Compositions for manipulating the lifespan and stress response of cells and organisms

Provided herein are methods and compositions for modulating the activity of sirtuin deacetylase protein family members; p53 activity; apoptosis; lifespan and sensitivity to stress of cells and organisms. Exemplary methods comprise contacting a cell with an activating compound, such as a flavone, stilbene, flavanone, isoflavone, catechin, chalcone, tannin or anthocyanidin; or an inhibitory compound, such as a sphingolipid, e.g., sphingosine.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

This patent application claims the benefit of priority from U.S. Provisional Patent Application No. 60/532,158, filed Dec. 23, 2003 and U.S. Provisional Patent Application No. 60/483,949, filed Jul. 1, 2003, each of which is herein incorporated by reference in its entirety.

BACKGROUND OF THE INVENTION

There is now good evidence from model organisms that the pace of aging can be regulated (Kenyon, C. Cell 105, 165-168 (2001)). Longevity regulatory genes have been identified in many eukaryotes, including rodents, flies, nematode worms and even single-celled organisms such as baker's yeast (reviewed in Sinclair, D. Mech Ageing Dev 123, 857-67 (2002); Hekimi, S. & Guarente, L. Genetics and the specificity of the aging process. Science 299, 1351-4 (2003)). These genes appear to be part of an evolutionarily conserved longevity pathway that evolved to promote survival in response to deteriorating environmental conditions (Kenyon, C. Cell 105, 165-168 (2001); Guarente, L. and Kenyon, C. Nature 408, 255-62. (2000). The yeast S. cerevisiae has proven a particularly useful model in which to study cell autonomous pathways of longevity (Sinclair, D. Mech Ageing Dev 123, 857-67 (2002)). In this organism, replicative lifespan is defined as the number of daughter cells an individual mother cell produces before dying. Yeast lifespan extension is governed by PNC1, a calorie restriction (CR)—and stress-responsive gene that depletes nicotinamide, a potent inhibitor of the longevity protein Sir2. Both PNC1 and SIR2 are required for lifespan extension by CR or mild stress (Lin et al. Science 289, 2126-8 (2000); Anderson et al. Nature 423, 181-5 (2003)) and additional copies of these genes extend lifespan 30-70% (Lin et al. Science 289, 2126-8 (2000); Anderson et al. Nature 423, 181-5 (2003); Kaeberlein et al. Genes Dev 13, 2570-80 (1999). Based on these results we proposed that CR may confer health benefits in a variety of species because it is a mild stress that induces a sirtuin-mediated organismal defense response (Anderson et al. Nature 423, 181-5 (2003).

Sir2, a histone deacetylase (HDAC), is the founding member of the sirtuin deacetylase family, which is characterized by a requirement for NAD+ as a co-substrate (Landry et al. Proc Natl Acad Sci USA 97, 5807-11 (2000); Imai et al. Nature 403, 795-800 (2000); Smith et al. Proc Natl Acad Sci USA 97, 6658-63 (2000); Tanner et al. Proc Natl Acad Sci USA 97, 14178-82 (2000); Tanny et al. Cell 99, 735-45 (1999); Tanny, J. C. and Moazed, D. Proc Natl Acad Sci USA 98, 415-20 (2001)). SIR2 was originally identified as a gene required for the formation of transcriptionally silent heterochromatin at yeast mating-type loci (Laurenson, P. and Rine, J. Microbiol Rev 56, 543-60. (1992)). Subsequent studies have shown that Sir2 suppresses recombination between repetitive DNA sequences at ribosomal RNA genes (rDNA)(Smith, J. S. and Boeke, J. D. Genes Dev 11, 241-54 (1997); Bryk et al. Genes Dev 11, 255-69 (1997); Gottlieb, S. and Esposito, R. E. Cell 56, 771-6 (1989)). Sir2 has also been implicated in the partitioning of carbonylated proteins to yeast mother cells during budding (Aguilaniu et al. Science (2003). Studies in C. elegans, mammalian cells, and the single-celled parasite Leishmania, indicate that the survival and longevity functions of sirtuins are conserved (Tissenbaum, H. A. and Guarente, L. Nature 410, 227-30 (2001); Vaziri et al. Cell 107, 149-59 (2001); Luo et al. Cell 107, 137-48 (2001); Vergnes et al. Gene 296, 139-50 (2002)). In C. elegans additional copies of sir-2.1 extend lifespan by 50% via the insulin/IGF-1 signalling pathway, the same pathway recently shown to regulate lifespan in rodents (Holzenberger et al. Nature 421, 182-7 (2003); Shimokawa et al. Faseb J 17, 1108-9 (2003); Tatar et al. Science 299, 1346-51 (2003)).

SUMMARY OF THE INVENTION

Provided herein are methods for activating a sirtuin deacetylase protein family member. The method may comprise contacting a sirtuin deacetylase protein family member with a compound having a structure selected from the group of formulas 1-25 and 31. Compounds falling within formulas 1-25 and 31 and activating a sirtuin protein are referred to herein as “activating compounds.” The activating compound may be a polyphenol compound, such as a plant polyphenol or an analog or derivative thereof. Exemplary compounds are selected from the group consisting of flavones, stilbenes, flavanones, isoflavones, catechins, chalcones, tannins and anthocyanidins or analog or derivative thereof. In illustrative embodiments, compounds are selected from the group of resveratrol, butein, piceatannol, isoliquiritgenin, fisetin, luteolin, 3,6,3′,4′-tetrahydroxyflavone, quercetin, and analogs and derivatives thereof. In certain embodiments, if the activating compound is a naturally occurring compound, it may not in a form in which it is naturally occurring.

The sirtuin deacetylase protein family member maybe the human SIRT1 protein or the yeast Sir2 protein.

The sirtuin deacetylase protein family member may be in a cell, in which case the method may comprise contacting the cell with an activating compound or introducing a compound into the cell. The cell may be in vitro. The cell may be a cell of a subject. The cell may be in a subject and the method may comprise administering the activating compound to the subject. Methods may further comprise determining the activity of the sirtuin deacetylase protein family member.

A cell may be contacted with an activating compound at a concentration of 0.1-100 μM. In certain embodiments, a cell is further contacted with an additional activating compound. In other embodiments, a cell is contacted with a least three different activating compounds.

Other methods encompassed herein include methods for inhibiting the activity of p53 in a cell and optionally protecting the cell against apoptosis, e.g., comprising contacting the cell with an activating compound at a concentration of less than about 0.5 μM. Another method comprises stimulating the activity of p53 in a cell and optionally inducing apoptosis in the cell, comprising contacting the cell with an activating compound at a concentration of at least 50 μM.

Also provided herein is a method for extending the lifespan of a eukaryotic cell, such as by increasing its resistance to stress, comprising contacting the cell with a compound selected from the group consisting of stilbene, flavone and chalcone family members. Such compounds are referred to as “lifespan extending compounds.” The compound may have the structure set forth in formula 7. Other compounds may be activating compounds having a structure set forth in any of formulas 1-25 and 30, provided they extend lifespan or increase resistance to stress. The compound may be selected from the group consisting of resveratrol, butein and fisetin and analogs and derivatives thereof. In certain embodiments, if the lifespan extending compound is a naturally occurring compound, it is not in a form in which it is naturally occurring. The method may further comprise determining the lifespan of the cell. The method may also further comprise contacting the cell with an additional compound or with at least three compounds selected from the group consisting of stilbene, flavone and chalcone family members or other lifespan extending compound. The cell may be contacted with a compound at a concentration of less than about 10 μM or at a concentration of about 10-100 μM. The cell may be in vitro or in vivo, it may be a yeast cell or a mammalian cell. If the cell is in a subject, the method may comprise administering the compound to the subject.

