 // NM_001005236 0.28 0.09 0.011 OR1L1 // olfactory receptor, family 1, subfamily L, member 1 // 3856075 ZNF682 NM_033196 // NM_033196 0.28 0.10 0.017 ZNF682 // zinc finger protein 682 // 19p12 // 91120 /// NM_00107734 3385951 NOX4 NM_016931 // NM_016931 0.28 0.06 0.002 NOX4 // NADPH oxidase 4 // 11q14.2-q21 // 50507 /// ENST00000263317 3523881 KDELC1 NM_024089 // NM_024089 0.28 0.06 0.002 KDELC1 // KDEL (Lys-Asp-Glu-Leu) containing 1 // 13q33 // 79070 /// 2632778 EPHA6 NM_001080448 // NM_001080448 0.28 0.09 0.010 EPHA6 // EPH receptor A6 // 3q11.2 // 285220 /// ENST00000389672 3373272 OR5W2 NM_001001960 // NM_001001960 0.28 0.10 0.015 OR5W2 // olfactory receptor, family 5, subfamily W, member 2 // 4017694 IRS4 NM_003604 // NM_003604 0.28 0.10 0.016 IRS4 // insulin receptor substrate 4 // Xq22.3 // 8471 /// ENST0000 3545311 KIAA1737 NM_033426 // NM_033426 0.28 0.07 0.003 KIAA1737 // KIAA1737 // 14q24.3 // 85457 /// ENST00000361786 // KIA 3753860 CCL5 NM_002985 // NM_002985 0.28 0.05 0.001 CCL5 // chemokine (C-C motif) ligand 5 // 17q11.2-q12 // 6352 /// E 3617312 SLC12A6 NM_001042496 // NM_001042496 0.27 0.07 0.005 SLC12A6 // solute carrier family 12 (potassium/chloride transpor 3351315 UBE4A NM_004788 // NM_004788 0.27 0.07 0.004 UBE4A // ubiquitination factor E4A (UFD2 homolog, yeast) // 11q23.3 3755396 CCDC49 NM_017748 // NM_017748 0.27 0.09 0.013 CCDC49 // coiled- coil domain containing 49 // 17q12 // 54883 /// EN 2870889 C5orf13 NM_004772 // NM_004772 0.27 0.09 0.010 C5orf13 // chromosome 5 open reading frame 13 // 5q22.1 // 9315 /// 2775259 RASGEF1B NM_152545 // NM_152545 0.27 0.10 0.015 RASGEF1B // RasGEF domain family, member 1B // 4q21.21- q21.22 // 15 3165624 0.27 0.06 0.003 2771654 CENPC1 NM_001812 // NM_001812 0.27 0.09 0.013 CENPC1 // centromere protein C 1 // 4q12-q13.3 // 1060 /// ENST0000 3784670 C18orf21 NM_031446 // NM_031446 0.27 0.08 0.008 C18orf21 // chromosome 18 open reading frame 21 // 18q12.2 // 83608 2364231 DDR2 NM_001014796 // NM_001014796 0.26 0.10 0.018 DDR2 // discoidin domain receptor tyrosine kinase 2 // 1q23.3 // 3921442 SH3BGR NM_007341 // NM_007341 0.26 0.08 0.007 SH3BGR // SH3 domain binding glutamic acid-rich protein // 21q22.3 2627368 C3orf49 BC015210 // BC015210 0.26 0.06 0.003 C3orf49 // chromosome 3 open reading frame 49 // 3p14.1 // 132200 3250699 EIF4EBP2 NM_004096 // NM_004096 0.26 0.10 0.018 EIF4EBP2 // eukaryotic translation initiation factor 4E binding pro 3237788 PLXDC2 NM_032812 // NM_032812 0.26 0.09 0.013 PLXDC2 // plexin domain containing 2 // 10p12.32- p12.31 // 84898 // 3285926 ZNF33B NM_006955 // NM_006955 0.26 0.10 0.018 ZNF33B // zinc finger protein 33B // 10q11.2 // 7582 /// ENST000003 3304475 ARL3 NM_004311 // NM_004311 0.26 0.08 0.008 ARL3 // ADP- ribosylation factor- like 3 // 10q23.3 // 403 /// ENST00 3364306 SOX6 NM_017508 // NM_017508 0.26 0.08 0.010 SOX6 // SRY (sex determining region Y)-box6 // 11p15.3 // 55553 // 3185498 SLC31A2 NM_001860 // NM_001860 0.25 0.09 0.015 SLC31A2 // solute carrier family 31 (copper transporters), member 2 3998766 KAL1 NM_000216 // NM_000216 0.25 0.07 0.006 KAL1 // Kallmann syndrome 1 sequence // Xp22.32 // 3730 /// ENST000 3143266 PSKH2 NM_033126 // NM_033126 0.25 0.07 0.006 PSKH2 // protein serine kinase H2 // 8q21.2 // 85481 /// ENST000002 3458911 CTDSP2 NM_005730 // NM_005730 0.25 0.06 0.003 CTDSP2 // CTD (carboxy-terminal domain, RNA polymerase II, polypept 3195034 PTGDS NM_000954 // NM_000954 0.25 0.08 0.010 PTGDS // prostaglandin D2 synthase 21 kDa (brain) // 9q34.2- q34.3 // 3854066 C19orf42 NM_024104 // NM_024104 0.25 0.08 0.010 C19orf42 // chromosome 19 open reading frame 42 // 19p13.11 // 7908 3819474 ANGPTL4 NM_139314 // NM_139314 0.25 0.06 0.004 ANGPTL4 // angiopoietin-like 4 // 19p13.3 // 51129 /// NM_001039667 3944084 TOM1 NM_005488 // NM_005488 0.25 0.07 0.006 TOM1 // target of myb1 (chicken) // 22q13.1 // 10043 /// ENST000003 3848243 INSR NM_000208 // NM_000208 0.24 0.09 0.014 INSR // insulin receptor // 19p13.3- p13.2 // 3643 /// NM_001079817 3168415 CLTA NM_007096 // NM_007096 0.24 0.08 0.009 CLTA // clathrin, light chain (Lca) // 9p13 // 1211 /// NM_00107667 2609462 CAV3 NM_033337 // NM_033337 0.24 0.07 0.007 CAV3 // caveolin 3 // 3p25 // 859 /// NM_001234 // CAV3 // caveolin 3393834 C11orf60 BC022856 // BC022856 0.24 0.06 0.003 C11orf60 // chromosome 11 open reading frame 60 // 11q23.3 // 56912 3755614 STAC2 NM_198993 // NM_198993 0.24 0.07 0.009 STAC2 // SH3 and cysteine rich domain 2 // 17q12 // 342667 /// ENST 3627363 NARG2 NM_024611 /// NM_024611 0.24 0.06 0.003 NARG2 // NMDA receptor regulated 2 // 15q22.2 // 79664 /// NM_00101 3212976 ZCCHC6 NM_024617 // NM_024617 0.24 0.08 0.014 ZCCHC6 // zinc finger, CCHC domain containing 6 // 9q21 // 79670 // 3275922 PRKCQ NM_006257 // NM_006257 0.24 0.05 0.002 PRKCQ // protein kinase C, theta // 10p15 // 5588 /// ENST000002631 3023825 C7orf45 BC017587 // BC017587 0.23 0.09 0.020 C7orf45 // chromosome 7 open reading frame 45 // 7q32.2 // 136263 // 3832906 IL29 NM_172140 // NM_172140 0.23 0.08 0.015 IL29 // interleukin 29 (interferon, lambda 1) // 19q13.13 // 282618 3529156 NGDN NM_015514 // NM_015514 0.23 0.08 0.012 NGDN // neuroguidin, EIF4E binding protein // 14q11.2 // 25983 /// 2620448 CLEC3B NM_003278 // NM_003278 0.23 0.08 0.014 CLEC3B // C-type lectin domain family 3, member B // 3p22-p21.3 // 3481296 SGCG NM_000231 // NM_000231 0.23 0.09 0.019 SGCG // sarcoglycan, gamma (35 kDa dystrophin- associated glycoprotei 3135184 RB1CC1 NM_014781 // NM_014781 0.23 0.07 0.008 RB1CC1 // RB1- inducible coiled- coil 1 // 8q11 // 9821 /// NM_001083 2421843 GBP3 NM_018284 // NM_018284 0.23 0.06 0.004 GBP3 // guanylate binding protein 3 // 1p22.2 // 2635 /// ENST00000 3385003 CREBZF NM_001039618 // NM_001039618 0.23 0.09 0.020 CREBZF // CREB/ATF bZIP transcription factor // 11q14 // 58487/ 3610804 IGF1R NM_000875 // NM_000875 0.23 0.08 0.013 IGF1R // insulin- like growth factor 1 receptor // 15q26.3 // 3480/ 3606304 AKAP13 NM_006738 // NM_006738 0.23 0.04 0.000 AKAP13 // A kinase (PRKA) anchor protein 13 // 15q24-q25 // 11214/ 2565579 ANKRD39 NM_016466 // NM_016466 0.23 0.05 0.003 ANKRD39 // ankyrin repeat domain 39 // 2q11.2 // 51239 /// ENST0000 2722151 RBPJ NM_005349 // NM_005349 0.22 0.07 0.008 RBPJ // recombination signal binding protein for immunoglobulin kap 3031533 GIMAP4 NM_018326 // NM_018326 0.22 0.08 0.017 GIMAP4 // GTPase, IMAP family member 4 // 7q36.1 // 55303 /// ENST0 3725481 UBE2Z NM_023079 // NM_023079 0.22 0.06 0.004 UBE2Z // ubiquitin- conjugating enzyme E2Z // 17q21.32 // 65264 /// 3549575 IFI27 NM_005532 // NM_005532 0.22 0.08 0.016 IFI27 // interferon, alpha-inducible protein 27 // 14q32 // 3429 // 3725035 NFE2L1 NM_003204 // NM_003204 0.22 0.07 0.011 NFE2L1 // nuclear factor (erythroid- derived 2)-like 1 // 17q21.3 // 3348748 C11orf1 NM_022761 // NM_022761 0.22 0.07 0.008 C11orf1 // chromosome 11 open reading frame 1 // 11q13-q22 // 64776 3722039 RAMP2 NM_005854 // NM_005854 0.22 0.05 0.003 RAMP2 // receptor (G protein-coupled) activity modifying protein 2 3886704 STK4 NM_006282 // NM_006282 0.22 0.07 0.012 STK4 // serine/threonine kinase 4 // 20q11.2- q13.2 // 6789 /// ENST 3645901 FLJ14154 NM_024845/ NM_024845 0.22 0.06 0.005 FLJ14154 // hypothetical protein FLJ14154 // 16p13.3 // 79903 /// N 3367673 MPPED2 NM_001584 // NM_001584 0.22 0.08 0.017 MPPED2 // metallophosphoesterase domain containing 2 // 11p13 // 74 3219885 PTPN3 NM_002829 // NM_002829 0.22 0.05 0.003 PTPN3 // protein tyrosine phosphatase, non- receptor type 3 // 9q31 3791466 0.22 0.06 0.007 3717635 ZNF207 NM_001098507 // NM_001098507 0.22 0.08 0.015 ZNF207 // zinc finger protein 207 // 17q11.2 // 7756 /// NM_0034 2648141 MBNL1 NM_021038 // NM_021038 0.22 0.07 0.009 MBNL1 // muscleblind-like (Drosophila) // 3q25 // 4154 /// NM_20729 2436938 PBXIP1 NM_020524 // NM_020524 0.21 0.05 0.002 PBXIP1 // pre-B- cell leukemia homeobox interacting protein 1 // 1q2 3299705 PANK1 NM_148977 // NM_148977 0.21 0.06 0.007 PANK1 // pantothenate kinase 1 // 10q23.31 // 53354 /// NM_148978/ 3628923 FAM96A NM_032231 // NM_032231 0.21 0.05 0.003 FAM96A // family with sequence similarity 96, member A // 15q22.31 2353669 CD2 NM_001767 // CD2 // NM_001767 0.21 0.06 0.006 CD2 molecule // 1p13 // 914 /// ENST00000369478 // CD2 // CD 3474450 PLA2G1B NM_000928 // NM_000928 0.21 0.08 0.016 PLA2G1B // phospholipase A2, group IB (pancreas) // 12q23-q24.1 // 3722417 NBR1 NM_031858 // NM_031858 0.21 0.08 0.017 NBR1 // neighbor of BRCA1 gene 1 // 17q21.31 // 4077 /// NM_005899 3234760 CUGBP2 NM_001025077 // NM_001025077 0.21 0.06 0.004 CUGBP2 // CUG triplet repeat, RNA binding protein 2 // 10p13 // 3627422 RORA NM_134260 // NM_134260 0.21 0.06 0.006 RORA // RAR- related orphan receptor A // 15q21-q22 // 6095 /// NM_0 3382061 XRRA1 NM_182969 // NM_182969 0.21 0.08 0.017 XRRA1 // X-ray radiation resistance associated 1 // 11q13.4 // 1435 3015338 STAG3 NM_012447 // NM_012447 0.21 0.06 0.007 STAG3 // stromal antigen 3 // 7q22.1 // 10734 /// ENST00000317296/ 2665720 ZNF385D NM_024697 // NM_024697 0.21 0.07 0.013 ZNF385D // zinc finger protein 385D // 3p24.3 // 79750 /// ENST0000 3154185 TMEM71 NM_144649 // NM_144649 0.21 0.06 0.009 TMEM71 // transmembrane protein 71 // 8q24.22 // 137835 /// ENST000 3789947 NEDD4L NM_015277 // NM_015277 0.21 0.08 0.016 NEDD4L // neural precursor cell expressed, developmentally down-reg 2688933 CD200R2 ENST00000383679 // ENST00000383679 0.21 0.08 0.016 CD200R2 // CD200 cell surface glycoprotein receptor isoform 2 3379644 CPT1A NM_001876 // NM_001876 0.21 0.04 0.001 CPT1A // carnitine palmitoyltransferase 1A (liver) // 11q13.1-q13.2 3677795 CREBBP NM_004380 // NM_004380 0.21 0.05 0.004 CREBBP // CREB binding protein (Rubinstein-Taybi syndrome) // 16p13 2358320 TARS2 NM_025150 // NM_025150 0.21 0.06 0.007 TARS2 // threonyl- tRNA synthetase 2, mitochondrial (putative) // 1q 3228373 TSC1 NM_000368 // NM_000368 0.20 0.06 0.006 TSC1 // tuberous sclerosis 1 // 9q34 // 7248 /// NM_001008567 // TS 3362795 RNF141 NM_016422 // NM_016422 0.20 0.08 0.019 RNF141 // ring finger protein 141 // 11p15.4 // 50862 /// ENST00000 3673684 CDT1 NM_030928 // NM_030928 0.20 0.07 0.015 CDT1 // chromatin licensing and DNA replication factor 1 // 16q24.3 3042881 HOXA7 NM_006896 // NM_006896 0.20 0.02 0.000 HOXA7 // homeobox A7 // 7p15-p14 // 3204 /// ENST00000396347 // HOX 3381817 UCP2 NM_003355 // NM_003355 0.20 0.05 0.005 UCP2 // uncoupling protein 2 (mitochondrial, proton carrier) // 11q 3415068 ANKRD33 NM_182608 // NM_182608 0.20 0.06 0.006 ANKRD33 // ankyrin repeat domain 33 // 12q13.13 // 341405 /// ENST0 3633403 SIN3A NM_015477 // NM_015477 0.20 0.07 0.014 SIN3A // SIN3 homolog A, transcription regulator (yeast) // 15q24.2 3380901 NUMA1 NM_006185 // NM_006185 0.19 0.04 0.002 NUMA1 // nuclear mitotic apparatus protein 1 // 11q13 // 4926 /// E 2598099 BARD1 NM_000465 // NM_000465 0.19 0.07 0.015 BARD1 // BRCA1 associated RING domain 1 // 2q34- q35 // 580 /// ENST 3139722 NCOA2 NM_006540 // NM_006540 0.19 0.06 0.010 NCOA2 // nuclear receptor coactivator 2 // 8q13.3 // 10499 /// ENST 3641871 LINS1 NM_018148 // NM_018148 0.19 0.06 0.013 LINS1 // lines homolog 1 (Drosophila) // 15q26.3 // 55180 /// NM_00 3401217 TULP3 NM_003324 // NM_003324 0.19 0.06 0.008 TULP3 // tubby like protein 3 // 12p13.3 // 7289 /// ENST0000022824 3741997 ANKFY1 NM_016376 // NM_016376 0.19 0.06 0.008 ANKFY1 // ankyrin repeat and FYVE domain containing 1 // 17p13.3 // 2622742 C3orf45 BC028000 // BC028000 0.19 0.06 0.013 C3orf45 // chromosome 3 open reading frame 45 // 3p21.31 // 132228/ 3845352 UQCR NM_006830 // NM_006830 0.19 0.06 0.014 UQCR // ubiquinol- cytochrome c reductase, 6.4 kDa subunit // 19p13.3 3960356 BAIAP2L2 NM_025045 // NM_025045 0.19 0.07 0.018 BAIAP2L2 // BAI1-associated protein 2-like 2 // 22q13.1 // 80115 // 3645947 CLUAP1 NM_015041 // NM_015041 0.19 0.06 0.012 CLUAP1 // clusterin associated protein 1 // 16p13.3 // 23059 /// NM 3835544 ZNF227 NM_182490 // NM_182490 0.18 0.06 0.011 ZNF227 // zinc finger protein 227 // — // 7770 /// ENST0000031304 3368748 FBXO3 NM_033406 // NM_033406 0.18 0.07 0.020 FBXO3 // F-box protein 3 // 11p13 // 26273 /// NM_012175 // FBXO3/ 3621623 ELL3 NM_025165 // NM_025165 0.18 0.05 0.005 ELL3 // elongation factor RNA polymerase II-like 3 // 15q15.3 // 80 3430552 PWP1 NM_007062 // NM_007062 0.18 0.07 0.016 PWP1 // PWP1 homolog (S. cerevisiae) // 12q23.3 // 11137 /// ENST00 2844908 BTNL9 NM_152547 // NM_152547 0.18 0.05 0.005 BTNL9 // butyrophilin-like 9 // 5q35.3 // 153579 /// ENST0000032770 4021508 ZNF280C NM_017666 // NM_017666 0.18 0.07 0.018 ZNF280C // zinc finger protein 280C // Xq25 // 55609 /// ENST000003 2489071 TET3 NM_144993 // NM_144993 0.18 0.04 0.003 TET3 // tet oncogene family member 3 // 2p13.1 // 200424 /// ENST00 2516879 HOXD8 NM_019558 // NM_019558 0.18 0.06 0.015 HOXD8 // homeobox D8 // 2q31.1 // 3234 /// ENST00000313173 // HOXD8 3740704 SMYD4 NM_052928 // NM_052928 0.18 0.06 0.012 SMYD4 // SET and MYND domain containing 4 // 17p13.3 // 114826 /// 3975467 UTX NM_021140 // NM_021140 0.18 0.06 0.013 UTX // ubiquitously transcribed tetratricopeptide repeat, X chromos 3699044 RFWD3 NM_018124 // NM_018124 0.18 0.06 0.011 RFWD3 // ring finger and WD repeat domain 3 // 16q22.3 // 55159 /// 3473083 MED13L NM_015335 // NM_015335 0.18 0.02 0.000 MED13L // mediator complex subunit 13-like // 12q24.21 // 23389 /// 2332711 PPIH NM_006347 // NM_006347 0.17 0.06 0.017 PPIH // peptidylprolyl isomerase H (cyclophilin H) // 1p34.1 // 104 3556990 JUB NM_032876 // JUB // NM_032876 0.17 0.04 0.004 jub, ajuba homolog (Xenopus laevis) // 14q11.2 // 84962 /// 2780143 BDH2 NM_020139 // NM_020139 0.17 0.05 0.006 BDH2 // 3- hydroxybutyrate dehydrogenase, type 2 // 4q24 // 56898 // 3899495 C20orf12 NM_001099407 // NM_001099407 0.17 0.05 