Methods for inhibiting sirtuins; inhibiting deacetylation of p53; stimulating apoptosis; shorting lifespan and rendering cells and organisms sensitive to stress are also encompassed. One method comprises contacting a sirtuin or cell or organism comprising such with an inhibitory compound having a formula selected from the group of formulas 26-29 and 31.

Also provided herein are compositions comprising, e.g., two compounds each having a formula selected from the group of formulas 1-31. Further provided herein are screening methods for identifying compounds, e.g., small molecules, that modulate sirtuins and/or modulate the life span or resistance to stress of cells. Methods may comprise (i) contacting a cell comprising a SIRT1 protein with a peptide of p53 comprising an acetylated residue 382 in the presence of an inhibitor of class I and class II HDAC under conditions appropriate for SIRT1 to deacetylate the peptide and (ii) determining the level of acetylation of the peptide, wherein a different level of acetylation of the peptide in the presence of the test compound relative to the absence of the test compound indicates that the test compound modulates SIRT1 in vivo.

BRIEF DESCRIPTION OF THE DRAWINGS

FIGS. 1a through 1d show the effects of resveratrol on the kinetics of recombinant human SIRT1. FIG. 1a shows resveratrol dose-response of SIRT1 catalytic rate at 25 μM NAD+, 25 μM p53-382 acetylated peptide. Relative initial rates are the mean of two determinations, each derived from the slopes of fluorescence (arbitrary fluorescence units, AFU) vs. time plots with data obtained at 0, 5, 10 and 20 min. of deacetylation. FIG. 1b shows the SIRT1 initial rate at 3 mM NAD+, as a function of p53-382 acetylated peptide concentration in the presence (Δ) or absence (▪) of 100 μM resveratrol. Lines represent non-linear least-squares fits to the Michaelis-Menten equation. Kinetic constants: Km(control, ▪)=64 μM, Km(+resveratrol, Δ)=1.8 μM; Vmax(control, ▪)=1107 AFU/min., Vmax(+resveratrol, Δ)=926 AFU/min. FIG. 1c shows the SIRT1 initial rate at 1 mM p53-382 acetylated peptide, as a function of NAD+ concentration, in the presence (Δ) or absence (▪) of 100 μM resveratrol. Lines represent non-linear least-squares fits to the Michaelis-Menten equation. Kinetic constants: Km(control, ▪)=558 μM, Km(+resveratrol, Δ)=101 μM; Vmax(control, ▪)=1863 AFU/min., Vmax(+resveratrol, Δ)=1749 AFU/min. FIG. 1d shows effects of resveratrol on nicotinamide inhibition of SIRT1. Kinetic constants are shown relative to those of the control (no nicotinamide, no resveratrol) and represent the mean of two determinations. Error bars are standard errors of the mean. The variable substrate in each experiment (N=NAD+, P=p53 acetylated peptide), the presence/absence of nicotinamide (±) and the resveratrol concentration (μM) are indicated beneath each pair of Km−Vmax bars.

FIG. 2a through 2d show the effects of polyphenols on Sir2 and S. cerevisiae lifespan. FIG. 2a shows the initial deacetylation rate of recombinant GST-Sir2 as a function of resveratrol concentration. Rates were determined at the indicated resveratrol concentrations, either with 100 μM ‘Fluor de Lys’ acetylated lysine substrate (FdL) plus 3 mM NAD+ (Δ) or with 200 μM p53-382 acetylated peptide substrate plus 200 μM NAD+ (▪). FIG. 2b shows lifespan analyses determined by micro-manipulating individual yeast cells as described Sinclair, D. A. and Guarente (Cell 91, 1033-42 (1997)) in complete 2% glucose medium with 10 μM of each compound, unless otherwise stated. Average lifespan was determined for wild type untreated (□), quercetin (◯) and piceatannol (●). FIG. 2c shows the average lifespan for wild type untreated (□), fisetin (◯), butein (), or resveratrol (Δ). FIG. 2d shows average lifespan for wild type untreated (□), and growth with resveratrol at 10 μM (Δ), 100 μM (●), or 500 μM (◯).

FIGS. 3a through 3f show resveratrol extending lifespan by mimicking CR and suppressing rDNA recombination. Yeast lifespans were determined as in FIG. 2. FIG. 3a shows average lifespan for wild type (wt) untreated (□), wild type+resveratrol (wt+R; ●) and glucose-restricted+resveratrol (CR+R; ◯). FIG. 3b shows average lifespans for wild type (□), sir2(Δ) sir2+resveratrol (sir2+R; ▴), pnc1 (◯), and pnc1+resveratrol (pnc1+R; ●). FIG. 3c shows resveratrol suppressing the frequency of ribosomal DNA recombination in the presence and absence of nicotinamide (NAM). Frequencies were determined by loss of the ADE2 marker gene from the rDNA locus (RDN1). FIG. 3d shows that resveratrol does not suppress rDNA recombination in a sir2 strain. FIG. 3e show that resveratrol and other sirtuin activators do not significantly increase rDNA silencing compared to a 2×SIR2 strain. Pre-treated cells (RDN1::URA3) were harvested and spotted as 10-fold serial dilutions on either SC or SC with 5-fluororotic acid (5-FOA). In this assay, increased rDNA silencing results in increased survival on 5-FOA medium. FIG. 3f show quantitation of the effect of resveratrol on rDNA silencing by counting numbers of surviving cells on FOA/total plated.

FIGS. 4a through 4e show resveratrol and other polyphenols stimulating SIRT1 activity in human cells. FIG. 4a shows a method for assaying intracellular deacetylase activity with a fluorogenic, cell-permeable substrate, FdL (‘Fluor de Lys’, BIOMOL). FdL (200 μM) is added to growth media and cells are incubated for 1-3 hours to allow FdL to enter the cells and the lysine-deacetylated product (deAc-FdL) to accumulate intracellularly. Cells are lysed with detergent in the presence of 1 μM TSA and 1 mM nicotinamide. Addition of the non-cell-permeable Developer (BIOMOL) releases a fluorophor, specifically from deAc-FdL. FIG. 4b shows SIRT1 activating polyphenols stimulating TSA-insensitive, FdL deacetylation by HeLa S3 cells. Cells were grown adherently in DMEM/10% FCS and treated for 1 hour with 200 μM FdL, 1 μM TSA and either vehicle (0.5% final DMSO, Control) or 500 μM of the indicated compound. Intracellular accumulation of deAc-FdL was then determined as described briefly in FIG. 4a. The intracellular deAc-FdL level for each compound (mean of six replicates) are plotted against the ratios to the control rate obtained in the in vitro SIRT1 polyphenol screen (see Table 1, Supplementary Tables 1 and 3). FIG. 4c shows U2OS osteosarcoma cells grown to ≧90% confluence in DMEM/10% FCS exposed to 0 or 10 grays of gamma irradiation (IR). Whole cell lysates were prepared 4 hours post-irradiation and were probed by Western blotting with indicated antibodies. FIG. 4d shows U2OS cells cultured as above and pre-treated with the indicated amounts of resveratrol or a 0.5% DMSO blank for 4 hours after which cells were exposed to 0 or 50 J/cm2 of UV radiation. Lysates were prepared and analyzed by Western blot as in FIG. 4c. FIG. 4e shows human embryonic kidney cells (HEK 293) expressing wild type SIRT1 or dominant negative SIRT1-H363Y (SIRT1-HY) protein cultured as described above, pre-treated with the indicated amounts of resveratrol or a 0.5% DMSO blank for 4 hours, and exposed to 50 J/cm2 of UV radiation as above. Lysates were prepared and analyzed as above.

FIG. 5 shows the deacetylation site preferences of recombinant SIRT1. Initial rates of deacetylation were determined for a series of fluorogenic acetylated peptide substrates based on short stretches of human histone H3, H4 and p53 sequence. Substrates examined include: H3-4-9 with the sequence K(Ac)QTARK(Ac) (SEQ ID NO:1); H3-9-14 with the sequence K(Ac)STGGK(Ac) (SEQ ID NO:2); H3-9-14/pS with the sequence K(Ac)—S(PO3)-TGGK(Ac) (SEQ ID NO:3); H3-14-18 with the sequence K(Ac)APRK(Ac) (SEQ ID NO:4); H4-1-5 with the sequence SGRGK(Ac)(SEQ ID NO:5); H4-12-16(Fluor de Lys-H4-AcK16) with the sequence KGGAK(Ac) (SEQ ID NO:6); H4-12-16/diAc with the sequence K(Ac)GGAK(Ac)(SEQ ID NO:7); p53-320 (Fluor de Lys-SIRT2) with the sequence QPKK(Ac)(SEQ ID NO:8); p53-373 with the sequence K(Ac)SKK(Ac)(SEQ ID NO:9); p53-382(Fluor de Lys-SIRT1 with the sequence RHKK(Ac) (SEQ ID NO:10); p53-382/di-Ac (Fluor de Lys-HDAC8) with the sequence RHK(Ac)K(Ac)(SEQ ID NO:11); and ε-acetyl lysine (Fluor de Lys, Fdl) wit the sequence K(Ac). All substrate were obtained from BIOMOL, Plymouth Meeting, Pa.). Recombinant human SIRT1 (1 μg, BIOMOL), was incubated for 10 minutes at 37° C. with 25 μM of the indicated fluorogenic acetylated peptide substrate and 500 μM NAD+. Reactions were stopped by the addition of 1 mM nicotinamide and the deacetylation-dependent fluorescent signal was determined.

FIG. 6a through 6c show intracellular deacetylation activity measured with a cell-permeable, fluorogenic HDAC and sirtuin substrate. HeLa S3 cells were grown to confluence in DMEM/10% FCS and then incubated with fresh medium containing 200 μM FdL for the indicated times at 37° C. Intracellular and medium levels of deacetylated substrate (deAc-FdL) were determined according to the manufacturer's instructions (HDAC assay kit, BIOMOL). All data points represent the mean of two determinations. FIG. 6a shows the concentration ratio of intracellular ([deAc-FdL]i) to medium ([deAc-FdL]o) concentrations in the presence (Δ) or absence (▪) of 1 μM trichostatin A (TSA). FIG. 6b shows total accumulation of deacetylated substrate (deAc-FdL) in the presence (Δ) or absence (▪) of 1 μM TSA. FIG. 6c shows intracellular accumulation of deacetylated substrate (deAc-FdL) in the presence (Δ) or absence (▪) of 1 μM TSA.

DETAILED DESCRIPTION OF THE INVENTION

Definitions

As used herein, the following terms and phrases shall have the meanings set forth below. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art.

The singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise. “Activating a sirtuin protein” refers to the action of producing an activated sirtuin protein, i.e., a sirtuin protein that is capable of performing at least one of its biological activities to at least some extent, e.g., with an increase of activity of at least about 10%, 50%, 2 fold or more. Biological activities of sirtuin proteins include deacetylation, e.g., of histones and p53; extending lifespan; increasing genomic stability; silencing transcription; and controlling the segregation of oxidized proteins between mother and daughter cells.

An “activating compound” refers to a compound that activates a sirtuin protein. Activating compounds may have a formula selected from the group of formulas 1-25 and 30.

A “form that is naturally occurring” when referring to a compound means a compound that is in a form, e.g., a composition, in which it can be found naturally. For example, since resveratrol can be found in red wine, it is present in red wine in a form that is naturally occurring. A compound is not in a form that is naturally occurring if, e.g., the compound has been purified and separated from at least some of the other molecules that are found with the compound in nature.

“Inhibiting a sirtuin protein” refers to the action of reducing at least one of the biological activities of a sirtuin protein to at least some extent, e.g., at least about 10%, 50%, 2 fold or more.

An “inhibitory compound” or “inhibiting compound” refers to a compound that inhibits a sirtuin protein. Inhibitory compounds may have a formula selected from the group of formulas 26-29 and 31.

A “naturally occurring compound” refers to a compound that can be found in nature, i.e., a compound that has not been designed by man. A naturally occurring compound may have been made by man or by nature.

“Replicative lifespan” which is used interchangeably herein with “lifespan” of a cell refers to the number of daughter cells produced by an individual “mother cell.” “Chronological aging,” on the other hand, refers to the length of time a population of non-dividing cells remains viable when deprived of nutrients. “Increasing the lifespan of a cell” or “extending the lifespan of a cell,” as applied to cells or organisms, refers to increasing the number of daughter cells produced by one cell; increasing the ability of cells or organisms to cope with stresses and combat damage, e.g., to DNA, proteins; and/or increasing the ability of cells or organisms to survive and exist in a living state for longer under a particular condition, e.g., stress. Lifespan can be increased by at least about 20%, 30%, 40%, 50%, 60% or between 20% and 70%, 30% and 60%, 40% and 60% or more using methods described herein.

“Sirtuin deacetylase protein family members;” “Sir2 family members;” “Sir2 protein family members;” or “sirtuin proteins” includes yeast Sir2, Sir-2.1, and human SIRT1 and SIRT2 proteins. Other family members include the four additional yeast Sir2-like genes termed “HST genes” (homologues of Sir two) HST1, HST2, HST3 and HST4, and the five other human homologues hSIRT3, hSIRT4, hSIRT5, hSIRT6 and hSIRT7 (Brachmann et al. (1995) Genes Dev. 9:2888 and Frye et al. (1999) BBRC 260:273). Preferred sirtuins are those that share more similarities with SIRT1, i.e., hSIRT1, and/or Sir2 than with SIRT2, such as those members having at least part of the N-terminal sequence present in SIRT1 and absent in SIRT2 such as SIRT3 has.

The terms “comprise” and “comprising” are used in the inclusive, open sense, meaning that additional elements may be included.

The term “including”is used to mean “including but not limited to”. “Including” and “including but not limited to” are used interchangeably.

The term “cis” is art-recognized and refers to the arrangement of two atoms or groups around a double bond such that the atoms or groups are on the same side of the double bond. Cis configurations are often labeled as (Z) configurations.

The term “trans” is art-recognized and refers to the arrangement of two atoms or groups around a double bond such that the atoms or groups are on the opposite sides of a double bond. Trans configurations are often labeled as (E) configurations.

The term “covalent bond” is art-recognized and refers to a bond between two atoms where electrons are attracted electrostatically to both nuclei of the two atoms, and the net effect of increased electron density between the nuclei counterbalances the internuclear repulsion. The term covalent bond includes coordinate bonds when the bond is with a metal ion.

The term “therapeutic agent” is art-recognized and refers to any chemical moiety that is a biologically, physiologically, or pharmacologically active substance that acts locally or systemically in a subject. Examples of therapeutic agents, also referred to as “drugs”, are described in well-known literature references such as the Merck Index, the Physicians Desk Reference, and The Pharmacological Basis of Therapeutics, and they include, without limitation, medicaments; vitamins; mineral supplements; substances used for the treatment, prevention, diagnosis, cure or mitigation of a disease or illness; substances which affect the structure or function of the body; or pro-drugs, which become biologically active or more active after they have been placed in a physiological environment.