0.008 C20orf12 // chromosome 20 open reading frame 12 // 20p11.23 // 5 3290875 ANK3 NM_020987 // NM_020987 0.17 0.03 0.001 ANK3 // ankyrin 3, node of Ranvier (ankyrin G) // 10q21 // 288 /// 3576014 C14orf102 NM_017970 // NM_017970 0.17 0.04 0.002 C14orf102 // chromosome 14 open reading frame 102 // 14q32.11 // 55 3644887 ATP6V0C NM_001694 // NM_001694 0.17 0.06 0.017 ATP6V0C // ATPase, H+ transporting, lysosomal 16 kDa, V0 subunit c/ 2648378 RAP2B NM_002886 // NM_002886 0.17 0.06 0.017 RAP2B // RAP2B, member of RAS oncogene family // 3q25.2 // 5912 /// 2362892 ATP1A2 NM_000702 // NM_000702 0.16 0.06 0.015 ATP1A2 // ATPase, Na+/K+ transporting, alpha 2 (+) polypeptide // 1 2361488 RHBG NM_020407 // NM_020407 0.16 0.06 0.014 RHBG // Rh family, B glycoprotein // 1q21.3 // 57127 /// ENST000003 3415915 PFDN5 NM_002624 // NM_002624 0.16 0.05 0.011 PFDN5 // prefoldin subunit 5 // 12q12 // 5204 /// NM_145897 // PFDN 3433796 PEBP1 NM_002567 // NM_002567 0.16 0.04 0.004 PEBP1 // phosphatidyl- ethanolamine binding protein 1 // 12q24.23 // 3788302 SMAD4 NM_005359 // NM_005359 0.16 0.05 0.012 SMAD4 // SMAD family member 4 // 18q21.1 // 4089 /// ENST0000039841 3436236 ZNF664 NM_152437 // NM_152437 0.16 0.06 0.016 ZNF664 // zinc finger protein 664 // 12q24.31 //144348 /// ENST000 3441542 TMEM16B NM_020373 // NM_020373 0.16 0.06 0.018 TMEM16B // transmembrane protein 16B // 12p13.3 // 57101 /// ENST00 3456353 CALCOCO1 NM_020898 // NM_020898 0.16 0.05 0.010 CALCOCO1 // calcium binding and coiled-coil domain 1 // 12q13.13 // 3888721 PTPN1 NM_002827 // NM_002827 0.16 0.06 0.020 PTPN1 // protein tyrosine phosphatase, non- receptor type 1 // 20q13 3138204 CYP7B1 NM_004820 // NM_004820 0.15 0.05 0.014 CYP7B1 // cytochrome P450, family 7, subfamily B, polypeptide 1 // 3278401 FRMD4A NM_018027 // NM_018027 0.15 0.05 0.009 FRMD4A // FERM domain containing 4A // 10p13 // 55691 /// ENST00000 3904226 RBM39 NM_184234 // NM_184234 0.15 0.05 0.015 RBM39 // RNA binding motif protein 39 // 20q11.22 // 9584 /// NM_00 3791850 SERPINB13 NM_012397 // NM_012397 0.15 0.04 0.005 SERPINB13 // serpin peptidase inhibitor, clade B (ovalbumin), membe 3665603 CTCF NM_006565 // NM_006565 0.15 0.04 0.004 CTCF // CCCTC- binding factor (zinc finger protein) // 16q21-q22.3/ 3969802 BMX NM_203281 // NM_203281 0.15 0.05 0.016 BMX // BMX non- receptor tyrosine kinase // Xp22.2 // 660 /// NM_001 3621276 HISPPD2A NM_014659 // NM_014659 0.14 0.04 0.005 HISPPD2A // histidine acid phosphatase domain containing 2A // 15q1 2325113 C1orf213 NM_138479 // NM_138479 0.14 0.05 0.012 C1orf213 // chromosome 1 open reading frame 213 // 1p36.12 // 14889 3681956 KIAA0430 NM_014647 // NM_014647 0.14 0.05 0.018 KIAA0430 // KIAA0430 // 16p13.11 // 9665 /// ENST00000396368 // KIA 3415193 GRASP NM_181711 // NM_181711 0.14 0.05 0.019 GRASP // GRP1 (general receptor for phosphoinositides 1)-associated 3249369 LRRTM3 NM_178011 // NM_178011 0.14 0.05 0.011 LRRTM3 // leucine rich repeat transmembrane neuronal 3 // 10q21.3/ 3874023 PTPRA NM_002836 // NM_002836 0.14 0.04 0.004 PTPRA // protein tyrosine phosphatase, receptor type, A // 20p13 // 3809621 FECH NM_001012515 // NM_001012515 0.14 0.04 0.009 FECH // ferrochelatase (protoporphyria) // 18q21.3 // 2235 /// N 3351385 MLL NM_005933 // NM_005933 0.14 0.05 0.016 MLL // myeloid/lymphoid or mixed-lineage leukemia (trithorax homolo 3288707 ERCC6 NM_000124 // NM_000124 0.14 0.05 0.016 ERCC6 // excision repair cross- complementing rodent repair deficien 3624607 MYO5A NM_000259 // NM_000259 0.14 0.04 0.006 MYO5A // myosin VA (heavy chain 12, myoxin) // 15q21 // 4644 /// EN 3353859 OR4D5 NM_001001965 // NM_001001965 0.14 0.05 0.017 OR4D5 // olfactory receptor, family 4, subfamily D, member 5 // 2823797 TSLP NM_033035 // NM_033035 0.14 0.05 0.013 TSLP // thymic stromal lymphopoietin // 5q22.1 // 85480 /// NM_1385 2414366 PPAP2B NM_003713 // NM_003713 0.13 0.04 0.007 PPAP2B // phosphatidic acid phosphatase type 2B // 1pter-p22.1 // 8 3878308 CSRP2BP NM_020536 // NM_020536 0.13 0.05 0.019 CSRP2BP // CSRP2 binding protein // 20p11.23 // 57325 /// NM_177926 4025771 CD99L2 NM_031462 // NM_031462 0.13 0.04 0.007 CD99L2 // CD99 molecule-like 2 // Xq28 // 83692 /// NM_134446 // CD 3414776 LETMD1 NM_015416 // NM_015416 0.13 0.05 0.014 LETMD1 // LETM1 domain containing 1 // 12q13.13 // 25875 /// NM_001 3645253 SRRM2 NM_016333 // NM_016333 0.13 0.04 0.007 SRRM2 // serine/arginine repetitive matrix 2 // 16p13.3 // 23524 // 2440700 ADAMTS4 NM_005099 // NM_005099 0.13 0.03 0.005 ADAMTS4 // ADAM metallopeptidase with thrombospondin type 1 motif, 2609870 BRPF1 NM_001003694 // NM_001003694 0.13 0.04 0.012 BRPF1 // bromodomain and PHD finger containing, 1 // 3p26-p25 // 3632298 ADPGK NM_031284 // NM_031284 0.13 0.04 0.007 ADPGK // ADP- dependent glucokinase // 15q24.1 // 83440 /// ENST0000 3184940 GNG10 NM_001017998 // NM_001017998 0.13 0.04 0.011 GNG10 // guanine nucleotide binding protein (G protein), gamma 1 3223776 C5 NM_001735 // C5 // NM_001735 0.13 0.04 0.008 complement component 5 // 9q33-q34 // 727 /// ENST00000223642 3922100 MX1 NM_002462 // NM_002462 0.12 0.04 0.015 MX1 // myxovirus (influenza virus) resistance 1, interferon-inducib 3960478 CSNK1E NM_001894 // NM_001894 0.12 0.04 0.018 CSNK1E // casein kinase 1, epsilon // 22q13.1 // 1454 /// NM_152221 3715703 SUPT6H NM_003170 // NM_003170 0.11 0.03 0.005 SUPT6H // suppressor of Ty 6 homolog (S. cerevisiae) // 17q11.2 // 2322818 PADI3 NM_016233 // NM_016233 0.11 0.03 0.006 PADI3 // peptidyl arginine deiminase, type III // 1p36.13 // 51702 2393740 KIAA0562 NM_014704 // NM_014704 0.11 0.03 0.009 KIAA0562 // KIAA0562 // 1p36.32 // 9731 /// ENST00000378230 // KIAA 3784509 ZNF271 NM_001112663 // NM_001112663 0.11 0.04 0.020 ZNF271 // zinc finger protein 271 // 18q12 // 10778 /// NM_00662 3372253 CUGBP1 NM_006560 // NM_006560 0.11 0.04 0.011 CUGBP1 // CUG triplet repeat, RNA binding protein 1 // 11p11 // 106 2948259 TRIM26 NM_003449 // NM_003449 0.11 0.03 0.006 TRIM26 // tripartite motif-containing 26 // 6p21.3 // 7726 /// ENST 3191900 NUP214 NM_005085 // NM_005085 0.11 0.03 0.003 NUP214 // nucleoporin 214 kDa // 9q34.1 // 8021 /// ENST00000359428 3105581 CA3 NM_005181 // CA3 // NM_005181 0.11 0.03 0.003 carbonic anhydrase III, muscle specific // 8q13-q22 // 761/ 3832457 RYR1 NM_000540 // NM_000540 0.11 0.03 0.006 RYR1 // ryanodine receptor 1 (skeletal) // 19q13.1 // 6261 /// NM_0 3936256 BCL2L13 NM_015367 // NM_015367 0.10 0.02 0.002 BCL2L13 // BCL2- like 13 (apoptosis facilitator) // 22q11 // 23786/ 3599280 PIAS1 NM_016166 // NM_016166 0.10 0.04 0.017 PIAS1 // protein inhibitor of activated STAT, 1 // 15q // 8554 /// 3755976 MED24 NM_014815 // NM_014815 0.10 0.04 0.019 MED24 // mediator complex subunit 24 // 17q21.1 // 9862 /// NM_0010 3656418 SRCAP NM_006662 // NM_006662 0.10 0.04 0.017 SRCAP // Snf2- related CREBBP activator protein // 16p11.2 // 10847 3943101 DEPDC5 NM_014662 // NM_014662 0.09 0.01 0.000 DEPDC5 // DEP domain containing 5 // 22q12.3 // 9681 /// NM_0010071 3960685 DMC1 NM_007068 // NM_007068 0.09 0.03 0.013 DMC1 // DMC1 dosage suppressor of mck1 homolog, meiosis-specific ho 2434776 CDC42SE1 NM_001038707 // NM_001038707 0.08 0.03 0.014 CDC42SE1 // CDC42 small effector 1 // 1q21.2 // 56882 /// NM_020 3438417 SFRS8 NM_004592 // NM_004592 0.08 0.03 0.016 SFRS8 // splicing factor, arginine/serine-rich 8 (suppressor-of- whi 3457696 PAN2 NM_014871 // NM_014871 0.08 0.02 0.008 PAN2 // PAN2 polyA specific ribonuclease subunit homolog (S. cerevi 2534615 SCLY NM_016510 // NM_016510 0.08 0.02 0.004 SCLY // selenocysteine lyase // 2q37.3 // 51540 /// ENST00000254663 2765865 RELL1 NM_001085400 // NM_001085400 0.07 0.02 0.002 RELL1 // RELT- like 1 // 4p14 // 768211 /// NM_001085399 // RELL1 3765642 INTS2 NM_020748 // NM_020748 0.05 0.01 0.005 INTS2 // integrator complex subunit 2 // 17q23.2 // 57508 /// ENST0 2906607 NFYA NM_002505 // NM_002505 −0.07 0.02 0.011 NFYA // nuclear transcription factor Y, alpha // 6p21.3 // 4800 /// 3168102 CREB3 NM_006368 // NM_006368 −0.07 0.02 0.010 CREB3 // cAMP responsive element binding protein 3 // 9pter-p22.1/ 3939365 SMARCB1 NM_003073 // NM_003073 −0.07 0.02 0.013 SMARCB1 // SWI/SNF related, matrix associated, actin dependent regu 3415229 NR4A1 NM_002135 // NM_002135 −0.07 0.03 0.015 NR4A1 // nuclear receptor subfamily 4, group A, member 1 // 12q13/ 2437801 ARHGEF2 NM_004723 // NM_004723 −0.09 0.02 0.002 ARHGEF2 // rho/rac guanine nucleotide exchange factor (GEF) 2 // 1q 3645565 THOC6 NM_024339 // NM_024339 −0.10 0.04 0.018 THOC6 // THO complex 6 homolog (Drosophila) // 16p13.3 // 79228 /// 2406766 MRPS15 NM_031280 // NM_031280 −0.11 0.03 0.003 MRPS15 // mitochondrial ribosomal protein S15 // 1p35-p34.1 // 6496 3553141 KIAA0329 NM_014844 // NM_014844 −0.11 0.04 0.018 KIAA0329 // KIAA0329 // 14q32.31 // 9895 /// ENST00000359520 // KIA 3297666 DYDC1 NM_138812 // NM_138812 −0.11 0.02 0.000 DYDC1 // DPY30 domain containing 1 // 10q23.1 // 143241 /// ENST000 3625674 RFXDC2 NM_022841 // NM_022841 −0.12 0.04 0.012 RFXDC2 // regulatory factor X domain containing 2 // 15q21.3 // 648 2926969 PDE7B NM_018945 // NM_018945 −0.12 0.04 0.013 PDE7B // phosphodiesterase 7B // 6q23-q24 // 27115 /// ENST00000308 3525313 COL4A1 NM_001845 // NM_001845 −0.12 0.04 0.014 COL4A1 // collagen, type IV, alpha 1 // 13q34 // 1282 /// ENST00000 2438892 FCRL5 NM_031281 // NM_031281 −0.12 0.04 0.009 FCRL5 // Fc receptor-like 5 // 1q21 // 83416 /// ENST00000361835 // 3220846 SUSD1 NM_022486 // NM_022486 −0.12 0.03 0.006 SUSD1 // sushi domain containing 1 // 9q31.3-q33.1 // 64420 /// ENS 3598430 SLC24A1 NM_004727 // NM_004727 −0.12 0.05 0.019 SLC24A1 // solute carrier family 24 (sodium/potassium/ calcium excha 3506431 RNF6 NM_005977 // NM_005977 −0.12 0.04 0.011 RNF6 // ring finger protein (C3H2C3 type) 6 // 13q12.2 // 6049 /// 3696057 SLC12A4 NM_005072 // NM_005072 −0.12 0.02 0.001 SLC12A4 // solute carrier family 12 (potassium/chloride transporter 2519577 COL3A1 NM_000090 // NM_000090 −0.12 0.04 0.012 COL3A1 // collagen, type III, alpha 1 (Ehlers- Danlos syndrome type 3734479 TMEM104 NM_017728 // NM_017728 −0.13 0.04 0.015 TMEM104 // transmembrane protein 104 // 17q25.1 // 54868 /// ENST00 3345157 PIWIL4 NM_152431 // NM_152431 −0.13 0.05 0.015 PIWIL4 // piwi-like 4 (Drosophila) // 11q21 // 143689 /// ENST00000 2949471 NEU1 NM_000434 // NM_000434 −0.13 0.04 0.013 NEU1 // sialidase 1 (lysosomal sialidase) // 6p21.3 // 4758 /// ENS 2599670 CRYBA2 NM_057093 // NM_057093 −0.13 0.04 0.014 CRYBA2 // crystallin, beta A2 // 2q34-q36 // 1412 /// NM_005209 // 3922444 ABCG1 NM_207628 // NM_207628 −0.13 0.03 0.003 ABCG1 // ATP- binding cassette, sub-family G (WHITE), member 1 // 21 2760371 WDR1 NM_017491 // NM_017491 −0.14 0.05 0.019 WDR1 // WD repeat domain 1 // 4p16.1 // 9948 /// NM_005112 // WDR1 2835440 TCOF1 NM_001008656 // NM_001008656 −0.14 0.04 0.007 TCOF1 // Treacher Collins- Franceschetti syndrome 1 // 5q32- q33.1 2451544 MYOG NM_002479 // NM_002479 −0.14 0.05 0.018 MYOG // myogenin (myogenic factor 4) // 1q31-q41 // 4656 /// ENST00 3745504 SCO1 NM_004589 // NM_004589 −0.14 0.03 0.003 SCO1 // SCO cytochrome oxidase deficient homolog 1 (yeast) // 17p12 2835213 PPARGC1B NM_133263 // NM_133263 −0.14 0.04 0.006 PPARGC1B // peroxisome proliferator- activated receptor gamma, coact 3704567 CBFA2T3 NM_005187 // NM_005187 −0.14 0.05 0.020 CBFA2T3 // core- binding factor, runt domain, alpha subunit 2; trans 2893562 RREB1 NM_002955 // NM_002955 −0.14 0.04 0.006 RREB1 // ras responsive element binding protein 1 // 6p25 // 6239/ 2672712 SCAP NM_012235 // NM_012235 −0.14 0.04 0.009 SCAP // SREBF chaperone // 3p21.31 // 22937 /// ENST00000265565 // 2768197 CORIN NM_006587 // NM_006587 −0.14 0.05 0.011 CORIN // corin, serine peptidase // 4p13-p12 // 10699 /// ENST00000 2495279 VWA3B NM_144992 // NM_144992 −0.14 0.04 0.006 VWA3B // von Willebrand factor A domain containing 3B // 2q11.2 // 2903588 PFDN6 NM_014260 // NM_014260 −0.14 0.05 0.014 PFDN6 // prefoldin subunit 6 // 6p21.3 // 10471 /// ENST00000399112 3031383 REPIN1 NM_013400 // NM_013400 −0.15 0.05 0.018 REPIN1 // replication initiator 1 // 7q36.1 // 29803 /// NM_014374 3754469 ACACA NM_198839 // NM_198839 −0.15 0.05 0.010 ACACA // acetyl- Coenzyme A carboxylase alpha // 17q21 // 31 /// NM 3767480 AXIN2 NM_004655 // NM_004655 −0.15 0.05 0.013 AXIN2 // axin 2 (conductin, axil) // 17q23-q24 // 8313 /// ENST0000 2954506 CRIP3 NM_206922 // NM_206922 −0.15 0.06 0.018 CRIP3 // cysteine- rich protein 3 // 6p21.1 // 401262 /// ENST000003 3845263 ADAMTSL5 NM_213604 // NM_213604 −0.15 0.06 0.016 ADAMTSL5 // ADAMTS-like 5 // 19p13.3 // 339366 /// ENST00000330475 2565143 STARD7 NM_020151 // NM_020151 −0.15 0.06 0.016 STARD7 // StAR- related lipid transfer (START) domain containing 7/ 2321960 PLEKHM2 NM_015164 // NM_015164 −0.16 0.05 0.009 PLEKHM2 // pleckstrin homology domain containing, family M (with RU 3829174 GPATCH1 NM_018025 // NM_018025 −0.16 0.03 0.001 GPATCH1 // G patch domain containing 1 // 19q13.11 // 55094 /// ENS 2798586 AHRR NM_020731 // NM_020731 −0.16 0.05 0.011 AHRR // aryl- hydrocarbon receptor repressor // 5p15.3 // 57491 /// 2362991 CASQ1 NM_001231 // NM_001231 −0.16 0.06 0.015 CASQ1 // calsequestrin 1 (fast-twitch, skeletal muscle) // 1q21 // 3954525 ZNF280B NM_080764 // NM_080764 −0.16 0.04 0.005 ZNF280B // zinc finger protein 280B // 22q11.22 // 140883 /// ENST0 4020991 ACTRT1 NM_138289 // NM_138289 −0.16 0.05 0.007 ACTRT1 // actin- related protein T1 // Xq25 // 139741 /// ENST000003 3982975 POU3F4 NM_000307 // NM_000307 −0.16 0.05 0.013 POU3F4 // POU class 3 homeobox 4 // Xq21.1 // 5456 /// ENST00000373 3963990 PKDREJ NM_006071 // NM_006071 −0.16 0.03 0.001 PKDREJ // polycystic kidney disease (polycystin) and REJ homolog (s 2436401 JTB NM_006694 // JTB // NM_006694 −0.16 0.06 0.014 jumping translocation breakpoint // 1q21 // 10899 /// NM_002 2759654 ABLIM2 NM_032432 // NM_032432 −0.16 0.05 0.007 ABLIM2 // actin binding LIM protein family, member 2 // 4p16- p15 // 2437329 CLK2 NM_003993 // NM_003993 −0.16 0.06 0.016 CLK2 // CDC-like kinase 2 // 1q21 // 1196 /// NR_002711 // CLK2P // 3401119 ITFG2 NM_018463 // NM_018463 −0.16 0.04 0.004 ITFG2 // integrin alpha FG-GAP repeat containing 2 // 12p13.33 // 5 3599709 GLCE NM_015554 // NM_015554 −0.16 0.06 0.014 GLCE // glucuronic acid epimerase // 15q23 // 26035 /// ENST0000026 3882413 C20orf114 NM_033197 // NM_033197 −0.16 0.06 0.020 C20orf114 // chromosome 20 open reading frame 114 // 20q11.21 // 92 3712922 C17orf39 NM_024052 // NM_024052 −0.16 0.06 0.017 C17orf39 // chromosome 17 open reading frame 39 // 17p11.2 // 79018 2473376 EFR3B BC049384 // BC049384 −0.17 0.05 0.009 EFR3B // EFR3 homolog B (S. cerevisiae) // 2p23.3 // 22979 /// ENST0 2607262 STK25 NM_006374 // NM_006374 −0.17 0.06 0.015 STK25 // serine/threonine kinase 25 (STE20 homolog, yeast) // 2q37. 