The term “therapeutic effect” is art-recognized and refers to a local or systemic effect in animals, particularly mammals, and more particularly humans caused by a pharmacologically active substance. The term thus means any substance intended for use in the diagnosis, cure, mitigation, treatment or prevention of disease or in the enhancement of desirable physical or mental development and/or conditions in an animal or human. The phrase “therapeutically-effective amount” means that amount of such a substance that produces some desired local or systemic effect at a reasonable benefit/risk ratio applicable to any treatment. The therapeutically effective amount of such substance will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. For example, certain compositions described herein may be administered in a sufficient amount to produce a at a reasonable benefit/risk ratio applicable to such treatment.

The term “synthetic” is art-recognized and refers to production by in vitro chemical or enzymatic synthesis.

The term “meso compound” is art-recognized and refers to a chemical compound which has at least two chiral centers but is achiral due to a plane or point of symmetry.

The term “chiral” is art-recognized and refers to molecules that have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules that are superimposable on their mirror image partner. A “prochiral molecule” is a molecule that has the potential to be converted to a chiral molecule in a particular process.

The term “stereoisomers” is art-recognized and refers to compounds that have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. In particular, “enantiomers” refer to two stereoisomers of a compound that are non-superimposable mirror images of one another. “Diastereomers”, on the other hand, refers to stereoisomers with two or more centers of dissymmetry and whose molecules are not mirror images of one another.

Furthermore, a “stereoselective process” is one that produces a particular stereoisomer of a reaction product in preference to other possible stereoisomers of that product. An “enantioselective process” is one that favors production of one of the two possible enantiomers of a reaction product.

The term “regioisomers” is art-recognized and refers to compounds that have the same molecular formula but differ in the connectivity of the atoms. Accordingly, a “regioselective process” is one that favors the production of a particular regioisomer over others, e.g., the reaction produces a statistically significant increase in the yield of a certain regioisomer.

The term “epimers” is art-recognized and refers to molecules with identical chemical constitution and containing more than one stereocenter, but which differ in configuration at only one of these stereocenters.

The term “ED50” is art-recognized. In certain embodiments, ED50 means the dose of a drug that produces 50% of its maximum response or effect, or alternatively, the dose that produces a pre-determined response in 50% of test subjects or preparations. The term “LD50” is art-recognized. In certain embodiments, LD50 means the dose of a drug that is lethal in 50% of test subjects. The term “therapeutic index” is an art-recognized term that refers to the therapeutic index of a drug, defined as LD50/ED50.

The term “structure-activity relationship” or “(SAR)” is art-recognized and refers to the way in which altering the molecular structure of a drug or other compound alters its biological activity, e.g., its interaction with a receptor, enzyme, nucleic acid or other target and the like.

The term “aliphatic” is art-recognized and refers to a linear, branched, cyclic alkane, alkene, or alkyne. In certain embodiments, aliphatic groups in the present compounds are linear or branched and have from 1 to about 20 carbon atoms.

The term “alkyl” is art-recognized, and includes saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. In certain embodiments, a straight chain or branched chain alkyl has about 30 or fewer carbon atoms in its backbone (e.g., C1-C30 for straight chain, C3-C30 for branched chain), and alternatively, about 20 or fewer. Likewise, cycloalkyls have from about 3 to about 10 carbon atoms in their ring structure, and alternatively about 5, 6 or 7 carbons in the ring structure. The term “alkyl” is also defined to include halosubstituted alkyls.

Moreover, the term “alkyl” (or “lower alkyl”) includes “substituted alkyls”, which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents may include, for example, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain may themselves be substituted, if appropriate. For instance, the substituents of a substituted alkyl may include substituted and unsubstituted forms of amino, azido, imino, amido, phosphoryl (including phosphonate and phosphinate), sulfonyl (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alkylthios, carbonyls (including ketones, aldehydes, carboxylates, and esters), —CN and the like. Exemplary substituted alkyls are described below. Cycloalkyls may be further substituted with alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-substituted alkyls, —CN, and the like.

The term “aralkyl” is art-recognized and refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).

The terms “alkenyl” and “alkynyl” are art-recognized and refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.

Unless the number of carbons is otherwise specified, “lower alkyl” refers to an alkyl group, as defined above, but having from one to about ten carbons, alternatively from one to about six carbon atoms in its backbone structure. Likewise, “lower alkenyl” and “lower alkynyl” have similar chain lengths.

The term “heteroatom” is art-recognized and refers to an atom of any element other than carbon or hydrogen. Illustrative heteroatoms include boron, nitrogen, oxygen, phosphorus, sulfur and selenium.

The term “aryl” is art-recognized and refers to 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as “aryl heterocycles” or “heteroaromatics.” The aromatic ring may be substituted at one or more ring positions with such substituents as described above, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, —CF3, —CN, or the like. The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is aromatic, e.g., the other cyclic rings may be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls.

The terms ortho, meta and para are art-recognized and refer to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively. For example, the names 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.

The terms “heterocyclyl” or “heterocyclic group” are art-recognized and refer to 3- to about 10-membered ring structures, alternatively 3- to about 7-membered rings, whose ring structures include one to four heteroatoms. Heterocycles may also be polycycles. Heterocyclyl groups include, for example, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxanthene, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, oxolane, thiolane, oxazole, piperidine, piperazine, morpholine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like. The heterocyclic ring may be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, —CF3, —CN, or the like.

The terms “polycyclyl” or “polycyclic group” are art-recognized and refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are “fused rings”. Rings that are joined through non-adjacent atoms are termed “bridged” rings. Each of the rings of the polycycle may be substituted with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, —CF3, —CN, or the like.

The term “carbocycle” is art-recognized and refers to an aromatic or non-aromatic ring in which each atom of the ring is carbon.

The term “nitro” is art-recognized and refers to —NO2; the term “halogen” is art-recognized and refers to —F, —Cl, —Br or —I; the term “sulfhydryl” is art-recognized and refers to —SH; the term “hydroxyl” means —OH; and the term “sulfonyl” is art-recognized and refers to —SO2. “Halide” designates the corresponding anion of the halogens, and “pseudohalide” has the definition set forth on page 560 of “Advanced Inorganic Chemistry” by Cotton and Wilkinson.

The terms “amine” and “amino” are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that may be represented by the general formulas:
wherein R50, R51 and R52 each independently represent a hydrogen, an alkyl, an alkenyl, —(CH2)m—R61, or R50 and R51, taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R61 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle or a polycycle; and m is zero or an integer in the range of 1 to 8. In certain embodiments, only one of R50 or R51 may be a carbonyl, e.g., R50, R51 and the nitrogen together do not form an imide. In other embodiments, R50 and R51 (and optionally R52) each independently represent a hydrogen, an alkyl, an alkenyl, or —(CH2)m—R61. Thus, the term “alkylamine” includes an amine group, as defined above, having a substituted or unsubstituted alkyl attached thereto, i.e., at least one of R50 and R51 is an alkyl group.

The term “acylamino” is art-recognized and refers to a moiety that may be represented by the general formula:
wherein R50 is as defined above, and R54 represents a hydrogen, an alkyl, an alkenyl or —(CH2)m—R61, where m and R61 are as defined above.

The term “amido” is art recognized as an amino-substituted carbonyl and includes a moiety that may be represented by the general formula:
wherein R50 and R51 are as defined above. Certain embodiments of amides may not include imides which may be unstable.