3755580 CACNB1 NM_199247 // NM_199247 −0.17 0.06 0.013 CACNB1 // calcium channel, voltage-dependent, beta 1 subunit // 17q 3402150 NTF3 NM_001102654 // NM_001102654 −0.17 0.06 0.020 NTF3 // neurotrophin 3 // 12p13 // 4908 /// NM_002527 // NTF3 // 3014714 ARPC1B NM_005720 // NM_005720 −0.17 0.06 0.020 ARPC1B // actin related protein 2/3 complex, subunit 1B, 41 kDa // 7 3723071 DBF4B NM_145663 // NM_145663 −0.17 0.04 0.002 DBF4B // DBF4 homolog B (S. cerevisiae) // 17q21.31|17q21 // 80174 2371255 SMG7 NM_173156 // NM_173156 −0.17 0.06 0.014 SMG7 // Smg-7 homolog, nonsense mediated mRNA decay factor (C. eleg 3217487 ALG2 NM_033087 // NM_033087 −0.17 0.06 0.011 ALG2 // asparagine-linked glycosylation 2 homolog (S. cerevisiae, a 3352159 LOC100130353 AK130019 // AK130019 −0.17 0.06 0.018 LOC100130353 // hypothetical protein LOC100130353 // 11q23.3 // 1001 3401259 TEAD4 NM_003213 // NM_003213 −0.17 0.07 0.020 TEAD4 // TEA domain family member 4 // 12p13.3-p13.2 // 7004 /// NM 3114618 RNF139 NM_007218 // NM_007218 −0.17 0.06 0.015 RNF139 // ring finger protein 139 // 8q24 // 11236 /// ENST00000303 2991150 TSPAN13 NM_014399 // NM_014399 −0.18 0.05 0.006 TSPAN13 // tetraspanin 13 // 7p21.1 // 27075 /// ENST00000262067 // 2875193 P4HA2 NM_004199 // NM_004199 −0.18 0.05 0.007 P4HA2 // procollagen- proline, 2- oxoglutarate 4- dioxygenase (proline 4011743 SLC7A3 NM_032803 // NM_032803 −0.18 0.06 0.009 SLC7A3 // solute carrier family 7 (cationic amino acid transporter, 3194015 LCN9 NM_001001676 // NM_001001676 −0.18 0.06 0.011 LCN9 // lipocalin 9 // 9q34.3 // 392399 /// ENST00000277526 // L 3741040 MNT NM_020310 // NM_020310 −0.18 0.04 0.003 MNT // MAX binding protein // 17p13.3 // 4335 /// ENST00000174618/ 3901851 ABHD12 NM_001042472 // NM_001042472 −0.18 0.05 0.004 ABHD12 // abhydrolase domain containing 12 // 20p11.21 // 26090 2324919 EPHB2 NM_017449 // NM_017449 −0.18 0.06 0.010 EPHB2 // EPH receptor B2 // 1p36.1-p35 // 2048 /// NM_004442 // EPH 3185976 COL27A1 NM_032888 // NM_032888 −0.18 0.06 0.009 COL27A1 // collagen, type XXVII, alpha 1 // 9q32 // 85301 /// ENST0 2855434 C5orf39 NM_001014279 // NM_001014279 −0.18 0.05 0.007 C5orf39 // chromosome 5 open reading frame 39 // 5p12 // 389289 2334476 MAST2 NM_015112 // NM_015112 −0.18 0.02 0.000 MAST2 // microtubule associated serine/threonine kinase 2 // 1p34.1 3962734 TTLL1 NM_001008572 // NM_001008572 −0.18 0.03 0.001 TTLL1 // tubulin tyrosine ligase-like family, member 1 // 22q13. 4017538 COL4A6 NM_033641 // NM_033641 −0.18 0.03 0.000 COL4A6 // collagen, type IV, alpha 6 // Xq22 // 1288 /// NM_001847 3141589 IL7 NM_000880 // IL7 // NM_000880 −0.19 0.05 0.006 interleukin 7 // 8q12-q13 // 3574 /// ENST00000263851 // IL7 2436826 KCNN3 NM_002249 // NM_002249 −0.19 0.06 0.008 KCNN3 // potassium intermediate/small conductance calcium-activated 3521174 ABCC4 NM_005845 // NM_005845 −0.19 0.07 0.017 ABCC4 // ATP- binding cassette, sub-family C (CFTR/MRP), member 4 // 3768280 C17orf58 NM_181656 // NM_181656 −0.19 0.07 0.017 C17orf58 // chromosome 17 open reading frame 58 // 17q24.2 // 28401 2363784 HSPA6 NM_002155 // NM_002155 −0.19 0.06 0.011 HSPA6 // heat shock 70 kDa protein 6 (HSP70B′) // 1q23 // 3310 /// E 3928211 GRIK1 NM_175611 // NM_175611 −0.19 0.06 0.011 GRIK1 // glutamate receptor, ionotropic, kainate 1 // 21q22.11 // 2 2758978 EVC2 NM_147127 // NM_147127 −0.19 0.06 0.012 EVC2 // Ellis van Creveld syndrome 2 (limbin) // 4p16.2-p16.1 // 13 3740664 C17orf91 NM_032895 // NM_032895 −0.19 0.07 0.015 C17orf91 // chromosome 17 open reading frame 91 // 17p13.3 // 84981 2782267 NEUROG2 NM_024019 // NM_024019 −0.20 0.06 0.010 NEUROG2 // neurogenin 2 // 4q25 // 63973 /// ENST00000313341 // NEU 3826542 ZNF738 BC034499 // BC034499 −0.20 0.05 0.003 ZNF738 // zinc finger protein 738 // 19p12 // 148203 /// AK291002 // 3966000 TYMP NM_001113756 // NM_001113756 −0.20 0.05 0.003 TYMP // thymidine phosphorylase // 22q13|22q13.33 // 1890 /// NM 3607447 ABHD2 NM_007011 // NM_007011 −0.20 0.05 0.005 ABHD2 // abhydrolase domain containing 2 // 15q26.1 // 11057 /// NM 3236448 SUV39H2 NM_024670 // NM_024670 −0.20 0.07 0.011 SUV39H2 // suppressor of variegation 3-9 homolog 2 (Drosophila) // 2528504 SPEG NM_005876 // NM_005876 −0.20 0.06 0.009 SPEG // SPEG complex locus // 2q35 // 10290 /// ENST00000312358 // 2730746 SLC4A4 NM_001098484 // NM_001098484 −0.20 0.06 0.007 SLC4A4 // solute carrier family 4, sodium bicarbonate cotranspor 2544662 DNMT3A NM_175629 // NM_175629 −0.20 0.06 0.007 DNMT3A // DNA (cytosine-5-)- methyltransferase 3 alpha // 2p23 // 17 2937625 C6orf208 BC101251 // BC101251 −0.20 0.06 0.007 C6orf208 // chromosome 6 open reading frame 208 // 6q27 // 80069 /// 3233157 UCN3 NM_053049 // NM_053049 −0.20 0.08 0.017 UCN3 // urocortin 3 (stresscopin) // 10p15.1 // 114131 /// ENST0000 2548172 FEZ2 NM_001042548 // NM_001042548 −0.21 0.03 0.000 FEZ2 // fasciculation and elongation protein zeta 2 (zygin II)/ 3877809 OTOR NM_020157 // NM_020157 −0.21 0.08 0.019 OTOR // otoraplin // 20p12.1-p11.23 // 56914 /// ENST00000246081 // 3839400 C19orf63 NM_175063 // NM_175063 −0.21 0.04 0.002 C19orf63 // chromosome 19 open reading frame 63 // 19q13.33 // 2843 3875108 C20orf196 AK292708 // AK292708 −0.21 0.06 0.006 C20orf196 // chromosome 20 open reading frame 196 // 20p12.3 // 1498 2970985 TSPYL4 NM_021648 // NM_021648 −0.21 0.07 0.011 TSPYL4 // TSPY- like 4 // 6q22.1 // 23270 /// ENST00000368611 // TSP 3189580 ZBTB43 NM_014007 // NM_014007 −0.21 0.08 0.017 ZBTB43 // zinc finger and BTB domain containing 43 // 9q33-q34 // 2 3407926 CMAS NM_018686 // NM_018686 −0.21 0.03 0.000 CMAS // cytidine monophosphate N- acetylneuraminic acid synthetase/ 3249886 TET1 NM_030625 // NM_030625 −0.21 0.06 0.007 TET1 // tet oncogene 1 // 10q21 // 80312 /// ENST00000373644 // TET 3151970 MTSS1 NM_014751 // NM_014751 −0.21 0.07 0.009 MTSS1 // metastasis suppressor 1 // 8p22 // 9788 /// ENST0000032506 3937183 DGCR8 NM_022720 // NM_022720 −0.21 0.06 0.008 DGCR8 // DiGeorge syndrome critical region gene 8 // 22q11.2 // 544 3958253 C22orf28 BC016707 // BC016707 −0.22 0.08 0.019 C22orf28 // chromosome 22 open reading frame 28 // 22q12 // 51493 // 3607503 ABHD2 NM_007011 // NM_007011 −0.22 0.07 0.010 ABHD2 // abhydrolase domain containing 2 // 15q26.1 // 11057 /// NM 2799030 SLC6A19 NM_001003841 // NM_001003841 −0.22 0.06 0.007 SLC6A19 // solute carrier family 6 (neutral amino acid transport 3870611 LILRB3 NM_001081450 // NM_001081450 −0.22 0.08 0.016 LILRB3 // leukocyte immunoglobulin- like receptor, subfamily B (w 3857811 C19orf12 NM_031448 // NM_031448 −0.22 0.08 0.019 C19orf12 // chromosome 19 open reading frame 12 // 19q12 // 83636/ 2500667 FBLN7 NM_153214 // NM_153214 −0.22 0.08 0.019 FBLN7 // fibulin 7 // 2q13 // 129804 /// ENST00000331203 // FBLN7/ 3523156 TMTC4 NM_032813 // NM_032813 −0.22 0.07 0.010 TMTC4 // transmembrane and tetratricopeptide repeat containing 4 // 2612371 EAF1 NM_033083 // NM_033083 −0.22 0.07 0.008 EAF1 // ELL associated factor 1 // 3p24.3 // 85403 /// ENST00000396 3988638 LONRF3 NM_001031855 // NM_001031855 −0.23 0.08 0.012 LONRF3 // LON peptidase N- terminal domain and ring finger 3 // X 3114240 C8orf32 BC008781 // BC008781 −0.23 0.08 0.016 C8orf32 // chromosome 8 open reading frame 32 // 8q24.13 // 55093 // 2460368 TTC13 NM_024525 // NM_024525 −0.23 0.08 0.014 TTC13 // tetratricopeptide repeat domain 13 // 1q42.2 // 79573 /// 2428425 PPM1J NM_005167 // NM_005167 −0.23 0.06 0.003 PPM1J // protein phosphatase 1J (PP2C domain containing) // 1p13.2 3194986 LCN12 NM_178536 // NM_178536 −0.23 0.06 0.004 LCN12 // lipocalin 12 // 9q34.3 // 286256 /// ENST00000371633 // LC 3642875 RAB11FIP3 NM_014700 // NM_014700 −0.23 0.07 0.010 RAB11FIP3 // RAB11 family interacting protein 3 (class II) // 16p13 2532378 CHRND NM_000751 // NM_000751 −0.23 0.08 0.018 CHRND // cholinergic receptor, nicotinic, delta // 2q33-q34 // 1144 2995667 ADCYAP1R1 NM_001118 // NM_001118 −0.23 0.05 0.002 ADCYAP1R1 // adenylate cyclase activating polypeptide 1 (pituitary) 3390641 ARHGAP20 NM_020809 // NM_020809 −0.23 0.05 0.003 ARHGAP20 // Rho GTPase activating protein 20 // 11q22.3-q23.1 // 57 2830465 MYOT NM_006790 // NM_006790 −0.23 0.07 0.007 MYOT // myotilin // 5q31 // 9499 /// ENST00000239926 // MYOT // myo 2452069 PIK3C2B NM_002646 // NM_002646 −0.23 0.02 0.000 PIK3C2B // phosphoinositide-3- kinase, class 2, beta polypeptide // 3744127 HES7 NM_032580 // NM_032580 −0.23 0.09 0.019 HES7 // hairy and enhancer of split 7 (Drosophila) // 17p13.1 // 84 3327057 FLJ14213 NM_024841 // NM_024841 −0.23 0.07 0.007 FLJ14213 // protor- 2 // 11p13-p12 // 79899 /// ENST00000378867 // F 2664332 COLQ NM_005677 // NM_005677 −0.23 0.07 0.006 COLQ // collagen- like tail subunit (single strand of homotrimer) of 3829160 C19orf40 NM_152266 // NM_152266 −0.23 0.08 0.012 C19orf40 // chromosome 19 open reading frame 40 // 19q13.11 // 9144 3708798 SENP3 NM_015670 // NM_015670 −0.23 0.06 0.005 SENP3 // SUMO1/sentrin/SM T3 specific peptidase 3 // 17p13 // 26168 2358700 MGC29891 NM_144618 // NM_144618 −0.23 0.09 0.019 MGC29891 // hypothetical protein MGC29891 // 1q21.2 // 126626 /// E 2755111 KLKB1 NM_000892 // NM_000892 −0.24 0.08 0.012 KLKB1 // kallikrein B, plasma (Fletcher factor) 1 // 4q34-q35 // 38 2568968 UXS1 NM_025076 // NM_025076 −0.24 0.08 0.011 UXS1 // UDP- glucuronate decarboxylase 1 // 2q12.2 // 80146 /// BC00 2748923 GUCY1B3 NM_000857 // NM_000857 −0.24 0.07 0.007 GUCY1B3 // guanylate cyclase 1, soluble, beta 3 // 4q31.3-q33 // 29 3816509 GADD45B NM_015675 // NM_015675 −0.24 0.09 0.016 GADD45B // growth arrest and DNA-damage- inducible, beta // 19p13.3 3376410 SLC22A24 BC034394 // BC034394 −0.24 0.07 0.007 SLC22A24 // solute carrier family 22, member 24 // 11q12.3 // 283238 3286393 ZNF32 NM_006973 // NM_006973 −0.24 0.08 0.010 ZNF32 // zinc finger protein 32 // 10q22-q25 // 7580 /// NM_0010053 2540157 ODC1 NM_002539 // NM_002539 −0.24 0.09 0.020 ODC1 // ornithine decarboxylase 1 // 2p25 // 4953 /// ENST000002341 2994835 CHN2 NM_004067 // NM_004067 −0.24 0.09 0.017 CHN2 // chimerin (chimaerin) 2 // 7p15.3 // 1124 /// NM_001039936/ 3603199 IDH3A NM_005530 // NM_005530 −0.24 0.05 0.001 IDH3A // isocitrate dehydrogenase 3 (NAD+) alpha // 15q25.1-q25.2/ 3040454 TWISTNB NM_001002926 // NM_001002926 −0.24 0.09 0.017 TWISTNB // TWIST neighbor // 7p15.3 // 221830 /// ENST0000022256 2497301 TMEM182 NM_144632 // NM_144632 −0.24 0.07 0.007 TMEM182 // transmembrane protein 182 // 2q12.1 // 130827 /// ENST00 3766716 TEX2 NM_018469 // NM_018469 −0.25 0.07 0.007 TEX2 // testis expressed 2 // 17q23.3 // 55852 /// ENST00000258991 3458819 CYP27B1 NM_000785 // NM_000785 −0.25 0.08 0.009 CYP27B1 // cytochrome P450, family 27, subfamily B, polypeptide 1/ 3368940 ABTB2 NM_145804 // NM_145804 −0.25 0.08 0.010 ABTB2 // ankyrin repeat and BTB (POZ) domain containing 2 // 11p13 3298924 MMRN2 NM_024756 // NM_024756 −0.25 0.07 0.006 MMRN2 // multimerin 2 // 10q23.2 // 79812 /// ENST00000372027 // MM 3529951 KIAA1305 NM_025081 // NM_025081 −0.25 0.08 0.011 KIAA1305 // KIAA1305 // 14q12 // 57523 /// BC008219 // KIAA1305 // 3006572 AUTS2 NM_015570 // NM_015570 −0.25 0.09 0.017 AUTS2 // autism susceptibility candidate 2 // 7q11.22 // 26053 /// 3025500 BPGM NM_001724 // NM_001724 −0.25 0.10 0.018 BPGM // 2,3- bisphosphoglycerate mutase // 7q31- q34 // 669 /// NM_19 2494709 CNNM4 NM_020184 // NM_020184 −0.26 0.09 0.016 CNNM4 // cyclin M4 // 2p12-p11.2 // 26504 /// ENST00000377075 // CN 3329983 PTPRJ NM_002843 // NM_002843 −0.26 0.08 0.010 PTPRJ // protein tyrosine phosphatase, receptor type, J // 11p11.2 2769346 LNX1 NM_032622 // NM_032622 −0.26 0.09 0.015 LNX1 // ligand of numb-protein X 1 // 4q12 // 84708 /// ENST0000030 3867195 FAM83E NM_017708 // NM_017708 −0.26 0.09 0.013 FAM83E // family with sequence similarity 83, member E // 19q13.32- 3790529 GRP NM_002091 // NM_002091 −0.26 0.05 0.001 GRP // gastrin- releasing peptide // 18q21.1-q21.32 // 2922 /// NM_0 3987029 TMEM164 NM_032227 // NM_032227 −0.26 0.10 0.018 TMEM164 // transmembrane protein 164 // Xq22.3 // 84187 /// ENST000 3526454 GRTP1 NM_024719 // NM_024719 −0.26 0.09 0.015 GRTP1 // growth hormone regulated TBC protein 1 // 13q34 // 79774/ 2438344 GPATCH4 NM_182679 // NM_182679 −0.26 0.07 0.006 GPATCH4 // G patch domain containing 4 // 1q22 // 54865 /// NM_0155 3132927 NKX6-3 NM_152568 // NM_152568 −0.27 0.09 0.014 NKX6-3 // NK6 homeobox 3 // 8p11.21 // 157848 /// ENST00000343444/ 2672376 TESSP2 NM_182702 // NM_182702 −0.27 0.09 0.013 TESSP2 // testis serine protease 2 // 3p21.31 // 339906 /// ENST000 2730347 C4orf35 NM_033122 // NM_033122 −0.27 0.10 0.019 C4orf35 // chromosome 4 open reading frame 35 // 4q13.3 // 85438 // 3921068 ETS2 NM_005239 // NM_005239 −0.27 0.03 0.000 ETS2 // v-ets erythroblastosis virus E26 oncogene homolog 2 (avian) 2532894 DGKD NM_152879 // NM_152879 −0.27 0.07 0.003 DGKD // diacylglycerol kinase, delta 130 kDa // 2q37.1 // 8527 /// N 4018454 AMOT NM_133265 // NM_133265 −0.27 0.09 0.012 AMOT // angiomotin // Xq23 // 154796 /// NM_001113490 // AMOT // an 3070507 RNF148 NM_198085 // NM_198085 −0.27 0.10 0.017 RNF148 // ring finger protein 148 // 7q31.33 // 378925 /// BC029264 3832256 SPINT2 NM_021102 // NM_021102 −0.27 0.10 0.017 SPINT2 // serine peptidase inhibitor, Kunitz type, 2 // 19q13.1 // 3371225 CHST1 NM_003654 // NM_003654 −0.27 0.07 0.005 CHST1 // carbohydrate (keratan sulfate Gal-6) sulfotransferase 1 // 3870494 TFPT NM_013342 // NM_013342 −0.27 0.09 0.010 TFPT // TCF3 (E2A) fusion partner (in childhood Leukemia) // 19q13 3863811 PSG9 NM_002784 // NM_002784 −0.28 0.09 0.011 PSG9 // pregnancy specific beta-1- glycoprotein 9 // 19q13.2 // 5678 3160175 VLDLR NM_003383 // NM_003383 −0.28 0.08 0.007 VLDLR // very low density lipoprotein receptor // 9p24 // 7436 /// 2794704 ASB5 NM_080874 // NM_080874 −0.28 0.11 0.019 ASB5 // ankyrin repeat and SOCS box-containing 5 // 4q34.2 // 14045 3908901 KCNB1 NM_004975 // NM_004975 −0.28 0.09 0.009 KCNB1 // potassium voltage- gated channel, Shab-related subfamily, m 3390852 FLJ45803 NM_207429 // NM_207429 −0.28 0.10 0.015 FLJ45803 // FLJ45803 protein // 11q23.1 // 399948 /// ENST000003554 2600689 EPHA4 NM_004438 // NM_004438 −0.29 0.07 0.003 EPHA4 // EPH receptor A4 // 2q36.1 // 2043 /// ENST00000281821 // E 3469597 NUAK1 NM_014840 // NM_014840 −0.29 0.09 0.009 NUAK1 // NUAK family, SNF1-like kinase, 1 // 12q23.3 // 9891 /// EN 3607232 ISG20L1 NM_022767 // NM_022767 −0.29 0.10 0.015 ISG20L1 // interferon stimulated exonuclease gene 20 kDa-like 1 // 1 2358426 ADAMTSL4 AK023606 // AK023606 −0.29 0.11 0.016 ADAMTSL4 // ADAMTS-like 4 // 1q21.2 // 54507 3853609 CYP4F2 NM_001082 // NM_001082 −0.29 0.11 0.016 CYP4F2 // cytochrome P450, family 4, subfamily F, polypeptide 2 // 2936971 KIF25 NM_030615 // NM_030615 −0.30 0.09 0.008 KIF25 // kinesin family member 25 // 6q27 // 3834 /// NM_005355 // 2997272 EEPD1 NM_030636 // NM_030636 −0.30 0.09 0.010 EEPD1 // endonuclease/ exonuclease/ phosphatase family domain contain 3961253 RPS19BP1 NM_194326 // NM_194326 −0.30 0.10 0.013 RPS19BP1 // ribosomal protein S19 binding protein 1 // 22q13.1 // 9 3082373 VIPR2 NM_003382 // NM_003382 −0.30 0.10 0.011 VIPR2 // vasoactive intestinal peptide receptor 2 // 7q36.3 // 7434 2340961 IL12RB2 NM_001559 // NM_001559 −0.30 0.08 0.005 IL12RB2 // interleukin 12 receptor, beta 2 // 1p31.3-p31.2 // 3595 2736462 BMPR1B NM_001203 // NM_001203 −0.30 0.08 0.004 BMPR1B // bone morphogenetic protein receptor, type IB // 4q22-q24 3774504 −0.30 0.11 0.016 3395958 OR8B4 NM_001005196 // NM_001005196 −0.30 0.11 0.018 OR8B4 // olfactory receptor, family 8, subfamily B, member 4 // 2806231 BXDC2 NM_018321 // NM_018321 −0.31 0.10 0.013 BXDC2 // brix domain containing 2 // 5p13.2 // 55299 /// ENST000003 2396858 NPPB NM_002521 // NM_002521 −0.31 0.11 0.016 NPPB // natriuretic peptide precursor B // 1p36.2 // 4879 /// ENST0 3233322 C10orf18 NM_017782 // NM_017782 −0.31 0.06 0.001 C10orf18 // chromosome 10 open reading frame 18 // 10p15.1 // 54906 2439101 FCRL1 NM_052938 // NM_052938 −0.31 0.06 0.001 FCRL1 // Fc receptor-like 1 // 1q21-q22 // 115350 /// ENST000003681 2413907 DHCR24 NM_014762 // NM_014762 −0.31 0.11 0.014 DHCR24 // 24- dehydrocholesterol reductase // 1p33- p31.1 // 1718 /// 3231186 C9orf37 NM_032937 // NM_032937 −0.31 0.09 0.008 C9orf37 // chromosome 9 open reading frame 37 // 9q34.3 // 85026 // 2669955 XIRP1 NM_194293 // NM_194293 −0.32 0.11 0.013 XIRP1 // xin actin- binding repeat containing 1 // 3p22.2 // 165904 3345222 AMOTL1 NM_130847 // NM_130847 −0.32 0.11 0.012 AMOTL1 // angiomotin like 1 // 11q14.3 // 154810 /// ENST0000031782 2573326 FLJ14816 BC112205 // BC112205 −0.32 0.11 0.016 FLJ14816 // hypothetical protein FLJ14816 // 2q14.2 // 84931 /// BC1 3349437 UNQ2550 AY358815 // AY358815 −0.32 0.09 0.005 UNQ2550 // SFVP2550 // 11q23.1 // 100130653 3951117 ACR NM_001097 // NM_001097 −0.32 0.12 0.017 ACR // acrosin // 22q13- qter|22q13.33 // 49 /// ENST00000216139 // 2489140 −0.32 0.07 0.002 2562115 LSM3 CR457185 // LSM3 // CR457185 −0.32 0.11 0.011 LSM3 homolog, U6 small nuclear RNA associated (S. cerevisiae 3572975 NGB NM_021257 // NM_021257 −0.33 0.09 0.004 NGB // neuroglobin // 14q24.3 // 58157 /// ENST00000298352 // NGB/ 2439350 OR6N1 NM_001005185 // NM_001005185 −0.33 0.10 0.009 OR6N1 // olfactory receptor, family 6, subfamily N, member 1 // 3590275 CHAC1 NM_024111 // NM_024111 −0.33 0.12 0.014 CHAC1 // ChaC, cation transport regulator homolog 1 (E. coli) // 15 2397898 HSPB7 NM_014424 // NM_014424 −0.33 0.12 0.015 HSPB7 // heat shock 27 kDa protein family, member 7 (cardiovascular) 2364677 PBX1 NM_002585 // NM_002585 −0.34 0.07 0.001 PBX1 // pre-B-cell leukemia homeobox 1 // 1q23 // 5087 /// ENST0000 2474409 DNAJC5G NM_173650 // NM_173650 −0.34 0.09 0.004 DNAJC5G // DnaJ (Hsp40) homolog, subfamily C, member 5 gamma // 2p2 3581373 −0.34 0.12 0.014 3508330 HSPH1 NM_006644 // NM_006644 −0.34 0.13 0.019 HSPH1 // heat shock 105 kDa/110 kDa protein 1 // 13q12.3 // 10808 /// 3751164 DHRS13 NM_144683 // NM_144683 −0.35 0.10 0.006 DHRS13 // dehydrogenase/ reductase (SDR family) member 13 // 17q11.2 2908179 VEGFA NM_001025366 // NM_001025366 −0.35 0.13 0.016 VEGFA // vascular endothelial growth factor A // 6p12 // 7422 // 3962448 dJ222E13.2 NR_002184 // NR_002184 −0.35 0.12 0.014 dJ222E13.2 // similar to CGI-96 // 22q13.2 // 91695 /// BC073834 // 3747638 LOC201164 BC031263 // BC031263 −0.35 0.09 0.004 LOC201164 // similar to CG12314 gene product // 17p11.2 // 201164 // 2821981 TMEM157 NM_198507 // NM_198507 −0.35 0.12 0.015 TMEM157 // transmembrane protein 157 // 5q21.1 // 345757 /// ENST00 3123675 PPP1R3B NM_024607 // NM_024607 −0.35 0.12 0.014 PPP1R3B // protein phosphatase 1, regulatory (inhibitor) subunit 3B 2656837 ST6GAL1 NM_173216 // NM_173216 −0.35 0.13 0.016 ST6GAL1 // ST6 beta-galactosamide alpha-2,6- sialyltranferase 1 // 3 3746574 PMP22 NM_000304 // NM_000304 −0.36 0.09 0.004 PMP22 // peripheral myelin protein 22 // 17p12- p11.2 // 5376 /// NM 2771342 EPHA5 NM_004439 // NM_004439 −0.36 0.09 0.003 EPHA5 // EPH receptor A5 // 4q13.1 // 2044 /// NM_182472 // EPHA5/ 2888674 MXD3 NM_031300 // NM_031300 −0.36 0.12 0.012 MXD3 // MAX dimerization protein 3 // 5q35.3 // 83463 /// ENST00000 2353477 ATP1A1 NM_000701 // NM_000701 −0.36 0.11 0.007 ATP1A1 // ATPase, Na+/K+ transporting, alpha 1 polypeptide // 1p21 3956984 ZMAT5 NM_019103 // NM_019103 −0.36 0.11 0.009 ZMAT5 // zinc finger, matrin type 5 // 22cen-q12.3 // 55954 /// NM 2551651 ATP6V1E2 NM_080653 // NM_080653 −0.37 0.13 0.017 ATP6V1E2 // ATPase, H+ transporting, lysosomal 31 kDa, V1 subunit E2 3578069 C14orf139 BC008299 // BC008299 −0.37 0.13 0.016 C14orf139 // chromosome 14 open reading frame 139 // 14q32.13 // 796 2428501 SLC16A1 NM_003051 // NM_003051 −0.37 0.14 0.018 SLC16A1 // solute carrier family 16, member 1 (monocarboxylic acid 3061621 TFPI2 NM_006528 // NM_006528 −0.37 0.09 0.002 TFPI2 // tissue factor pathway inhibitor 2 // 7q22 // 7980 /// ENST 3705516 LOC100131454 AF229804 // AF229804 −0.38 0.11 0.008 LOC100131454 // similar to hCG1646635 // 17p13.3 // 100131454 /// EN 3306299 XPNPEP1 NM_020383 // NM_020383 −0.38 0.14 0.018 XPNPEP1 // X- prolyl aminopeptidase (aminopeptidase P) 1, soluble // 2763550 PPARGC1A NM_013261 // NM_013261 −0.38 0.13 0.012 PPARGC1A // peroxisome proliferator- activated receptor gamma, coact 2769063 USP46 NM_022832 // NM_022832 −0.38 0.13 0.013 USP46 // ubiquitin specific peptidase 46 // 4q12 // 64854 /// ENST0 3806459 ST8SIA5 NM_013305 // NM_013305 −0.38 0.10 0.004 ST8SIA5 // ST8 alpha-N-acetyl- neuraminide alpha- 2,8-sialyltransfera 3190151 SLC25A25 NM_001006641 // NM_001006641 −0.39 0.09 0.003 SLC25A25 // solute carrier family 25 (mitochondrial carrier; pho 2489172 MTHFD2 NM_001040409 // NM_001040409 −0.39 0.05 0.000 MTHFD2 // methylenetetra- hydrofolate dehydrogenase (NADP+ depende 2952065 PPIL1 NM_016059 // NM_016059 −0.39 0.10 0.005 PPIL1 // peptidylprolyl isomerase (cyclophilin)-like 1 // 6p21.1 // 3382015 CHRDL2 NM_015424 // NM_015424 −0.39 0.10 0.003 CHRDL2 // chordin-like 2 // 11q14 // 25884 /// ENST00000263671 // C 2711139 ATP13A5 NM_198505 // NM_198505 −0.40 0.11 0.005 ATP13A5 // ATPase type 13A5 // 3q29 // 344905 /// ENST00000342358/ 2633917 RG9MTD1 NM_017819 // NM_017819 −0.41 0.14 0.013 RG9MTD1 // RNA (guanine-9-) methyltransferase domain containing 1/ 2974671 C6orf192 NM_052831 // NM_052831 −0.41 0.15 0.018 C6orf192 // chromosome 6 open reading frame 192 // 6q22.3-q23.3 // 2982270 FLJ27255 ENST00000355047 // ENST00000355047 −0.41 0.12 0.007 FLJ27255 // hypothetical LOC401281 // 6q25.3 // 401281 /// AK 2778273 PGDS NM_014485 // NM_014485 −0.41 0.08 0.001 PGDS // prostaglandin D2 synthase, hematopoietic // 4q22.3 // 27306 3005332 RCP9 NM_014478 // NM_014478 −0.41 0.14 0.013 RCP9 // calcitonin gene-related peptide-receptor component protein 2650393 PPM1L NM_139245 // NM_139245 −0.42 0.12 0.006 PPM1L // protein phosphatase 1 (formerly 2C)-like // 3q26.1 // 1517 3463056 CSRP2 NM_001321 // NM_001321 −0.42 0.11 0.005 CSRP2 // cysteine and glycine-rich protein 2 // 12q21.1 // 1466 /// 2459405 −0.43 0.10 0.003 2570238 NPHP1 NM_000272 // NM_000272 −0.43 0.06 0.000 NPHP1 // nephronophthisis 1 (juvenile) // 2q13 // 4867 /// NM_20718 2840616 NPM1 NM_002520 // NM_002520 −0.43 0.14 0.010 NPM1 // nucleophosmin (nucleolar phosphoprotein B23, numatrin) // 5 3601051 NEO1 NM_002499 // NM_002499 −0.43 0.09 0.002 NEO1 // neogenin homolog 1 (chicken) // 15q22.3-q23 // 4756 /// ENS 3936515 TUBA8 NM_018943 // NM_018943 −0.43 0.10 0.002 TUBA8 // tubulin, alpha 8 // 22q11.1 // 51807 /// ENST00000330423/ 2725013 UCHL1 NM_004181 // NM_004181 −0.44 0.11 0.004 UCHL1 // ubiquitin carboxyl-terminal esterase L1 (ubiquitin thioles 2380590 TGFB2 NM_003238 // NM_003238 −0.44 0.16 0.017 TGFB2 // transforming growth factor, beta 2 // 1q41 // 7042 /// ENS 2496382 NPAS2 NM_002518 // NM_002518 −0.46 0.10 0.002 NPAS2 // neuronal PAS domain protein 2 // 2q11.2 // 4862 /// ENST00 3841574 LILRB1 NM_006669 // NM_006669 −0.46 0.16 0.015 LILRB1 // leukocyte immunoglobulin- like receptor, subfamily B (with 3726960 NME2 NM_001018137 // NM_001018137 −0.47 0.16 0.013 NME2 // non- metastatic cells 2, protein (NM23B) expressed in // 2649367 PTX3 NM_002852 // NM_002852 −0.47 0.11 0.002 PTX3 // pentraxin- related gene, rapidly induced by IL-1 beta // 3q2 2909483 GPR111 NM_153839 // NM_153839 −0.47 0.13 0.006 GPR111 // G protein-coupled receptor 111 // 6p12.3 // 222611 /// EN 2881950 SLC36A2 NM_181776 // NM_181776 −0.48 0.12 0.004 SLC36A2 // solute carrier family 36 (proton/amino acid symporter), 3441190 FGF6 NM_020996 // NM_020996 −0.48 0.12 0.004 FGF6 // fibroblast growth factor 6 // 12p13 // 2251 /// ENST0000022 3028911 C7orf34 NM_178829 // NM_178829 −0.49 0.18 0.019 C7orf34 // chromosome 7 open reading frame 34 // 7q34 // 135927 /// 2830861 EGR1 NM_001964 // NM_001964 −0.49 0.19 0.020 EGR1 // early growth response 1 // 5q31.1 // 1958 /// ENST000002399 3323891 GAS2 NM_177553 // NM_177553 −0.49 0.16 0.011 GAS2 // growth arrest-specific 2 // 11p14.3-p15.2 // 2620 /// NM_00 2497252 SLC9A2 NM_003048 // NM_003048 −0.50 0.11 0.002 SLC9A2 // solute carrier family 9 (sodium/hydrogen exchanger), memb 3018484 GPR22 NM_005295 // NM_005295 −0.51 0.15 0.008 GPR22 // G protein-coupled receptor 22 // 7q22- q31.1 // 2845 /// EN 2712632 TFRC NM_003234 // NM_003234 −0.51 0.12 0.003 TFRC // transferrin receptor (p90, CD71) // 3q29 // 7037 /// ENST00 3214451 NFIL3 NM_005384 // NM_005384 −0.53 0.14 0.004 NFIL3 // nuclear factor, interleukin 3 regulated // 9q22 // 4783 // 2435981 S100A12 NM_005621 // NM_005621 −0.54 0.19 0.014 S100A12 // S100 calcium binding protein A12 // 1q21 // 6283 /// ENS 3320675 RIG U32331 // RIG // U32331 −0.54 0.10 0.001 regulated in glioma // 11p15.1 // 10530 3290746 SLC16A9 NM_194298 // NM_194298 −0.54 0.15 0.006 SLC16A9 // solute carrier family 16, member 9 (monocarboxylic acid 3055703 NSUN5C NM_032158 // NM_032158 −0.57 0.17 0.008 NSUN5C // NOL1/NOP2/Sun domain family, member 5C // 7q11.23 // 2602 3265494 TRUB1 NM_139169 // NM_139169 −0.57 0.17 0.008 TRUB1 // TruB pseudouridine (psi) synthase homolog 1 (E. coli) // 1 3374213 OR1S2 NM_001004459 // NM_001004459 −0.58 0.20 0.013 OR1S2 // olfactory receptor, family 1, subfamily S, member 2 // 3318253 OR51L1 NM_001004755 // NM_001004755 −0.59 0.18 0.009 OR51L1 // olfactory receptor, family 51, subfamily L, member 1/ 3294280 DNAJC9 NM_015190 // NM_015190 −0.59 0.22 0.018 DNAJC9 // DnaJ (Hsp40) homolog, subfamily C, member 9 // 10q22.2 // 2899095 HIST1H4A NM_003538 // NM_003538 −0.60 0.16 0.005 HIST1H4A // histone cluster 1, H4a // 6p21.3 // 8359 /// ENST000003 2378068 G0S2 NM_015714 // NM_015714 −0.63 0.22 0.016 G0S2 // G0/G1switch 2 // 1q32.2-q41 // 50486 /// ENST00000367029 // 3737677 LOC100129503 AF218021 // AF218021 −0.64 0.19 0.007 LOC100129503 // hypothetical protein LOC100129503 // 17q25.3 // 1001 3300115 PPP1R3C NM_005398 // NM_005398 −0.69 0.26 0.020 PPP1R3C // protein phosphatase 1, regulatory (inhibitor) subunit 3C 3279058 ACBD7 NM_001039844 // NM_001039844 −0.69 0.13 0.001 ACBD7 // acyl- Coenzyme A binding domain containing 7 // 10p13 // 4031156 RPS4Y2 NM_001039567 // NM_001039567 −0.71 0.17 0.003 RPS4Y2 // ribosomal protein S4, Y-linked 2 // Yq11.223 // 140032 2979246 RAET1L NM_130900 // NM_130900 −0.75 0.26 0.013 RAET1L // retinoic acid early transcript 1L // 6q25.1 // 154064 /// 3321150 ARNTL NM_001178 // NM_001178 −0.80 0.20 0.004 ARNTL // aryl hydrocarbon receptor nuclear translocator-like // 11p 3862873 CYP2A6 NM_000762 // NM_000762 −1.12 0.34 0.009 CYP2A6 // cytochrome P450, family 2, subfamily A, polypeptide 6 //