The term “alkylthio” refers to an alkyl group, as defined above, having a sulfur radical attached thereto. In certain embodiments, the “alkylthio” moiety is represented by one of —S-alkyl, —S-alkenyl, —S-alkynyl, and —S—(CH2)m—R61, wherein m and R61 are defined above. Representative alkylthio groups include methylthio, ethyl thio, and the like.

The term “carbonyl” is art recognized and includes such moieties as may be represented by the general formulas:
wherein X50 is a bond or represents an oxygen or a sulfur, and R55 and R56 represents a hydrogen, an alkyl, an alkenyl, —(CH2)m—R61 or a pharmaceutically acceptable salt, R56 represents a hydrogen, an alkyl, an alkenyl or —(CH2)m—R61, where m and R61 are defined above. Where X50 is an oxygen and R55 or R56 is not hydrogen, the formula represents an “ester”. Where X50 is an oxygen, and R55 is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when R55 is a hydrogen, the formula represents a “carboxylic acid”. Where X50 is an oxygen, and R56 is hydrogen, the formula represents a “formate”. In general, where the oxygen atom of the above formula is replaced by sulfur, the formula represents a “thiolcarbonyl” group. Where X50 is a sulfur and R55 or R56 is not hydrogen, the formula represents a “thiolester.” Where X50 is a sulfur and R55 is hydrogen, the formula represents a “thiolcarboxylic acid.” Where X50 is a sulfur and R56 is hydrogen, the formula represents a “thiolformate.” On the other hand, where X50 is a bond, and R55 is not hydrogen, the above formula represents a “ketone” group. Where X50 is a bond, and R55 is hydrogen, the above formula represents an “aldehyde” group.

The terms “alkoxyl” or “alkoxy” are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like. An “ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as may be represented by one of —O-alkyl, —O-alkenyl, —O-alkynyl, —O—(CH2)m—R61, where m and R61 are described above.

The term “sulfonate” is art recognized and refers to a moiety that may be represented by the general formula:
in which R57 is an electron pair, hydrogen, alkyl, cycloalkyl, or aryl.

The term “sulfate” is art recognized and includes a moiety that may be represented by the general formula:
in which R57 is as defined above.

The term “sulfonamido” is art recognized and includes a moiety that may be represented by the general formula:
in which R50 and R56 are as defined above.

The term “sulfamoyl” is art-recognized and refers to a moiety that may be represented by the general formula:
in which R50 and R51 are as defined above.

The term “sulfonyl” is art-recognized and refers to a moiety that may be represented by the general formula:
in which R58 is one of the following: hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl.

The term “sulfoxido” is art-recognized and refers to a moiety that may be represented by the general formula:
in which R58 is defined above.

The term “phosphoryl” is art-recognized and may in general be represented by the formula:
wherein Q50 represents S or O, and R59 represents hydrogen, a lower alkyl or an aryl. When used to substitute, e.g., an alkyl, the phosphoryl group of the phosphorylalkyl may be represented by the general formulas:
wherein Q50 and R59, each independently, are defined above, and Q51 represents O, S or N. When Q50 is S, the phosphoryl moiety is a “phosphorothioate”.

The term “phosphoramidite” is art-recognized and may be represented in the general formulas:
wherein Q51, R50, R51 and R59 are as defined above.

The term “phosphonamidite” is art-recognized and may be represented in the general formulas:
wherein Q51, R50, R51 and R59 are as defined above, and R60 represents a lower alkyl or an aryl.

Analogous substitutions may be made to alkenyl and alkynyl groups to produce, for example, aminoalkenyls, aminoalkynyls, amidoalkenyls, amidoalkynyls, iminoalkenyls, iminoalkynyls, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls.

The definition of each expression, e.g. alkyl, m, n, and the like, when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.

The term “selenoalkyl” is art-recognized and refers to an alkyl group having a substituted seleno group attached thereto. Exemplary “selenoethers” which may be substituted on the alkyl are selected from one of —Se-alkyl, —Se-alkenyl, —Se-alkynyl, and —Se—(CH2)m—R61, m and R61 being defined above.

The terms triflyl, tosyl, mesyl, and nonaflyl are art-recognized and refer to trifluoromethanesulfonyl, p-toluenesulfonyl, methanesulfonyl, and nonafluorobutanesulfonyl groups, respectively. The terms triflate, tosylate, mesylate, and nonaflate are art-recognized and refer to trifluoromethanesulfonate ester, p-toluenesulfonate ester, methanesulfonate ester, and nonafluorobutanesulfonate ester functional groups and molecules that contain said groups, respectively.

The abbreviations Me, Et, Ph, Tf, Nf, Ts, and Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, nonafluorobutanesulfonyl, p-toluenesulfonyl and methanesulfonyl, respectively. A more comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears in the first issue of each volume of the Journal of Organic Chemistry; this list is typically presented in a table entitled Standard List of Abbreviations.

Certain compounds contained in compositions described herein may exist in particular geometric or stereoisomeric forms. In addition, compounds may also be optically active. Contemplated herein are all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are encompassed herein.

If, for instance, a particular enantiomer of a compound is desired, it may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers. Alternatively, where the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.

It will be understood that “substitution” or “substituted with” includes 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., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, or other reaction.

The term “substituted” is also 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, aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described herein above. The permissible substituents may be one or more and the same or different for appropriate organic compounds. Heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Compounds are not intended to be limited in any manner by the permissible substituents of organic compounds.

The chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 67th Ed., 1986-87, inside cover. Also, the term “hydrocarbon” is contemplated to include all permissible compounds having at least one hydrogen and one carbon atom. In a broad aspect, the permissible hydrocarbons include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic organic compounds that may be substituted or unsubstituted.

The term “protecting group” is art-recognized and refers to temporary substituents that protect a potentially reactive functional group from undesired chemical transformations. Examples of such protecting groups include esters of carboxylic acids, silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones, respectively. The field of protecting group chemistry has been reviewed by Greene and Wuts in Protective Groups in Organic Synthesis (2nd ed., Wiley: N.Y., 1991).

The term “hydroxyl-protecting group” is art-recognized and refers to those groups intended to protect a hydrozyl group against undesirable reactions during synthetic procedures and includes, for example, benzyl or other suitable esters or ethers groups known in the art.

The term “carboxyl-protecting group” is art-recognized and refers to those groups intended to protect a carboxylic acid group, such as the C-terminus of an amino acid or peptide or an acidic or hydroxyl azepine ring substituent, against undesirable reactions during synthetic procedures and includes. Examples for protecting groups for carboxyl groups involve, for example, benzyl ester, cyclohexyl ester, 4-nitrobenzyl ester, t-butyl ester, 4-pyridylmethyl ester, and the like.