4. Identification of Ursolic Acid as an Inhibitor of Fasting-Induced Muscle Atrophy.

The Connectivity Map describes the effects of >1300 bioactive small molecules on global mRNA expression in several cultured cell lines, and contains search algorithms that permit comparisons between compound-specific mRNA expression signatures and mRNA expression signatures of interest (Lamb J, et al. (2006) Science (New York, N.Y. 313(5795):1929-1935). It was hypothesized herein that querying the Connectivity Map with the mRNA expression signature of fasting (atrophy signature-1) would identify inhibitors of atrophy-associated gene expression and thus, potential inhibitors of muscle atrophy. It was also reasoned herein that increasing the specificity of the query would enhance the output. To this end, as described herein, an evolutionarily conserved mRNA expression signature of fasting was discovered by comparing the effect of fasting on human skeletal muscle to the effect of a 24 h fast on mouse skeletal muscle. The mouse studies were described previously (Ebert S M, et al. (2010) Molecular endocrinology 24(4):790-799). Altogether, 35 mRNAs that were increased by fasting and 40 mRNAs that were decreased by fasting were identified in both human and mouse skeletal muscle (Table 2; the data in column labeled “Change” show mean changes in loge hybridization signals between fasting and fed states for the species indicated, [Mean log2 mRNA levels for fasted] minus [Mean loge mRNA levels in unfasted]; P-values were determined with paired t-tests). The data shown in Table 2 includes all mRNAs whose levels were increased by fasting in human muscle (P≤0.02) and in mouse muscle (P≤0.05), and all mRNAs whose levels were decreased by fasting in human muscle (P≤0.02) and in mouse muscle (P≤0.05). Of the mRNAs shown in Table 2, 63 mRNAs were represented on the HG-U133A arrays used in the Connectivity Map (FIG. 4A). These mRNAs (31 increased by fasting and 32 decreased by fasting) were used to query the Connectivity Map for candidate small molecule inhibitors of muscle atrophy.

TABLE 2 FASTING-REGULATED MRNAs COMMON TO HUMAN AND MOUSE SKELETAL MUSCLE. Human Mouse Mean Log2 Mean Log2 Change Change (Fasting- (Fasting- mRNA Protein Fed) P Fed) P PDK4 pyruvate dehydrogenase 2.15 0.000 1.91 0.000 kinase, isozyme 4 TXNIP thioredoxin interacting 0.85 0.004 0.60 0.038 protein FBXO32 F-box protein 32 0.82 0.002 2.13 0.000 SLC38A2 solute carrier family 38, 0.62 0.001 0.33 0.036 member 2 UCP3 uncoupling protein 3 0.59 0.000 1.02 0.001 (mitochondrial, proton carrier) ZFAND5 zinc finger, AN1-type 0.51 0.005 0.57 0.001 domain 5 HMOX1 heme oxygenase 0.46 0.006 0.17 0.035 (decycling) 1 SESN1 sestrin 1 0.46 0.004 1.51 0.001 GABARAPL1 GABA(A) receptor- 0.39 0.004 1.18 0.000 associated protein like 1 CAT catalase 0.39 0.003 0.85 0.001 CITED2 Cbp/p300-interacting 0.37 0.005 0.29 0.010 transactivator, with Glu/Asp- rich carboxy-terminal domain ABCA1 ATP-binding cassette, sub- 0.37 0.016 0.26 0.018 family A (ABC1), member 1 FBXL20 F-box and leucine-rich 0.35 0.002 0.46 0.001 repeat protein 20 XPO4 exportin 4 0.31 0.009 0.22 0.022 HERPUD1 homocysteine-inducible, 0.29 0.003 0.27 0.029 endoplasmic reticulum stress-inducible, ubiquitin- like domain 1 ACOX1 acyl-Coenzyme A oxidase 0.29 0.013 0.53 0.006 1, palmitoyl NOX4 NADPH oxidase 4 0.28 0.002 0.41 0.018 UBE4A ubiquitination factor E4A 0.27 0.004 1.08 0.010 (UFD2 homolog, yeast) INSR insulin receptor 0.24 0.014 0.58 0.003 IGF1R insulin-like growth factor 1 0.23 0.013 0.40 0.001 receptor PANK1 pantothenate kinase 1 0.21 0.007 0.78 0.000 NBR1 neighbor of BRCA1 gene 1 0.21 0.017 0.39 0.009 RORA RAR-related orphan 0.21 0.006 0.39 0.006 receptor A TMEM71 transmembrane protein 71 0.21 0.009 0.40 0.008 CPT1A carnitine 0.21 0.001 0.21 0.020 palmitoyltransferase 1A (liver) UCP2 uncoupling protein 2 0.20 0.005 0.33 0.024 (mitochondrial, proton carrier) TULP3 tubby like protein 3 0.19 0.008 0.22 0.008 MED13L mediator complex subunit 0.18 0.000 0.23 0.011 13-like CALCOCO1 calcium binding and coiled 0.16 0.010 0.31 0.028 coil domain 1 MYO5A myosin VA (heavy chain 12, 0.14 0.006 0.36 0.012 myoxin) PPAP2B phosphatidic acid 0.13 0.007 0.09 0.029 phosphatase type 2B SRRM2 serine/arginine repetitive 0.13 0.007 0.24 0.040 matrix 2 ADPGK ADP-dependent 0.13 0.007 0.16 0.009 glucokinase SUPT6H suppressor of Ty 6 homolog 0.11 0.005 0.26 0.036 (S. cerevisiae) SFRS8 splicing factor, 0.08 0.016 0.13 0.011 arginine/serine-rich 8 NFYA nuclear transcription factor −0.07 0.011 −0.31 0.045 Y, alpha MRPS15 mitochondrial ribosomal −0.11 0.003 −0.25 0.001 protein S15 PDE7B phosphodiesterase 7B −0.12 0.013 −0.51 0.011 WDR1 WD repeat domain 1 −0.14 0.019 −0.21 0.047 ACACA acetyl-Coenzyme A −0.15 0.010 −0.22 0.041 carboxylase alpha AXIN2 axin 2 (conductin, axil) −0.15 0.013 −0.12 0.046 CASQ1 calsequestrin 1 (fast-twitch, −0.16 0.015 −0.26 0.015 skeletal muscle) ZNF280B zinc finger protein 280B −0.16 0.005 −0.34 0.046 JTB jumping translocation −0.16 0.014 −0.42 0.030 breakpoint CACNB1 calcium channel, voltage- −0.17 0.013 −0.43 0.003 dependent, beta 1 subunit ALG2 asparagine-linked −0.17 0.011 −0.39 0.019 glycosylation 2 homolog TSPAN13 tetraspanin 13 −0.18 0.006 −0.30 0.028 P4HA2 procollagen-proline, 2- −0.18 0.007 −0.12 0.012 oxoglutarate 4-dioxygenase, alpha II polypeptide TTLL1 tubulin tyrosine ligase-like −0.18 0.001 −0.29 0.043 family, member 1 SUV39H2 suppressor of variegation 3- −0.20 0.011 −0.26 0.014 9 homolog 2 (Drosophila) SLC4A4 solute carrier family 4, −0.20 0.007 −0.69 0.003 sodium bicarbonate cotransporter, member 4 DNMT3A DNA (cytosine-5-)- −0.20 0.007 −0.48 0.000 methyltransferase 3 alpha FEZ2 fasciculation and elongation −0.21 0.000 −0.50 0.019 protein zeta 2 (zygin II) MTSS1 metastasis suppressor 1 −0.21 0.009 −0.22 0.033 TMTC4 transmembrane and −0.22 0.010 −0.17 0.035 tetratricopeptide repeat containing 4 PPM1J protein phosphatase 1J −0.23 0.003 −0.30 0.012 (PP2C domain containing) ARHGAP20 Rho GTPase activating −0.23 0.003 −0.22 0.013 protein 20 ABTB2 ankyrin repeat and BTB −0.25 0.010 −0.18 0.005 (POZ) domain containing 2 CNNM4 cyclin M4 −0.26 0.016 −0.27 0.005 GRTP1 growth hormone regulated −0.26 0.015 −0.54 0.002 TBC protein 1 RNF148 ring finger protein 148 −0.27 0.017 −0.35 0.014 SPINT2 serine peptidase inhibitor, −0.27 0.017 −0.23 0.026 Kunitz type, 2 PBX1 pre-B-cell leukemia −0.34 0.001 −0.22 0.000 homeobox 1 HSPH1 heat shock 105 kDa/110 kDa −0.34 0.019 −0.20 0.043 protein 1 VEGFA vascular endothelial growth −0.35 0.016 −0.26 0.002 factor A PMP22 peripheral myelin protein 22 −0.36 0.004 −0.13 0.012 PPARGC1A peroxisome proliferative −0.38 0.012 −0.39 0.030 activated receptor, gamma, coactivator 1 alpha ST8SIA5 ST8 alpha-N-acetyl- −0.38 0.004 −0.48 0.011 neuraminide alpha-2,8- sialyltransferase 5 PPIL1 peptidylprolyl isomerase −0.39 0.005 −0.52 0.016 (cyclophilin)-like 1 PPM1L protein phosphatase 1 −0.42 0.006 −0.46 0.000 (formerly 2C)-like NEO1 neogenin homolog 1 −0.43 0.002 −0.31 0.037 (chicken) TGFB2 transforming growth factor, −0.44 0.017 −0.30 0.003 beta 2 PTX3 pentraxin-related gene, −0.47 0.002 −0.48 0.000 rapidly induced by IL-1 beta GAS2 growth arrest-specific 2 −0.49 0.011 −0.23 0.044 TFRC transferrin receptor (p90, −0.51 0.003 −1.37 0.011 CD71)

The left side of FIG. 4B shows the 10 Connectivity Map instances (or data sets) with the most significant positive correlations (P<0.004) to the effect of fasting in skeletal muscle. The connectivity score, represented on the y-axis, is a measure of the strength of the correlation (Lamb J, et al. (2006) Science (New York, N.Y. 313(5795):1929-1935); the compound and cell-line is shown below the bar representing the Connectivity Score. Of these, 6 involved wortmannin or LY-294002 (inhibitors of phosphoinositide 3-kinase (PI3K)) or rapamycin (an inhibitor of the mammalian target of rapamycin complex 1 (mTORC1)). Since PI3K and mTORC1 mediate effects of insulin and IGF-I, and since insulin/IGF-I signaling inhibits muscle atrophy and atrophy-associated changes in skeletal muscle mRNA expression (Bodine S C, et al. (2001) Nat Cell Biol 3(11):1014-1019; Sandri M, et al. (2004) Cell 117(3):399-412), these results lent confidence that the Connectivity Map might be used to identify potential inhibitors of muscle atrophy. The right side of FIG. 4B shows the 10 Connectivity Map instances with the most significant negative correlations (P<0.004) to the effect of fasting in skeletal muscle. These compounds, whose effects on cultured cell lines were opposite to the effect of fasting on muscle, included metformin (an insulin-sensitizing agent widely used to treat type 2 diabetes), as well as ursolic acid. Further experiments focused on metformin and ursolic acid. To test the hypothesis that metformin and ursolic acid might reduce fasting-induced muscle atrophy, each compound was administered, or vehicle alone, via i.p. injection to C57BL/6 mice. The mice were then fasted, and after 12 hours of fasting, the mice received a second dose of the compound or vehicle. After 24 hours of fasting, the blood glucose was measured and muscles were harvested. The data shown in FIGS. 4C-4H are means±SEM from 16 mice. Both metformin (250 mg/kg) and ursolic acid (200 mg/kg) significantly reduced fasting blood glucose (FIGS. 4C and 4D). The effects of metformin and ursolic acid on fasting-induced muscle atrophy were also examined, i.e. the effect of 24 h fast (relative to ad lib feeding) on wet weight of lower hindlimb skeletal muscle (bilateral tibialis anterior (“TA” muscle), gastrocnemius, and soleus; see FIGS. 4E-4G). In the absence of metformin and ursolic acid, fasting reduced muscle weight by 9% (FIG. 4E). Although metformin did not alter muscle weight in fasted mice (FIG. 4F), ursolic acid increased it by 7±2% (FIG. 4G). Moreover, consistent with the predicted inhibitory effect on fasting-induced gene expression described herein, ursolic acid reduced fasting levels of atrogin-1 and MuRF1 mRNA levels in the TA muscles of fasted mice (FIG. 4H; the data shown are normalized to the levels in vehicle-treated mice, which were set at 1). In FIGS. 4E-4H, each data point represents one mouse and the horizontal bars denote the means. In FIGS. 4C-4H, P-values were determined using unpaired t-tests. Thus, ursolic acid, but not metformin, decreased fasting-induced muscle atrophy.

5. Ursolic Acid Reduces Denervation-Induced Muscle Atrophy.

The Connectivity Map was queried with a second mRNA expression signature, atrophy signature-2 (described above), to determine if this muscle atrophy signature would also correlate with ursolic acid, among other compounds. As described above, atrophy signature-2 was an mRNA expression signature identified as described herein for human skeletal muscle mRNAs that were induced or repressed by fasting and also by spinal cord injury (“SCI”). The studies of the effects of SCI on human skeletal muscle gene expression were described previously (Adams C M, et al. (2011) Muscle Nerve. 43(1):65-75). Using this approach with the muscle atrophy expression signatures described herein, there were 18 human mRNAs that were increased by fasting and SCI, and 17 human mRNAs that were decreased by fasting and SCI, and are shown in Table 3 (“Change” represents mean changes in loge hybridization signals for pairs as indicated, e.g. fasting and fed states for column labeled “(Fasting-Fed)” or untrained and trained for the column labeled “(Untrained-Trained)”). The data in Table 3 include all mRNAs whose levels were increased by fasting (P≤0.02) and by SCI (P≤0.05), and all mRNAs whose levels were decreased by fasting (P≤0.02) and by SCI (P≤0.05). P-values in Table 3 were determined with paired t-tests.

TABLE 3 HUMAN SKELETAL MUSCLE MRNAS INDUCED OR REPRESSED BY FASTING AND SCI. EFFECT OF FASTING EFFECT OF SCI Change Change (Fasting- (Untrained- mRNA Protein Fed) P Trained) P OR1D4 olfactory receptor, family 1, 0.50 0.019 0.65 0.030 subfamily D, member 4 RHOBTB1 Rho-related BTB domain 0.48 0.001 0.71 0.032 containing 1 TSPAN8 tetraspanin 8 0.39 0.015 1.79 0.023 FLJ33996 hypothetical protein FLJ33996 0.39 0.019 0.68 0.020 NUPR1 nuclear protein 1 0.35 0.007 0.65 0.030 IRS2 insulin receptor substrate 2 0.34 0.004 0.21 0.035 NPC2 Niemann-Pick disease, type C2 0.30 0.011 0.39 0.042 KLF11 Kruppel-like factor 11 0.29 0.011 0.22 0.034 ZNF682 zinc finger protein 682 0.28 0.017 0.72 0.013 NOX4 NADPH oxidase 4 0.28 0.002 0.56 0.007 PLXDC2 plexin domain containing 2 0.26 0.013 0.38 0.022 CTDSP2 CTD small phosphatase 2 0.25 0.003 0.34 0.021 CAV3 caveolin 3 0.24 0.007 0.56 0.020 IGF1R insulin-like growth factor 1 0.23 0.013 0.63 0.040 receptor FLJ14154 hypothetical protein FLJ14154 0.22 0.005 0.30 0.021 CUGBP2 CUG triplet repeat, RNA 0.21 0.004 0.14 0.034 binding protein 2 MLL myeloid/lymphoid or mixed- 0.14 0.016 0.30 0.040 lineage leukemia SUPT6H suppressor of Ty 6 homolog 0.11 0.005 0.19 0.024 MRPS15 mitochondrial ribosomal protein −0.11 0.003 −0.33 0.001 S15 RFXDC2 regulatory factor X domain −0.12 0.012 −0.10 0.037 containing 2 PDE7B phosphodiesterase 7B −0.12 0.013 −0.39 0.011 PFDN6 prefoldin subunit 6 −0.14 0.014 −0.42 0.021 ZNF280B zinc finger protein 280B −0.16 0.005 −0.30 0.028 TSPAN13 tetraspanin 13 −0.18 0.006 −0.56 0.023 TTLL1 tubulin tyrosine ligase-like −0.18 0.001 −0.37 0.020 family, member 1 CMAS cytidine monophosphate N- −0.21 0.000 −0.22 0.025 acetylneuraminic acid synthetase C8orf32 chromosome 8 open reading −0.23 0.016 −0.11 0.049 frame 32 GUCY1B3 guanylate cyclase 1, soluble, −0.24 0.007 −0.24 0.008 beta 3 ZNF32 zinc finger protein 32 −0.24 0.010 −0.21 0.030 VLDLR very low density lipoprotein −0.28 0.007 −0.16 0.015 receptor HSPB7 heat shock 27 kDa protein −0.33 0.015 −0.77 0.032 family, member 7 (cardiovascular) VEGFA vascular endothelial growth −0.35 0.016 −0.43 0.020 factor A SLC16A1 solute carrier family 16, −0.37 0.018 −0.94 0.015 member 1 PPARGC1A peroxisome proliferative −0.38 0.012 −0.74 0.001 activated receptor, gamma, coactivator 1 alpha C6orf192 chromosome 6 open reading −0.41 0.018 −0.39 0.042 frame 192

Of the mRNAs listed in Table 3, 29 were represented on the HG-U133A arrays used in the Connectivity Map (FIG. 5A), but only 10 were common to the 63 mRNAs used in the first Connectivity Map query described above for atrophy signature-1 (IGF-IR, NOX4, SUPT6H, MRPS15, PDE7B, PGC-1a, TSPAN13, TTLL1, VEGFA and ZNF280B). The mRNAs listed in FIG. 5A represent human muscle atrophy signature-2: mRNAs altered by both fasting and SCI in human muscle. These mRNAs, as described above, were used to query the Connectivity Map. Inclusion criteria were: P≤0.02 in fasted human muscle (by t-test), P≤0.05 in untrained, paralyzed muscle (by t-test), and the existence of complimentary probes on HG-U133A arrays. Connectivity Map instances with the most significant positive and negative correlations to the effect of fasting and SCI in human muscle. P<0.005 for all compounds are shown in FIG. 5B. The results partially overlapped with the results of the first search: both search strategies identified LY-294002, wortmannin and rapamycin as predicted mimics of atrophy-inducing stress, and ursolic acid (but not metformin) as a predicted inhibitor (FIG. 5B).

Because atrophy signature-2 utilized data from SCI subjects, it was hypothesized that ursolic acid might reduce denervation-induced muscle atrophy. To test this, the left hindlimb muscles a denervation-induced skeletal muscle atrophy model in mouse was used. Briefly, on day 0, the left hindlimbs of C57BL/6 mice were denervated by transsecting the left sciatic nerve. This approach allowed the right hindlimb to serve as an intra-subject control. Mice were then administered ursolic acid (200 mg/kg) or an equivalent volume of vehicle alone (corn oil) via i.p. injection twice daily for seven days. During this time, mice continued to have ad libitum access to food. On day 7, muscle tissues were harvested for analysis, and the left (denervated) and right (innervated) hindlimb muscles in both groups (ursolic acid vs. vehicle administration) were compared. Ursolic acid significantly decreased denervation-induced muscle loss (FIG. 5C). In FIG. 5C, weights of the left (denervated) lower hindlimb muscles were normalized to weights of the right (innervated) lower hindlimb muscles from the same mouse. Each data point represents one mouse, and horizontal bars denote the means and the P-value was determined using an unpaired t-test. Histologically, this effect of ursolic acid was reflected as an increase in the size of denervated skeletal muscle fiber diameter in denervated gastrocnemius (D) and TA (E) muscles (FIGS. 5D and 5E, respectivel). The data shown in FIGS. 5D and 5E are from >2500 muscle fibers per condition; P<0.0001 by unpaired t-test. Thus, ursolic acid reduced denervation-induced muscle atrophy.

6. Ursolic Acid Induces Skeletal Muscle Hypertrophy.

The results from the denervation-induced muscle atrophy model suggested that ursolic acid reduced muscle atrophy, thus the hypothesis that ursolic acid might promote muscle hypertrophy in the absence of an atrophy-inducing stress was reasonable. Mice were provided ad lib access to either standard chow (control diet) or standard chow supplemented with 0.27% ursolic acid (ursolic acid diet) for 5 weeks before grip strength was measured and tissues were harvested. After five weeks, mice administered ursolic had increased lower hindlimb muscle weight (FIG. 6A), quadriceps weight (FIG. 6B), and upper forelimb muscle (triceps and biceps) weight (FIG. 6C). Each data point in FIGS. 6A-6C represents one mouse, and horizontal bars denote the means. The effect of ursolic acid in this study on skeletal muscle fiber size distribution is shown in FIG. 6D. Each distribution represents measurements of >800 triceps muscle fibers from 7 animals (>100 measurements/animal); P<0.0001. The effect of ursolic acid on peak grip strength (normalized to body weight) is shown in FIG. 6E. Each data point represents one mouse, and horizontal bars denote the means. Non-normalized grip strength data were 157±9 g (control diet) and 181±6 g (ursolic acid diet) (P=0.04).

Moreover, dietary ursolic acid increased the specific force generated by muscles ex vivo (FIG. 7). Briefly, six-week old male C57BL/6 mice were provided either standard diet or diet containing 0.27% ursolic acid for 16 weeks before being euthanized. The lower hindlimb was removed (by transsecting the upper hindlimb mid-way through the femur), and placed in Krebs solution aerated with 95% O2 and 5% CO2. The gastrocnemius, soleus and tibialis anterior muscles, as well as the distal half of the tibia and fibula were then removed and discarded, leaving the extensor digitorum longus and peroneus muscles with their origins and insertions intact. A suture was placed through the proximal tendon and secured to the distal femur fragment. This ex vivo preparation was then mounted vertically in a water jacket bath (Aurora Scientific 1200A Intact Muscle Test System, filled with aerated Krebs solution) by attaching the suture to a servo-controlled lever (superiorly) and clamping the metatarsals (inferiorly). Passive muscle force was adjusted to a baseline of 1 g, and then muscles were stimulated with supramaximal voltage (80 V) at 100 Hz. The mean time from euthanasia to maximal force measurements was 10 min. After force measurements, muscles were removed and weighed in order to calculate specific titanic force. Maximal tetanic force and muscle weight did not differ between the two groups (P=0.20 and 0.26, respectively). Data are means±SEM from 5-6 mice per diet. P-values were determined with a t-test. Together, the data in FIGS. 6 and 7 provide morphological and functional evidence that ursolic acid induced skeletal muscle hypertrophy.

7. Ursolic Acid Induces Trophic Changes in Skeletal Muscle Gene Expression.

The foregoing results suggested that ursolic acid might alter skeletal muscle gene expression. To test this hypothesis, an unbiased approach was used, specifically exon expression arrays were used to analyze gastrocnemius muscle mRNA expression in mice that had been fed diets lacking or containing ursolic acid for 5 weeks. Mice were provided ad lib access to either standard chow (control diet) or standard chow supplemented with 0.27% ursolic acid (ursolic acid diet) for 5 weeks before gastrocnemius muscle RNA was harvested and analyzed by Affymetrix Mouse Exon 1.0 ST arrays (n=4 arrays per diet). Each array assessed pooled gastrocnemius RNA from two mice. Stringent criteria were used for ursolic acid-induced effects on mRNA levels (P<0.005), and mRNAs with low levels of expression were disregarded (i.e. only transcripts that were increased to a mean loge hybridization signal≥8, or repressed from a mean log2 hybridization signal≥8 were included). The results were that ursolic acid decreased 18 mRNAs and increased 51 mRNAs (out of >16,000 mRNAs analyzed. The results are shown in Table 4 (“Change” is the meang loge change or difference between mice on ursolic acid diet and control diet, i.e. [Mean loge mRNA levels in ursolic acid diet] minus [Mean loge mRNA levels in control diet]).

TABLE 4 MOUSE SKELETAL MUSCLE MRNAS INDUCED OR REPRESSED BY URSOLIC ACID. mRNA Protein Change P Smox spermine oxidase 0.81 0.001 Lyz2 lysozyme 2 0.71 0.001 C3 complement component 3 0.70 0.000 Tyrobp TYRO protein tyrosine kinase binding 0.69 0.001 protein Lum lumican 0.61 0.001 Igf1 insulin-like growth factor 1 0.56 0.005 Fmo1 flavin containing monooxygenase 1 0.47 0.000 Ostn osteocrin 0.43 0.001 Nampt nicotinamide phosphoribosyltransferase 0.41 0.003 H19 H19 fetal liver mRNA 0.39 0.004 Hipk2 homeodomain interacting protein kinase 2 0.38 0.002 Fbp2 fructose bisphosphatase 2 0.37 0.003 Gpx1 glutathione peroxidase 1 0.36 0.001 Sepp1 selenoprotein P, plasma, 1 0.35 0.004 Parp3 poly (ADP-ribose) polymerase family, 0.32 0.001 member 3 Hspb8 heat shock protein 8 0.32 0.000 Musk muscle, skeletal, receptor tyrosine kinase 0.31 0.004 Fhl3 four and a half LIM domains 3 0.31 0.005 Hsph1 heat shock 105 kDa/110 kDa protein 1 0.30 0.001 Arfgap2 ADP-ribosylation factor GTPase activating 0.30 0.001 protein 2 Cd24a CD24a antigen 0.28 0.002 Sepx1 selenoprotein X 1 0.28 0.003 Hk2 hexokinase 2 0.26 0.003 Ggct gamma-glutamyl cyclotransferase 0.24 0.005 Trip10 thyroid hormone receptor interactor 10 0.23 0.000 Npc1 Niemann Pick type C1 0.22 0.001 Asb5 ankyrin repeat and SOCs box-containing 5 0.21 0.001 Vps29 vacuolar protein sorting 29 (S. pombe) 0.20 0.000 Ahsa2 AHA1, activator of heat shock protein 0.18 0.001 ATPase homolog 2 Lsm14a LSM14 homolog A (SCD6, S. cerevisiae) 0.18 0.004 Pdha1 pyruvate dehydrogenase E1 alpha 1 0.18 0.001 Trappc2l trafficking protein particle complex 2-like 0.16 0.004 Ube2l3 ubiquitin-conjugating enzyme E2L 3 0.16 0.003 Ctsb cathepsin B 0.16 0.003 D0H4S114 DNA segment, human D4S114 0.15 0.004 Psma2 proteasome (prosome, macropain) 0.15 0.005 subunit, alpha type 2 Mrpl46 mitochondrial ribosomal protein L46 0.15 0.001 Eef1e1 eukaryotic translation elongation factor 1 0.15 0.002 epsilon 1 Krr1 KRR1, small subunit (SSU) processome 0.15 0.005 component, homolog Ndufaf4 NADH dehydrogenase (ubiquinone) 1 0.14 0.005 alpha subcomplex, assembly factor 4 Ndufs2 NADH dehydrogenase (ubiquinone) Fe—S 0.14 0.002 protein 2 2610507B11Rik RIKEN cDNA 2610507B11 gene 0.14 0.000 Ssr4 signal sequence receptor, delta 0.14 0.000 Ndufs4 NADH dehydrogenase (ubiquinone) Fe—S 0.14 0.003 protein 4 Sqstm1 sequestosome 1 0.12 0.001 Gfm1 G elongation factor, mitochondrial 1 0.12 0.003 2310016M24Rik RIKEN cDNA 2310016M24 gene 0.12 0.004 Sod2 superoxide dismutase 2, mitochondrial 0.12 0.001 Prdx5 peroxiredoxin 5 0.10 0.005 BC004004 cDNA sequence BC004004 0.06 0.001 Ghitm growth hormone inducible transmembrane 0.05 0.005 protein Foxn3 forkhead box N3 −0.09 0.000 Klhl31 kelch-like 31 (Drosophila) −0.09 0.001 Acadm acyl-Coenzyme A dehydrogenase, −0.11 0.001 medium chain Eif4g3 eukaryotic translation initiation factor 4 −0.12 0.005 gamma, 3 Nrap nebulin-related anchoring protein −0.14 0.003 Golga4 golgi autoantigen, golgin subfamily a, 4 −0.14 0.003 Paip2b poly(A) binding protein interacting protein −0.16 0.000 2B Pde4dip phosphodiesterase 4D interacting protein −0.18 0.001 (myomegalin) Sfpq splicing factor proline/glutamine rich −0.18 0.005 Pnn pinin −0.18 0.002 D4Wsu53e DNA segment, Chr 4, Wayne State −0.18 0.003 University 53, expressed Mlec malectin −0.19 0.003 Cacna1s calcium channel, voltage-dependent, L −0.22 0.001 type, alpha 1S Sfrs5 splicing factor, arginine/serine-rich 5 −0.22 0.005 (SRp40, HRS) Nnt nicotinamide nucleotide transhydrogenase −0.24 0.002 Adprhl1 ADP-ribosylhydrolase like 1 −0.26 0.002 Ddit4l DNA-damage-inducible transcript 4-like −0.32 0.000 Fbxo32 F-box protein 32 (Atrogin-1) −0.35 0.001

As discussed above, atrogin-1 and MuRF1 are transcriptionally up-regulated by atrophy-inducing stresses (see FIG. 2B and Sacheck J M, et al. (2007) Faseb J21(1):140-155), and they are required for muscle atrophy (Bodine S C, et al. (2001) Science (New York, N.Y. 294(5547):1704-1708). Moreover, in the studies of fasted mice as described herein above, ursolic acid reduced atrogin-1 and MuRF1 mRNAs (FIG. 4H). Consistent with that finding, the arrays indicated that dietary ursolic acid reduced atrogin-1 mRNA, which was the most highly repressed mRNA (FIG. 8A). The results shown in FIG. 8A represent a subset of the mRNAs from Table 4 which had the greatest increase or decrease in expression level in response to ursolic acid. Although MuRF1 mRNA was not measured by the arrays used in these experiments, qPCR analysis confirmed that dietary ursolic acid repressed both atrogin-1 and MuRF1 mRNAs (FIG. 8B; data are means±SEM). Interestingly, one of the most highly up-regulated muscle mRNAs was IGF1 (FIGS. 8A and 8B), which encodes insulin-like growth factor-I (IGF-I), a locally generated autocrine/paracrine hormone. IGF1 mRNA is known to be transcriptionally induced in hypertrophic muscle (Hameed M, et al. (2004) The Journal of physiology 555(Pt 1):231-240; Adams G R & Haddad F (1996) J Appl Physiol 81(6):2509-2516; Gentile M A, et al. (2010) Journal of molecular endocrinology 44(1):55-73). In addition, increased skeletal muscle IGF1 expression reduces denervation-induced muscle atrophy (Shavlakadze T, et al. (2005) Neuromuscul Disord 15(2):139-146), and stimulates muscle hypertrophy (Barton-Davis E R, et al. (1998) Proceedings of the National Academy of Sciences of the United States of America 95(26):15603-15607; Musaro A, et al. (2001) Nature Genetics 27(2):195-200). Moreover, by stimulating skeletal muscle insulin/IGF-I signaling, IGF-I represses atrogin-1 and MuRF mRNAs (Sacheck J M, et al. (2004) Am J Physiol Endocrinol Metab 287(4):E591-601; Frost R A, et al. (2009) J Cell Biochem 108(5):1192-1202.), as well as DDIT4L mRNA (ibid), which, after atrogin-1 mRNA, was the second most highly repressed mRNA in muscle from ursolic acid-treated mice (FIG. 8A). Thus, 5 weeks of dietary ursolic acid altered skeletal muscle gene expression in a manner known to reduce atrophy and promote hypertrophy, and muscle-specific IGF1 induction emerged as a likely contributing mechanism in ursolic acid-induced muscle hypertrophy. The effect of ursolic acid on plasma IGF-I levels was also determined, which primarily reflect growth hormone-mediated hepatic IGF-I production (Yakar S, et al. (1999) Proceedings of the National Academy of Sciences of the United States of America 96(13):7324-7329). Although diets containing 0.14% or 0.27% ursolic acid increased muscle mass (described in greater detail below; FIG. 10A), neither increased plasma IGF-I (FIG. 8C). For the data in FIG. 8C, mice were provided ad lib access to either standard chow (control diet) or standard chow supplemented with the indicated concentration of ursolic acid for 7 weeks before plasma IGF-I levels were measured. Each data point represents one mouse, and horizontal bars denote the means. P-values were determined by one-way ANOVA with Dunnett's post-test. Of note, exon expression arrays indicated that ursolic acid increased levels of all measured IGF1 exons (exons 2-6; FIG. 9A). The data in FIG. 9A are mean exon-specific log2 hybridization signals from the arrays described in Table 2. However, ursolic acid did not alter levels of mRNAs encoding myostatin (which reduces muscle mass, for example see Lee S J (2004) Annu Rev Cell Dev Biol 20:61-86), or twist or myogenin (which are induced by IGF-I during development, for example see Dupont J, et al. (2001) The Journal of biological chemistry 276(28):26699-26707; Tureckova J, et al. (2001) The Journal of biological chemistry 276(42):39264-39270). Moreover, ursolic acid did not alter the amount of IGF1 mRNA in adipose tissue (FIG. 9B). Briefly, the data shown in FIG. 9B were obtained as follows: mice were provided ad lib access to either standard chow (control diet) or standard chow supplemented with 0.27% ursolic acid (ursolic acid diet) for 7 weeks before retroperitoneal adipose tissue was harvested for qPCR quantification of IGF1 mRNA. The data shown are means ±SEM from 5 mice per group. Without wishing to be bound by a particular theory, ursolic acid-mediated IGF1 induction may be localized to skeletal muscle.