The term “amino-blocking group” is art-recognized and refers to a group which will prevent an amino group from participating in a reaction carried out on some other functional group, but which can be removed from the amine when desired. Such groups are discussed by in Ch. 7 of Greene and Wuts, cited above, and by Barton, Protective Groups in Organic Chemistry ch. 2 (McOmie, ed., Plenum Press, New York, 1973). Examples of suitable groups include acyl protecting groups such as, to illustrate, formyl, dansyl, acetyl, benzoyl, trifluoroacetyl, succinyl, methoxysuccinyl, benzyl and substituted benzyl such as 3,4-dimethoxybenzyl, o-nitrobenzyl, and triphenylmethyl; those of the formula —COOR where R includes such groups as methyl, ethyl, propyl, isopropyl, 2,2,2-trichloroethyl, 1-methyl-1-phenylethyl, isobutyl, t-butyl, t-amyl, vinyl, allyl, phenyl, benzyl, p-nitrobenzyl, o-nitrobenzyl, and 2,4-dichlorobenzyl; acyl groups and substituted acyl such as formyl, acetyl, chloroacetyl, dichloroacetyl, trichloroacetyl, trifluoroacetyl, benzoyl, and p-methoxybenzoyl; and other groups such as methanesulfonyl, p-toluenesulfonyl, p-bromobenzenesulfonyl, p-nitrophenylethyl, and p-toluenesulfonyl-aminocarbonyl. Preferred amino-blocking groups are benzyl (—CH2C6H5), acyl [C(O)R1] or SiR13 where R1 is C1-C4 alkyl, halomethyl, or 2-halo-substituted-(C2-C4 alkoxy), aromatic urethane protecting groups as, for example, carbonylbenzyloxy (Cbz); and aliphatic urethane protecting groups such as t-butyloxycarbonyl (Boc) or 9-fluorenylmethoxycarbonyl (FMOC).

The definition of each expression, e.g. lower alkyl, m, n, p and the like, when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.

The term “electron-withdrawing group” is art-recognized, and refers to the tendency of a substituent to attract valence electrons from neighboring atoms, i.e., the substituent is electronegative with respect to neighboring atoms. A quantification of the level of electron-withdrawing capability is given by the Hammett sigma (σ) constant. This well known constant is described in many references, for instance, March, Advanced Organic Chemistry 251-59 (McGraw Hill Book Company: New York, 1977). The Hammett constant values are generally negative for electron donating groups (σ(P)=−0.66 for NH2) and positive for electron withdrawing groups (σ(P)=0.78 for a nitro group), σ(P) indicating para substitution. Exemplary electron-withdrawing groups include nitro, acyl, formyl, sulfonyl, trifluoromethyl, cyano, chloride, and the like. Exemplary electron-donating groups include amino, methoxy, and the like.

The term “small molecule” is art-recognized and refers to a composition which has a molecular weight of less than about 2000 amu, or less than about 1000 amu, and even less than about 500 amu. Small molecules may be, for example, nucleic acids, peptides, polypeptides, peptide nucleic acids, peptidomimetics, carbohydrates, lipids or other organic (carbon containing) or inorganic molecules. Many pharmaceutical companies have extensive libraries of chemical and/or biological mixtures, often fungal, bacterial, or algal extracts, which can be screened with any of the assays described herein. The term “small organic molecule” refers to a small molecule that is often identified as being an organic or medicinal compound, and does not include molecules that are exclusively nucleic acids, peptides or polypeptides.

The term “modulation” is art-recognized and refers to up regulation (i.e., activation or stimulation), down regulation (i.e., inhibition or suppression) of a response, or the two in combination or apart.

The term “treating” is art-recognized and refers to curing as well as ameliorating at least one symptom of any condition or disease.

The term “prophylactic” or “therapeutic” treatment is art-recognized and refers to administration of a drug to a host. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, i.e., it protects the host against developing the unwanted condition, whereas if administered after manifestation of the unwanted condition, the treatment is therapeutic (i.e., it is intended to diminish, ameliorate or maintain the existing unwanted condition or side effects therefrom).

A “patient,” “subject” or “host” to be treated by the subject method may mean either a human or non-human animal.

The term “mammal” is known in the art, and exemplary mammals include humans, primates, bovines, porcines, canines, felines, and rodents (e.g., mice and rats).

The term “bioavailable” when referring to a compound is art-recognized and refers to a form of a compound that allows for it, or a portion of the amount of compound administered, to be absorbed by, incorporated to, or otherwise physiologically available to a subject or patient to whom it is administered.

The term “pharmaceutically-acceptable salts” is art-recognized and refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds, including, for example, those contained in compositions described herein.

The term “pharmaceutically acceptable carrier” is art-recognized and refers to a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the subject composition and its components and not injurious to the patient. Some examples of materials which may serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.

The terms “systemic administration,” “administered systemically,” “peripheral administration” and “administered peripherally” are art-recognized and refer to the administration of a subject composition, therapeutic or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes.

The terms “parenteral administration” and “administered parenterally” are art-recognized and refer to modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intra-articulare, subcapsular, subarachnoid, intraspinal, and intrastemal injection and infusion.

Exemplary Methods and Compositions

Provided herein are methods for activating a sirtuin deacetylase protein family member (referred to as “sirtuin protein”). The methods may comprise contacting the sirtuin deacetylase protein family member with a compound, such as a polyphenol, e.g. a plant polyphenol, and referred to herein as “activation compound” or “activating compound.” Exemplary sirtuin deacetylase proteins include the yeast silent information regulator 2 (Sir2) and human SIRT1. Other family members include proteins having a significant amino acid sequence homology and biological activity, e.g., the ability to deacetylate target proteins, such as histones and p53, to those of Sir2 and SIRT1.

Exemplary activating compounds are those selected from the group consisting of flavones, stilbenes, flavanones, isoflavanones, catechins, chalcones, tannins and anthocyanidins. Exemplary stilbenes include hydroxystilbenes, such as trihydroxystilbenes, e.g., 3,5,4′-trihydroxystilbene (“resveratrol”). Resveratrol is also known as 3,4′5-stilbenetriol. Tetrahydroxystilbenes, e.g., piceatannol, are also encompassed. Hydroxychalones including trihydroxychalones, such as isoliquiritigenin, and tetrahydroxychalones, such as butein, can also be used. Hydroxyflavones including tetrahydroxyflavones, such as fisetin, and pentahydroxyflavones, such as quercetin, can also be used.

In one embodiment, methods for activating a sirtuin protein comprise an activating compound that is a stilbene or chalcone compound of formula 1:
wherein, independently for each occurrence,

R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 represent H, alkyl, aryl, heteroaryl, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

R represents H, alkyl, or aryl;

M represents O, NR, or S;

A-B represents a bivalent alkyl, alkenyl, alkynyl, amido, sulfonamido, diazo, ether, alkylamino, alkylsulfide, or hydrazine group; and

n is 0 or 1.

In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 0. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 1. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein A-B is ethenyl. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein A-B is —CH2CH(Me)CH(Me)CH2—. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein M is O. In a further embodiment, the methods comprises a compound of formula 1 and the attendant definitions, wherein R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 are H. In a further embodiment, the method comprise a compound of formula 1 and the attendant definitions, wherein R2, R4, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein R2, R4, R′2 and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein R3, R5, R′2 and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein R1, R3, R5, R′2 and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein R2 and R′2 are OH; R4 is O-β-D-glucoside; and R′3 is OCH3. In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein R2 is OH; R4 is O-β-D-glucoside; and R′3 is OCH3.