8. Ursolic Acid Enhances Skeletal Muscle IGF-I Signaling.

Although muscle-specific IGF1 induction is characteristic of, and contributes to, muscle hypertrophy, it may be a relatively late event that promotes hypertrophy after it has been initiated by other stimuli (Adams G R, et al. (1999) J Appl Physiol 87(5):1705-1712). Without wishing to be bound by a particular theory, it is possible that ursolic acid might have a more proximal effect on insulin/IGF-I signaling. In a previous study of non-muscle cell lines (CHO/IR and 3T3-L1 cells), ursolic acid enhanced insulin-mediated Akt activation (Jung S H, et al. (2007) The Biochemical journal 403(2):243-250). To determine whether ursolic acid might have a similar effect in skeletal muscle, the level of phosphorylated Akt was assessed in quadriceps muscles of mice fed diets lacking or containing ursolic acid. Briefly, mice were provided ad lib access to either standard chow (control diet) or standard chow supplemented with 0.27% ursolic acid for 16 weeks. Total protein extracts from quadriceps muscles were subjected to SDS-PAGE, followed by immunoblot analysis for phosphorylated and total Akt, as indicated. A representative immunoblot is shown in FIG. 8D. Immunoblot data were quantitated as follows: in each mouse, the level of phospho-Akt was normalized to the level of total Akt; these ratios were then normalized to the average phospho-Akt/total Akt ratio from control mice and the results are shown in FIG. 8E (data are means±SEM from 9 mice per diet. P-value was determined by unpaired t-test). The data show that in quadriceps, ursolic acid increased Akt phosphorylation by 1.8-fold.

The effect of ursolic acid on Akt activation was examined in C2C12 skeletal myotubes, a well-established in vitro model of skeletal muscle (Sandri M, et al. (2004) Cell 117(3):399-412; Stitt T N, et al. (2004) Mol Cell 14(3):395-403). Use of an in vitro system, such as C2C12 skeletal myotubes, circumvented potentially confounding effects from non-muscle tissues, and enabled a determination of whether IGF-I or insulin was required for ursolic acid's effect. The latter consideration was important because circulating IGF-I and insulin are always present in healthy animals. Use of an in vitro system also allowed testing of a clearly defined concentration of ursolic acid (10 μM, similar what was used in the Connectivity Map (8.8 μM)) for a clearly defined time of incubation (20 min). These considerations were important because the in vivo pharmacokinetic properties of ursolic acid are not yet known.

For the data shown in FIGS. 8F-8K, serum-starved C2C12 myotubes were treated in the absence or presence of ursolic acid (10 μM) and/or IGF-I (10 nM), as indicated. For studies of the IGF-I receptor, cells were harvested 2 min later, and protein extracts were subjected to immunoprecipitation with anti-IGF-I receptor β antibody, followed by immunoblot analysis with anti-phospho-tyrosine or anti-IGF-I receptor antibodies to assess phospho- and total IGF-I receptor, respectively. For other studies, cells were harvested 20 min after addition of ursolic acid and/or IGF-I, and immunoblot analyses were performed using total cellular protein extracts and antibodies specific for the phosphorylated or total proteins indicated. Representative immunoblots showing effect of ursolic acid on IGF-I-mediated phosphorylation of Akt (FIG. 8F), S6K (FIG. 8G) and IGF-I receptor (FIG. 8H). Data from immunoblots was quantitated as follows: levels in the presence of ursolic acid and IGF-I were normalized to levels in the presence of IGF-I alone, which were set at 1 and are indicated by the dashed line. The data shown in FIG. 8I are means±SEM from ≥3 experiments.

For the data shown in FIGS. 9C-9F, serum-starved C2C12 myotubes were treated in the absence or presence of ursolic acid (10 μM), insulin (10 nM) and/or IGF-I (10 nM), as indicated. For studies of the insulin receptor, cells were harvested 2 min later, and protein extracts were subjected to immunoprecipitation with anti-insulin receptor β antibody, followed by immunoblot analysis with anti-phospho-insulin receptor β (Y1162/1163) or anti-insulin receptor β antibodies to assess phospho- and total insulin receptor, respectively. For other studies, cells were harvested 20 min after addition of ursolic acid, insulin and/or IGF-I, and immunoblot analyses were performed using total cellular protein extracts and antibodies specific for the phosphorylated or total proteins indicated.

When serum-starved myotubes were treated with ursolic acid alone, Akt phosphorylation did not increase (FIG. 8F). However, in the presence of IGF-I, ursolic acid increased Akt phosphorylation by 1.9-fold (FIGS. 8F and 8I). Ursolic acid also increased Akt phosphorylation in the presence of insulin (FIG. 9C). Thus, ursolic acid enhanced IGF-I-mediated and insulin-mediated Akt phosphorylation. The finding that ursolic acid enhanced muscle Akt activity in vivo and in vitro was consistent with the finding that ursolic acid's mRNA expression signature negatively correlated with the mRNA expression signatures of LY-294002 and wortmannin (FIGS. 4B and 5B), which inhibit insulin/IGF-I signaling upstream of Akt. However, ursolic acid's signature also negatively correlated with the signature of rapamycin, which inhibits insulin/IGF-I signaling downstream of Akt.

Although ursolic acid alone did not increase S6K phosphorylation (FIG. 9D), it enhanced IGF-I-mediated and insulin-mediated S6K phosphorylation (FIGS. 8G, 8I and 9D). To further investigate the mechanism, the effect of ursolic acid on the IGF-I receptor was examined. Ursolic acid increased IGF-I receptor phsophorylation in the presence but not the absence of IGF-I (FIGS. 8H and 8I). Similarly, ursolic acid increased insulin receptor phosphorylation in the presence but not the absence of insulin (FIG. 9E). Both of these effects were rapid, occurring within 2 minutes after the addition of ursolic acid and either IGF-I or insulin. Consistent with enhanced signaling at the level of the IGF-I and insulin receptors, ursolic acid also enhanced IGF-I-mediated and insulin-mediated ERK phosphorylation (FIGS. 8J and 9F). Moreover, ursolic acid enhanced IGF-I-mediated phosphorylation (inhibition) of FoxO transcription factors, which activate transcription of atrogin-1 and MuRF1 mRNAs (FIG. 8K; Sandri M, et al. (2004) Cell 117(3):399-412; Stitt T N, et al. (2004) Mol Cell 14(3):395-403.). Without wishing to be bound by a particular theory, ursolic acid represses atrophy-associated gene expression and promotes muscle hypertrophy by increasing activity of the IGF-I and insulin receptors.

9. Ursolic Acid Reduces Adiposity.

Mice were provided ad lib access to standard chow supplemented with the indicated concentration (weight percent in chow, either 0.14% or 0.28% as indicated in FIG. 10) of ursolic acid for 7 weeks before tissues were harvested for analysis. Data are means±SEM from 10 mice per diet. Data for the effects of ursolic acid on weights of skeletal muscle (quadriceps+triceps), epididymal fat, retroperitoneal fat and heart are shown in FIG. 10A. The P-values, determined by one-way ANOVA with post-test for linear trend, were <0.001 for muscle; 0.01 and 0.04 for epididymal and retroperitoneal fat, respectively; and 0.46 for heart. The data show that 7 weeks of dietary ursolic acid increased skeletal muscle weight in a dose-dependent manner, with a peak effect at 0.14% ursolic acid. Interestingly, although ursolic acid increased muscle weight, it did not increase total body weight (FIG. 10B; P-values were 0.71 and 0.80 for initial and final weights, respectively).

The data in FIG. 10A also show that 7 weeks of dietary ursolic acid reduced the weight of epididymal and retroperitoneal fat depots, with a peak effect at 0.14%. In another study, mice were provided ad lib access to either standard chow (control diet) or standard chow supplemented with 0.27% ursolic acid (ursolic acid diet) for 5 weeks. The relationship between skeletal muscle weight (quadriceps, triceps, biceps, TA, gastrocnemius and soleus) and retroperitoneal adipose weight is shown in FIG. 10C. Each data point in FIG. 10C represents one mouse; P<0.001 for both muscle and adipose by unpaired t-test. The data show that 5 weeks of ursolic acid administration (0.14%) also reduced adipose weight. Thus, muscle and fat weights were inversely related. Without wishing to be bound by a particular theory, ursolic acid-treated mice contain less fat because, in part, ursolic acid increases Akt activity (see FIGS. 8 and 9), and muscle-specific increases in Akt activity reduce adiposity as a secondary consequence of muscle hypertrophy (Lai K M, et al. (2004) Molecular and cellular biology 24(21):9295-9304; Izumiya Y, et al. (2008) Cell metabolism 7(2): 159-172).

Ursolic acid reduced adipose weight by reducing adipocyte size as shown by data in FIGS. 10D-10F. FIG. 10D shows a representative H&E stain of retroperitoneal fat for animals feed a control data or a chow with 0.27% ursolic acid as indicated. The data in FIG. 10D are shown quantitatively in FIG. 10E in terms of adipocyte diameter, where data point represents theaverage diameter of ≥125 retroperitoneal adipocytes from one mouse. The retroperitoneal adipocyte size distribution. Each distribution represents combined adipocyte measurements (>1000 per diet) from FIG. 10E.

The changes in adipocyte size were accompanied by a significant reduction in plasma leptin levels, which correlated closely with adipose weight (see FIGS. 10G and 10H). In FIG. 10G, each data point represents one mouse, and horizontal bars denote the means. P-values were determined by t-test. In FIG. 10H, each data point represents one mouse. Importantly, ursolic acid also significantly reduced plasma triglyceride (FIG. 10I) and cholesterol (FIG. 10J). In FIGS. 10I and 10J, each data point represents one mouse, and horizontal bars denote the means. P-values were determined by unpaired t-test. Although ursolic acid reduced leptin, it did not alter food intake (FIG. 11A). In this study, mice were provided ad lib access to either standard chow (control diet) or standard chow supplemented with 0.27% ursolic acid (ursolic acid diet) for 4 weeks. Mice were then moved to a comprehensive animal metabolic monitoring system (CLAMS; Columbus Instruments, Columbus, Ohio) and provided with ad lib access to the same diets. Food consumption was measured for 48 hours. Data are means±SEM from 6 mice per group. However, ursolic acid did not alter weights of heart (FIG. 10A), liver or kidney (FIGS. 11B and 11C), nor did it elevate plasma markers of hepatotoxicity or nephrotoxicity (alanine aminotransferase, bilirubin and creatinine; see FIGS. 11D-11F). The data in FIGS. 11B-11F were obtained as follows: mice were provided ad lib access to either standard chow (control diet) or standard chow supplemented with 0.27% ursolic acid (ursolic acid diet) for 5 weeks before tissues and plasma were harvested for the indicated measurements; each data point represents one mouse, and horizontal bars denote the means. For FIG. 11, P-values were determined with unpaired t-tests. Thus, dietary ursolic acid had two major effects: skeletal muscle hypertrophy and reduced adiposity.

10. Ursolic Acid Reduces Weight Gain and White Adipose Tissue.

The findings that ursolic acid increased skeletal muscle and decreased adiposity suggested that ursolic acid might increase energy expenditure, which would lead to obesity resistance. To test this, C57BL/6 mice were given ad libitum access to a high fat diet (HFD; Teklad TD.93075; 55% calories from fat) lacking or containing 0.27% ursolic acid. After 7 weeks, mice from each group were studied for three days in comprehensive lab animal monitoring systems (“CLAMS”; Columbus Instruments). In the CLAMS, mice were maintained on the same diet they had been eating since the beginning of the experiment. Following CLAMS, tissues were harvested for analysis. In high fat-fed mice, ursolic acid dramatically reduced weight gain, and this effect was apparent within one week (FIG. 12A). As previously observed in mice fed ursolic acid and standard chow (FIG. 6), ursolic acid increased grip strength and muscle mass (FIGS. 12B and 12C). Moreover, ursolic acid reduced retroperitoneal and epididymal fat (FIGS. 12D and 12E). Interestingly, in the scapular fat pad, which contains a mixture of white and thermogenic brown fat, ursolic acid reduced white fat (FIG. 12F), but increased brown fat (FIG. 12G). Importantly, increased skeletal muscle and brown adipose tissue would be predicted to increase energy expenditure. Indeed, CLAMS revealed that ursolic acid increased energy expenditure (FIG. 12H), providing an explanation for how ursolic acid reduces adiposity and obesity. Remarkably, CLAMS analysis revealed that ursolic acid-treated mice consumed more food (FIG. 121), even though they gained less weight (FIG. 12A). For the data shown in FIG. 12A, data are means±SEM from 12 control mice and 15 treated mice, but it should be noted that some error bars are too small to see; P<0.01 at 1 wk and each subsequent time point. In FIGS. 12B-12I, each data point represents one mouse and horizontal bars denote the means. P-values were determined with unpaired t-tests.

11. Ursolic Acid Reduces Obesity-Related Pre-Diabetes, Diabetes, Fatty Liver Disease and Hypercholesterolemia.

The study was carried out as follows: C57BL/6 mice were given ad libitum access to a high fat diet (“HFD”; Teklad TD.93075; 55% calories from fat) lacking or containing 0.27% ursolic acid. After 5 weeks, mice were fasted for 16 h before blood glucose was measured via the tail vein (FIG. 13A). Normal fasting blood glucose: ≤100 mg/dl. (B-I) After 7 weeks, liver and plasma were harvested for analysis (FIGS. 13B-13I). The data shown in FIG. 13A suggest that most mice fed HFD without ursolic acid for 6 weeks developed impaired fasting glucose (pre-diabetes) or diabetes. Importantly, this was prevented by ursolic acid (FIG. 13A). In addition, mice fed HFD without ursolic acid developed fatty liver disease, as evidenced by increased liver weight (>30% increase above normal mouse liver weight of 1500 mg; FIG. 13B), hepatocellular lipid accumulation (FIG. 13C, H&E stain at 20× magnification; FIG. 13D, lipid-staining osmium at 10× magnification), and elevated plasma liver function tests (FIG. 13E, AST; 13F, ALT; 13G, alkaline phosphatase (labeled as “Alk. Phos. in figure); and, 13H, cholesterol). However, ursolic acid prevented all of these hepatic changes (FIG. 13B-13G). In addition, ursolic acid reduced obesity-related hypercholesterolemia (FIG. 13H). In FIGS. 13A, 13B, and 13E-13H, each data point represents one mouse and horizontal bars denote the means.

12. Oleanolic Acid Does Not Increase Skeletal Muscle Mass.

The effect of ursolic acid on skeletal muscle weight and liver weight was compared to the effects by oleanolic acid and metformin. Metformin was a compound identified from atrophy signature-1, but not atrophy signature-2. Oleanolic acid, like ursolic acid is a pentacyclic acid triterpane. This is a structurally similar compound to ursolic acid. However, the two compounds are distinct: oleanolic acid has two methyl groups at position 20, whereas ursolic acid has a single methyl group at each of positions 19 and 20 (compare FIGS. 14A and 14D). Both ursolic acid and oleanolic acid reduce blood glucose, adiposity and hepatic steatosis (Wang Z H, et al. (2010) European journal of pharmacology 628(1-3):255-260; Jayaprakasam B, et al. (2006) J Agric Food Chem 54(1):243-248; de Melo C L, et al. (2010) Chem Biol Interact 185(1):59-65). In addition, both ursolic acid and oleanolic acid possess a large number of cellular effects and biochemical targets, including nearly equivalent inhibition of protein tyrosine phosphatases (“PTPs”; see Zhang W, et al. (2006) Biochimica et biophysica acta 1760(10):1505-1512; Qian S, et al. (2010) J Nat Prod 73(11):1743-1750; Zhang Y N, et al. (2008) Bioorg Med Chem 16(18):8697-8705). However, the effects of these compounds on skeletal muscle mass were not known.

Because some PTPs (particularly PTP1B) dephosphorylate (inactivate) the insulin receptor, PTP inhibition represented a potential mechanism to explain ursolic acid-mediated enhancement of insulin signaling. Thus, because oleanolic acid and ursolic acid inhibit PTP1B and other PTPs with similar efficacy and potency in vitro (Qian S, et al. (2010) J Nat Prod 73(11):1743-1750; Zhang Y N, et al. (2008) Bioorg Med Chem 16(18):8697-8705), testing oleanolic acid's effects on skeletal mass tests the potential role of PTP inhibition. It should be noted that neither ursolic acid nor oleanolic acid is known to inhibit PTPs in vivo, and neither of these compounds are known to enhance IGF-I signaling. Moreover, ursolic acid's capacity to inhibit PTPs has been disputed based on ursolic acid's failure to delay insulin receptor de-phosphorylation in cultured cells (Jung S H, et al. (2007) The Biochemical journal 403(2):243-250), and ursolic acid's capacity to act as an insulin mimetic (Jung S H, et al. (2007) The Biochemical journal 403(2):243-250). In addition, global and muscle-specific PTP1B knockout mice do not possess increased muscle mass, although they are resistant to obesity and obesity-related disorders (Delibegovic M, et al. (2007) Molecular and cellular biology 27(21):7727-7734; Klaman L D, et al. (2000) Molecular and cellular biology 20(15):5479-5489). Furthermore, ursolic acid increases pancreatic beta cell mass and serum insulin levels in vivo, perhaps via its anti-inflammatory effects (Wang Z H, et al. (2010) European journal of pharmacology 628(1-3):255-260; Jayaprakasam B, et al. (2006) J Agric Food Chem 54(1):243-248; de Melo C L, et al. (2010) Chem Biol Interact 185(1):59-65). Importantly, inflammation is now recognized as a central pathogenic mechanism in muscle atrophy, metabolic syndrome, obesity, fatty liver disease and type 2 diabetes. Thus, the existing data suggest at least four mechanisms to explain ursolic acid's capacity to increase insulin signaling in vivo: PTP inhibition, direct stimulation of the insulin receptor, increased insulin production, and reduced inflammation. Of these four potential mechanisms, only the latter three have been demonstrated in vivo.