In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 0; A-B is ethenyl; and R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 are H (trans stilbene). In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 1; A-B is ethenyl; M is O; and R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 are H (chalcone). In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 0; A-B is ethenyl; R2, R4, and R′3 are OH; and R1, R3, R5, R′1, R′2, R′4, and R′5 are H (resveratrol). In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 0; A-B is ethenyl; R2, R4, R′2 and R′3 are OH; and R1, R3, R5, R′1, R′4 and R′5 are H (piceatannol). In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 1; A-B is ethenyl; M is O; R3, R5, R′2 and R′3 are OH; and R1, R2, R4, R′1, R′4, and R′5 are H (butein). In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 1; A-B is ethenyl; M is O; R1, R3, R5, R′2 and R′3 are OH; and R2, R4, R′1, R′4, and R′5 are H (3,4,2′,4′,6′-pentahydroxychalcone). In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 0; A-B is ethenyl; R2 and R′2 are OH, R4 is O-β-D-glucoside, R′3 is OCH3; and R1, R3, R5, R′1, R′4, and R′5 are H (rhapontin). In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 0; A-B is ethenyl; R2 is OH, R4 is O-β-D-glucoside, R′3 is OCH3; and R1, R3, R5, R′1, R′2, R′4, and R′5 are H (deoxyrhapontin). In a further embodiment, the methods comprise a compound of formula 1 and the attendant definitions, wherein n is 0; A-B is —CH2CH(Me)CH(Me)CH2—; R2, R3, R′2, and R′3 are OH; and R1, R4, R5, R′1, R′4, and R′5 are H (NDGA).

In another embodiment, methods for activating a sirtuin protein comprise an activating compound that is a flavanone compound of formula 2:

wherein, independently for each occurrence,

R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, R′5, and R″ represent H, alkyl, aryl, heteroaryl, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

R represents H, alkyl, or aryl;

M represents H2, O, NR, or S;

Z represents CR, O, NR, or S; and

X represents CR or N; and

Y represents CR or N.

In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein X and Y are both CH. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein M is O. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein. M is H2. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein Z is O. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R″ is H. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R″ is OH. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R″ is an ester. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R1 is
In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R1, R2, R3, R4, R′1, R′2, R′3, R′4, R′5 and R″ are H. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R2, R4, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R4, R′2, R′3, and R″ are OH. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R2, R4, R′2, R′3, and R″ are OH. In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein R2, R4, R′2, R′3, R′4, and R″ are OH.

In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein X and Y are CH; M is O; Z and O; R″ is H; and R1, R2, R3, R4, R′1, R′2, R′3, R′4, R′5 and R″ are H (flavanone). In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein X and Y are CH; M is O; Z and O; R″ is H; R2, R4, and R′3 are OH; and R1, R3, R′1, R′2, R′4, and R′5 are H (naringenin). In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein X and Y are CH; M is O; Z and O; R″ is OH; R2, R4, R′2, and R′3 are OH; and R1, R3, R′1, R′4, and R′5 are H (3,5,7,3′,4′-pentahydroxyflavanone). In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein X and Y are CH; M is H2; Z and O; R″ is OH; R2, R4, R′2, and R′3, are OH; and R1, R3, R′1, R′4 and R′5 are H (epicatechin). In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein X and Y are CH; M is H2; Z and O; R″ is OH; R2, R4, R′2, R′3, and R′4 are OH; and R1, R3, R′1, and R′5 are H (gallocatechin). In a further embodiment, the methods comprise a compound of formula 2 and the attendant definitions, wherein X and Y are CH; M is H2; Z and O; R″ is
R2, R4, R′2, R′3, R′4, and R″ are OH; and R1, R3, R′1, and R′5 are H (epigallocatechin gallate).

In another embodiment, methods for activating a sirtuin protein comprise an activating compound that is an iso flavanone compound of formula 3:

wherein, independently for each occurrence,

R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, R′5, and R″1, represent H, alkyl, aryl, heteroaryl, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

R represents H, alkyl, or aryl;

M represents H2, O, NR, or S;

Z represents CR, O, NR, or S; and

X represents CR or N; and

Y represents CR or N.

In another embodiment, methods for activating a sirtuin protein comprise an activating compound that is a flavone compound of formula 4:

wherein, independently for each occurrence,

R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5, represent H, alkyl, aryl, heteroaryl, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

R″ is absent or represents H, alkyl, aryl, heteroaryl, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

R represents H, alkyl, or aryl;

M represents H2, O, NR, or S;

Z represents CR, O, NR, or S; and

X represents CR or N when R″ is absent or C when R″ is present.

In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CR. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein Z is O. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein M is O. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R″ is H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R″ is OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R′2, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R4, R′2, R′3, and R′4 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R4, R′2, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R3, R′2, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R4, R′2, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R′2, R′3, and R′4 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R4, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R3, R4, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R4, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R3, R′1, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2 and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R1, R2, R′2, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R3, R′1, and R′2 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R′3 is OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R4 and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2 and R4 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R4, R′1, and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R4 is OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R4, R′2, R′3, and R′4 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R2, R′2, R′3, and R′4 are OH. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein R1, R2, R4, R′2, and R′3 are OH.

In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; and R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 are H (flavone). In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C; R″ is OH; Z is O; M is O; R2, R′2, and R′3 are OH; and R1, R3, R4, R′1, R′4, and R′5 are H (fisetin). In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R2, R4, R′2, R′3, and R′4 are OH; and R1, R3, R′1, and R′5 are H (5,7,3′,4′,5′-pentahydroxyflavone). In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R2, R4, R′2, and R′3 are OH; and R1, R3, R′1, R′4, and R′5 are H (luteolin). In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C, R″ is OH; Z is O; M is O; R3, R′2, and R′3 are OH; and R1, R2, R4, R′1, R′4, and R′5 are H (3,6,3′,4′-tetrahydroxyflavone). In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C, R″ is OH;Zis O; M is O; R2, R4, R′2, and R′3 are OH; and R1, R3, R′1, R′4, and R′5 are H (quercetin). In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R2, R′2, R′3, and R′4 are OH; and R1, R3, R4, R′1, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C; R″ is OH; Z is O; M is O; R2, R4, and R′3 are OH; and R1, R3, R′1, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R2, R3, R4, and R′3 are OH; and R1, R′1, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R2, R4, and R′3 are OH; and R1, R3, R′1, R′2, R′4, and R′5 are H. In a embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C, R″ is OH; Z is O; M is O; R3, R′1, and R′3 are OH; and R1, R2, R4, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R2 and R′3 are OH; and R1, R3, R4, R′1, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C, R″ is OH; Z is O; M is O; R1, R2, R′2, and R′3 are OH; and R1, R2, R4, R′3, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C; R″ is OH; Z is O; M is O; R3, R′1, and R′2 are OH; and R1, R2, R4; R′3, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R′3 is OH; and R1, R2, R3, R4, R′1, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R4 and R′3 are OH; and R1, R2, R3, R′1, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R2 and R4 are OH; and R1, R3, R′1, R′2, R′3, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C; R″ is OH; Z is O; M is O; R2, R4, R′1, and R′3 are OH; and R1, R3, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is CH; R″ is absent; Z is O; M is O; R4 is OH; and R1, R2, R3, R′1, R′2, R′3, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C; R″ is OH; Z is O; M is O; R2, R4, R′2, R′3, and R′4 are OH; and R1, R3, R′1, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C; R″ is OH; Z is O; M is O; R2, R′2, R′3, and R′4 are OH; and R1, R3, R4, R′1, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 4 and the attendant definitions, wherein X is C; R″ is OH; Z is O; M is O; R1, R2, R4, R′2, and R′3 are OH; and R3, R′1, R′4, and R′5 are H.

In another embodiment, methods for activating a sirtuin protein comprise an activating compound that is an iso flavone compound of formula 5:

wherein, independently for each occurrence,

R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5, represent H, alkyl, aryl, heteroaryl, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

R″ is absent or represents H, alkyl, aryl, heteroaryl, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

R represents H, alkyl, or aryl;

M represents H2, O, NR, or S;

Z represents CR, O, NR, or S; and

Y represents CR or N when R″ is absent or C when R″ is present.