To compare the effects of ursolic acid and oleanolic acid on skeletal muscle and liver weight, C57BL/6 mice were administered ursolic acid (200 mg/kg), oleanolic acid (200 mg/kg), or vehicle alone (corn oil) via i.p. injection. Mice were then fasted, and after 12 hours of fasting, mice received a second dose of ursolic acid, oleanolic acid, or vehicle. After 24 hours of fasting, lower hindlimb skeletal muscles and liver were harvested and weighed. As shown previously, ursolic acid increased skeletal muscle weight (FIG. 14B), but not liver weight (FIG. 14C). In contrast, oleanolic acid increased liver weight (FIG. 14F), but not skeletal muscle weight (FIG. 14E). Interestingly, metformin (250 mg/kg) resembled oleanolic acid in biological effect: it increased liver weight (FIG. 141), but not muscle weight (FIG. 14H). Without wishing to be bound by a particular theory, ursolic acid increases skeletal muscle and inhibit muscle atrophy by a pathway that does not involve PTP inhibition.

13. Targeted Inhibition of PTP1B Does Not Induce Skeletal Muscle Hypertrophy.

To further rule out the potential role of PTP1B inhibition in skeletal muscle hypertrophy, PTP1B expression was specifically reduced in mouse skeletal muscle by transfecting plasmid DNA constructed to express RNA interference constructs. Briefly, C57BL/6 mouse tibialis anterior muscles were transfected with 20 μg pCMV-miR-control (control plasmid transfected in the left TA) or either 20 μg pCMV-miR-PTP1B #1 (encoding miR-PTP1B #1; transfected in the right TA) or 20 μg pCMV-miR-PTP1B #2 (encoding miR-PTP1B #2; transfected in the right TA). miR-PTP1B #1 and miR-PTP1B #2 encode two distinct RNA interference (RNAi) constructs targeting distinct regions of PTP1B mRNA. Tissue was harvested 10 days following transfection.

Of note with regard to FIG. 15A, mRNA measurements were taken from the entire TA muscle. Because electroporation transfects only a portion of muscle fibers, the data underestimate PTP1B knockdown in transfected muscle fibers. In FIG. 15A, mRNA levels in the right TA were normalized to levels in the left TA, which were set at 1; data are means±SEM from 3 mice. In FIG. 15B, in each TA muscle, the mean diameter of >300 transfected fibers was determined; data are means±SEM from 3 TA muscles per condition. For both FIGS. 15A and 15B, P-values were determined with one-tailed paired t-tests.

Although both miR-PTP1B constructs reduced PTP1B mRNA (FIG. 15A), neither increased skeletal muscle fiber diameter (FIG. 15B). These data demonstrate that targeted PTP1B inhibition does not cause muscle fiber hypertrophy. Without wishing to be bound by a particular theory, ursolic acid does not increase skeletal muscle by inhibiting PTP1B.

14. Ursolic Acid Serum Levels Associated with Increased Muscle Mass and Decreased Adiposity.

To determine the dose-response relationship between dietary ursolic acid and muscle and adipose weight, C57BL/6 mice were fed standard chow containing varying amounts of ursolic acid for 7 weeks. Serum ursolic acid levels from mice were determined as described above. As shown previously in FIG. 10A, ursolic acid increased skeletal muscle weight and decreased weight of retroperitoneal and epididymal fat pads in a dose-dependent manner, but did not alter heart weight (FIG. 16A; data are means±SEM). These effects of ursolic acid were discernable at 0.035% ursolic acid and were maximal at doses ≥0.14% ursolic acid. Serum was collected from these same mice at the time of necropsy, and then measured random serum ursolic acid levels via ultra high performance liquid chromatography (UPLC). The data indicate that ursolic acid serum levels in the range of 0.25-0.5 μg/ml are sufficient to increase muscle mass and decrease adiposity (FIG. 16B; data are means±SEM). Of note, 0.5 μg/ml equals 1.1 μM ursolic acid, close to the dose used in the Connectivity Map (8.8 μM) and in the C2C12 experiments (10 μM) described above.

The data described herein indicate that ursolic acid reduced muscle atrophy and stimulated muscle hypertrophy in mice. Importantly, ursolic acid's effects on muscle were accompanied by reductions in adiposity, fasting blood glucose and plasma leptin, cholesterol and triglycerides, as well as increases in the ratio of skeletal muscle to fat, the amount of brown fat, the ratio of brown fat to white fat, and increased energy expenditure. Without wishing to be bound by a particular theory, ursolic acid reduced muscle atrophy and stimulated muscle hypertrophy by enhancing skeletal muscle IGF-I expression and IGF-I signaling, and inhibiting atrophy-associated skeletal muscle mRNA expression.

All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention.

More specifically, certain agents which are both chemically and physiologically related can be substituted for the agents described herein while the same or similar results can be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

G. REFERENCES

The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.

1. Bodine S C, et al. (2001) Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol 3(11):1014-1019.

2. Sandri M, et al. (2004) Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117(3):399-412.

3. Stitt T N, et al. (2004) The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol Cell 14(3):395-403.

4. Hu Z, et al. (2009) Endogenous glucocorticoids and impaired insulin signaling are both required to stimulate muscle wasting under pathophysiological conditions in mice. The Journal of clinical investigation 119(10):3059-3069.

5. Dobrowolny G, et al. (2005) Muscle expression of a local Igf-1 isoform protects motor neurons in an ALS mouse model. The Journal of cell biology 168(2): 193-199.

6. Kandarian S C & Jackman R W (2006) Intracellular signaling during skeletal muscle atrophy. Muscle & nerve 33(2):155-165.

7. Hirose M, et al. (2001) Long-term denervation impairs insulin receptor substrate-1-mediated insulin signaling in skeletal muscle. Metabolism: clinical and experimental 50(2):216-222.

8. Pallafacchina G, et al. (2002) A protein kinase B-dependent and rapamycin-sensitive pathway controls skeletal muscle growth but not fiber type specification. Proceedings of the National Academy of Sciences of the United States of America 99(14):9213-9218.

9. Sandri M (2008) Signaling in muscle atrophy and hypertrophy. Physiology (Bethesda) 23:160-170.

10. Glass D J (2005) Skeletal muscle hypertrophy and atrophy signaling pathways. The international journal of biochemistry & cell biology 37(10):1974-1984.

11. Lecker S H, et al. (2004) Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. Faseb J 18(1):39-51.

12. Sacheck J M, et al. (2007) Rapid disuse and denervation atrophy involve transcriptional changes similar to those of muscle wasting during systemic diseases. Faseb J 21(1):140-155.

13. Jagoe R T, et al. (2002) Patterns of gene expression in atrophying skeletal muscles: response to food deprivation. Faseb J 16(13):1697-1712.

14. Sandri M, et al. (2006) PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proceedings of the National Academy of Sciences of the United States of America 103(44):16260-16265.

15. Wenz T, et al. (2009) Increased muscle PGC-1alpha expression protects from sarcopenia and metabolic disease during aging. Proceedings of the National Academy of Sciences of the United States of America 106(48):20405-20410.

16. Bodine S C, et al. (2001) Identification of ubiquitin ligases required for skeletal muscle atrophy. Science (New York, N.Y. 294(5547):1704-1708.

17. Lagirand-Cantaloube J, et al. (2008) The initiation factor eIF3-f is a major target for atroginl/MAFbx function in skeletal muscle atrophy. The EMBO journal 27(8): 1266-1276.

18. Cohen S, et al. (2009) During muscle atrophy, thick, but not thin, filament components are degraded by MuRF1-dependent ubiquitylation. The Journal of cell biology 185(6):1083-1095.

19. Adams V, et al. (2008) Induction of MuRF1 is essential for TNF-alpha-induced loss of muscle function in mice. Journal of molecular biology 384(1):48-59.

20. Leger B, et al. (2006) Human skeletal muscle atrophy in amyotrophic lateral sclerosis reveals a reduction in Akt and an increase in atrogin-1. Faseb J 20(3):583-585.

21. Doucet M, et al. (2007) Muscle atrophy and hypertrophy signaling in patients with chronic obstructive pulmonary disease. American journal of respiratory and critical care medicine 176(3):261-269.

22. Levine S, et al. (2008) Rapid disuse atrophy of diaphragm fibers in mechanically ventilated humans. The New England journal of medicine 358(13):1327-1335.

23. Adams C M, et al. (2011) Altered mRNA expression after long-term soleus electrical stimulation training in humans with paralysis. Muscle & nerve 43(1):65-75.

24. Ebert S M, et al. (2010) The transcription factor ATF4 promotes skeletal myofiber atrophy during fasting. Molecular endocrinology 24(4):790-799.

25. Lamb J, et al. (2006) The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science (New York, N.Y. 313 (5795): 1929-1935.

26. Frighetto R T S, et al. (2008) Isolation of ursolic acid from apple peels by high speed counter-current chromatography. Food Chemistry 106:767-771.

27. Liu J (1995) Pharmacology of oleanolic acid and ursolic acid. Journal of ethnopharmacology 49(2):57-68.

28. Liu J (2005) Oleanolic acid and ursolic acid: research perspectives. Journal of ethnopharmacology 100(1-2):92-94.

29. Wang Z H, et al. (2010) Anti-glycative effects of oleanolic acid and ursolic acid in kidney of diabetic mice. European journal of pharmacology 628(1-3):255-260.

30. Jang S M, et al. (2009) Ursolic acid enhances the cellular immune system and pancreatic beta-cell function in streptozotocin-induced diabetic mice fed a high-fat diet. Int Immunopharmacol 9(1): 113-119.

31. Jung S H, et al. (2007) Insulin-mimetic and insulin-sensitizing activities of a pentacyclic triterpenoid insulin receptor activator. The Biochemical journal 403(2):243-250.

32. Zhang W, et al. (2006) Ursolic acid and its derivative inhibit protein tyrosine phosphatase 1B, enhancing insulin receptor phosphorylation and stimulating glucose uptake. Biochimica et biophysica acta 1760(10):1505-1512.

33. Goldstein B J, et al. (2000) Tyrosine dephosphorylation and deactivation of insulin receptor substrate-1 by protein-tyrosine phosphatase 1B. Possible facilitation by the formation of a ternary complex with the Grb2 adaptor protein. The Journal of biological chemistry 275(6):4283-4289.

34. Delibegovic M, et al. (2007) Improved glucose homeostasis in mice with muscle-specific deletion of protein-tyrosine phosphatase 1B. Molecular and cellular biology 27(21): 7727-7734.

35. Zabolotny J M, et al. (2004) Transgenic overexpression of protein-tyrosine phosphatase 1B in muscle causes insulin resistance, but overexpression with leukocyte antigen-related phosphatase does not additively impair insulin action. The Journal of biological chemistry 279(23):24844-24851.

36. Sivakumar G, et al. (2009) Plant-based corosolic acid: future anti-diabetic drug? Biotechnol J 4(12):1704-1711.

37. Ebert S M, et al. (2010) The transcription factor ATF4 promotes skeletal myofiber atrophy during fasting. Molecular Endocrinology 24(4):790-799.

38. Dubowitz V, et al. (2007) Muscle biopsy: a practical approach (Saunders Elsevier, Philadelphia) 3rd Ed pp XIII, 611 s.

39. Hishiya A, et al. (2006) A novel ubiquitin-binding protein ZNF216 functioning in muscle atrophy. The EMBO journal 25(3):554-564.

40. Adams C M, et al. (2011) Altered mRNA expression after long-term soleus electrical stimulation training in humans with paralysis. Muscle Nerve. 43(1):65-75

41. Hameed M, et al. (2004) The effect of recombinant human growth hormone and resistance training on IGF-I mRNA expression in the muscles of elderly men. The Journal of physiology 555(Pt 1): 231-240.

42. Adams G R & Haddad F (1996) The relationships among IGF-1, DNA content, and protein accumulation during skeletal muscle hypertrophy. J Appl Physiol 81(6):2509-2516.

43. Gentile M A, et al. (2010) Androgen-mediated improvement of body composition and muscle function involves a novel early transcriptional program including IGF1, mechano growth factor, and induction of {beta}-catenin. Journal of molecular endocrinology 44(1):55-73.

44. Shavlakadze T, et al. (2005) Insulin-like growth factor I slows the rate of denervation induced skeletal muscle atrophy. Neuromuscul Disord 15(2):139-146.

45. Sacheck J M, et al. (2004) IGF-I stimulates muscle growth by suppressing protein breakdown and expression of atrophy-related ubiquitin ligases, atrogin-1 and MuRF1. Am J Physiol Endocrinol Metab 287(4):E591-601.

46. Frost R A, et al. (2009) Regulation of REDD1 by insulin-like growth factor-I in skeletal muscle and myotubes. J Cell Biochem 108(5):1192-1202.

47. Lee S J (2004) Regulation of muscle mass by myostatin. Annu Rev Cell Dev Biol 20:61-86.

48. Dupont J, et al. (2001) Insulin-like growth factor 1 (IGF-1)-induced twist expression is involved in the anti-apoptotic effects of the IGF-1 receptor. The Journal of biological chemistry 276(28):26699-26707.

49. Tureckova J, et al. (2001) Insulin-like growth factor-mediated muscle differentiation: collaboration between phosphatidylinositol 3-kinase-Akt-signaling pathways and myogenin. The Journal of biological chemistry 276(42):39264-39270.

50. Yakar S, et al. (1999) Normal growth and development in the absence of hepatic insulin-like growth factor I. Proceedings of the National Academy of Sciences of the United States of America 96(13):7324-7329.

51. Adams G R, et al. (1999) Time course of changes in markers of myogenesis in overloaded rat skeletal muscles. J Appl Physiol 87(5):1705-1712.

52. Lai K M, et al. (2004) Conditional activation of akt in adult skeletal muscle induces rapid hypertrophy. Molecular and cellular biology 24(21):9295-9304.

53. Izumiya Y, et al. (2008) Fast/Glycolytic muscle fiber growth reduces fat mass and improves metabolic parameters in obese mice. Cell metabolism 7(2):159-172.

54. Jayaprakasam B, et al. (2006) Amelioration of obesity and glucose intolerance in high-fat-fed C57BL/6 mice by anthocyanins and ursolic acid in Cornelian cherry (Cornus mas). J Agric Food Chem 54(1):243-248.

55. de Melo C L, et al. (2010) Oleanolic acid, a natural triterpenoid improves blood glucose tolerance in normal mice and ameliorates visceral obesity in mice fed a high-fat diet. Chem Biol Interact 185(1):59-65.

56. Qian S, et al. (2010) Synthesis and biological evaluation of oleanolic acid derivatives as inhibitors of protein tyrosine phosphatase 1B. J Nat Prod 73(11):1743-1750.

57. Zhang Y N, et al. (2008) Oleanolic acid and its derivatives: new inhibitor of protein tyrosine phosphatase 1B with cellular activities. Bioorg Med Chem 16(18):8697-8705.

58. Klaman L D, et al. (2000) Increased energy expenditure, decreased adiposity, and tissue-specific insulin sensitivity in protein-tyrosine phosphatase 1B-deficient mice. Molecular and cellular biology 20(15):5479-5489.

59. Reagan-Shaw S, Nihal M, & Ahmad N (2008) Dose translation from animal to human studies revisited. Faseb J 22(3):659-661.

60. Barton-Davis E R, et al. (1998) Viral mediated expression of insulin-like growth factor I blocks the aging-related loss of skeletal muscle function. Proceedings of the National Academy of Sciences of the United States of America 95(26):15603-15607.

61. Musarò A, et al. (2001) Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nature Genetics 27(2):195-200.

62. Zhou X, et al. (2010) Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell 142(4):531-43.

Claims

1. A method for: in an animal, the method comprising administering to the animal ursolic acid, or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein said animal is selected from the group consisting of a primate, domesticated fish, domesticated crustacean, domesticated mollusk, poultry, rabbit, dog, cat, and livestock.

(a) increasing skeletal muscle mass;
(b) reducing skeletal muscle atrophy;
(c) increasing muscular strength; or
(d) promoting muscle growth

2. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in an amount of greater than or equal to 100 mg per day.

3. The method of claim 1, wherein the animal is a mammal.

4. The method of claim 1, wherein the animal is a human.

5. The method of claim 1, wherein the animal is selected from the group consisting of a dog, cat, pig, cow, horse, goat, bison, sheep, chicken, turkey, duck, goose, and domesticated fish.

6. The method of claim 1, wherein the ursolic acid is present as a pharmaceutically acceptable salt selected from salts derived from aluminum, ammonium, calcium, copper (-ic and -ous), ferric, ferrous, lithium, magnesium, manganese, potassium, sodium, or zinc; salts of primary, secondary, and tertiary amines; and salts derived from arginine, betaine, caffeine, choline, N,N′-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholinc, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, or tromethamine.

7. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in an amount of greater than 200 mg per day.

8. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in an amount of greater than 300 mg per day.

9. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in an amount of greater than 400 mg per day.

10. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in an amount of greater than 500 mg per day.

11. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in an amount of greater than 750 mg per day.

12. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in an amount of greater than 1000 mg per day.

13. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in a composition that further comprises one or more adjuvants.

14. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in a dosage of from about 0.1 to about 50 mg/kg per day.

15. The method of claim 4, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in a dosage of from about 0.1 to about 50 mg/kg per day.

16. The method of claim 7, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in a dosage of from about 0.1 to about 50 mg/kg per day.

17. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in a composition that further comprises a preservative.

18. The method of claim 17, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof is administered to the animal in an amount of greater than or equal to 100 mg per day, and wherein the animal is selected from a domesticated fish, poultry, pig, cow, horse, goat, bison, sheep, dog, cat, and bison.

19. The method of claim 1, wherein the ursolic acid or pharmaceutically acceptable salt, hydrate, or solvate thereof has been isolated from a natural source.

20. The method of claim 1, for:

(b) reducing skeletal muscle atrophy;
(c) increasing muscular strength; or
(d) promoting muscle growth.
Patent History
Publication number: 20180118657
Type: Application
Filed: Nov 6, 2017
Publication Date: May 3, 2018
Applicants: UNIVERSITY OF IOWA RESEARCH FOUNDATION (IOWA CITY, IA), THE UNITED STATES OF AMERICA AS REPRESENTED BY THE SECRETARY OF THE DEPARTMENT OF VETERANS AFFAIRS (WASHINGTON, DC)
Inventors: Christopher M. ADAMS (Iowa City, IA), Steven D. KUNKEL (Iowa City, IA), Manish SUNEJA (Coralville, IA), Michael WELSH (Riverside, IA)
Application Number: 15/804,590
Classifications
International Classification: C07C 61/29 (20060101); C07D 265/30 (20060101); A61K 31/191 (20060101); A61K 31/198 (20060101); A61K 31/215 (20060101); A61K 31/19 (20060101); C07C 69/12 (20060101); C07C 69/757 (20060101); C07C 235/82 (20060101); C07J 53/00 (20060101);