In a further embodiment, the methods comprise a compound of formula 5 and the attendant definitions, wherein Y is CR. In a further embodiment, the methods comprise a compound of formula 5 and the attendant definitions, wherein Y is CH. In a further embodiment, the methods comprise a compound of formula 5 and the attendant definitions, wherein Z is O. In a further embodiment, the methods comprise a compound of formula 5 and the attendant definitions, wherein M is O. In a further embodiment, the methods comprise a compound of formula 5 and the attendant definitions, wherein R2 and R′3 are OH. In a further embodiment, the methods comprise a compound of formula 5 and the attendant definitions, wherein R2, R4, and R′3 are OH.

In a further embodiment, the methods comprise a compound of formula 5 and the attendant definitions, wherein Y is CH; R″ is absent; Z is O; M is O; R2 and R′3 are OH; and R1, R3, R4, R′1, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound of formula 5 and the attendant definitions, wherein Y is CH; R″ is absent; Z is O; M is O; R2, R4, and R′3 are OH; and R1, R3, R′1, R′2, R′4, and R′5 and H.

In another embodiment, methods for activating a sirtuin protein comprise an activating compound that is an anthocyanidin compound of formula 6:

wherein, independently for each occurrence,

R3, R4, R5, R6, R7, R8, R′2, R′3, R′4, R′5, and R′6 represent H, alkyl, aryl, heteroaryl, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

R represents H, alkyl, or aryl; and

A represents an anion selected from the following: Cl, Br, or I.

In a further embodiment, the methods comprise a compound of formula 6 and the attendant definitions, wherein A is Cl. In a further embodiment, the methods comprise a compound of formula 6 and the attendant definitions, wherein R3, R5, R7, and R′4 are OH. In a further embodiment, the methods comprise a compound of formula 6 and the attendant definitions, wherein R3, R5, R7, R′3, and R′4 are OH. In a further embodiment, the methods comprise a compound of formula 6 and the attendant definitions, wherein R3, R5, R7, R′3, R′4, and R′5 are OH.

In a further embodiment, the methods comprise a compound of formula 6 and the attendant definitions, wherein A is Cl; R3, R5, R7, and R′4 are OH; and R4, R6, R8, R′2, R′3, R′5, and R′6 are H. In a further embodiment, the methods comprise a compound of formula 6 and the attendant definitions, wherein Ais Cl; R3, R5, R7, R′3, and R′4 are OH; and R4, R6, R8, R′2, R′5, and R′6 are H. In a further embodiment, the methods comprise a compound of formula 6 and the attendant definitions, wherein A is Cl; R3, R5, R7, R′3, R′4, and R′5 are OH; and R4, R6, R8, R′2, and R′6 are H.

Methods for activating a sirtuin protein may also comprise a stilbene, chalcone, or flavone compound represented by formula 7:

wherein, independently for each occurrence,

M is absent or O;

R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 represent H, alkyl, aryl, heteroary alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

Ra represents H or the two Ra form a bond;

R represents H, alkyl, or aryl; and

n is 0 or 1.

In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein n is 0. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein n is 1. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein M is absent. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein M is O. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein Ra is H. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein M is O and the two Ra form a bond.

In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein R5 is H. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein R5 is OH. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein R1, R3, and R′3 are OH. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein R2, R4, R′2, and R′3 are OH. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein R2, R′2, and R′3 are OH. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein R2 and R4 are OH.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 7 and the attendant definitions, wherein n is 0; M is absent; Ra is H; R5 is H; R1, R3, and R′3 are OH; and R2, R4, R′1, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein n is 1; M is absent; Ra is H; R5 is H; R2, R4, R′2, and R′3 are OH; and R1, R3, R′1, R′4, and R′5 are H. In a further embodiment, the methods comprise an activating compound represented by formula 7 and the attendant definitions, wherein n is 1; M is O; the two Ra form a bond; R5 is OH; R2, R′2, and R′3 are OH; and R1, R3, R4, R′1, R′4, and R′5 are H.

Other compounds for activating sirtuin deacetylase protein family members include compounds having a formula selected from the group consisting of formulas 8-25 and 30 set forth below.

R═H, alkyl, aryl, heterocyclyl, or heteroaryl

R′═H, halogen, NO2, SR, OR, NR2, alkyl, aryl, or carboxy

R═H, alkyl, aryl, heterocyclyl, or heteroaryl

wherein, independently for each occurrence,

R′═H, halogen, NO2, SR, OR, NR2, alkyl, aryl, or carboxy

R═H, alkyl, aryl, heterocyclyl, or heteroaryl

wherein, independently for each occurrence,

L represents CR2, O, NR, or S;

R represents H, alkyl, aryl, aralkyl, or heteroaralkyl; and

R′ represents H, halogen, NO2, SR, OR, NR2, alkyl, aryl, or carboxy.

wherein, independently for each occurrence,

L represents CR2, O, NR, or S;

W represents CR or N;

R represents H, alkyl, aryl, aralkyl, or heteroaralkyl;

Ar represents a fused aryl or heteroaryl ring; and

R′ represents H, halogen, NO2, SR, OR, NR2, alkyl, aryl, or carboxy.

wherein, independently for each occurrence,

L represents CR2, O, NR, or S;

R represents H, alkyl, aryl, aralkyl, or heteroaralkyl; and

R′ represents H, halogen, NO2, SR, OR, NR2, alkyl, aryl, or carboxy.

wherein, independently for each occurrence,

L represents CR2, O, NR, or S;

R represents H, alkyl, aryl, aralkyl, or heteroaralkyl; and

R′ represents H, halogen, NO2, SR, OR, NR2, alkyl, aryl, or carboxy.

Methods for activating a sirtuin protein may also comprise a stilbene, chalcone, or flavone compound represented by formula 30:

wherein, independently for each occurrence,

D is a phenyl or cyclohexyl group;

R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 represent H, alkyl, aryl, heteroary, alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, carboxyl, azide, ether; or any two adjacent R or R′ groups taken together form a fused benzene or cyclohexyl group;

R represents H, alkyl, or aryl; and

A-B represents an ethylene, ethenylene, or imine group;

provided that when A-B is ethenylene and R′3 is H: R3 is not OH when R1, R2, R4, and R5 are H; and R2 and R4 are not OMe when R1, R3, and R5 are H; and R3 is not OMe when R1, R2, R4, and R5 are H.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein D is a phenyl group.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is an ethenylene or imine group.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is an ethenylene group.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein R2 is OH.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein R4 is OH

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein R2 and R4 are OH.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein D is a phenyl group; and A-B is an ethenylene group.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein D is a phenyl group; A-B is an ethenylene group; and R2 and R4 are OH.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is Cl.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is OH.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is H.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is CH2CH3.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is F.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is Me.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is an azide.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is SMe.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is NO2.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is CH(CH3)2.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is OMe.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; R′2 is OH; and R′3 is OMe.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 is OH; R4 is carboxyl; and R′3 is OH.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is carboxyl.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 and R′4 taken together form a fused benzene ring.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; and R4 is OH.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OCH2OCH3; and R′3 is SMe.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is carboxyl.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a cyclohexyl ring; and R2 and R4 are OH.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; and R3 and R4 are OMe.

In a further embodiment, the methods include contacting a cell with an activating compound represented by formula 30 and the attendant definitions, wherein A-B is ethenylene; D is a phenyl ring; R2 and R4 are OH; and R′3 is OH.

Exemplary activating compounds are those listed in the appended Tables having a ratio to control rate of more than one. A preferred compound of formula 8 is Dipyridamole; a preferred compound of formula 12 is Hinokitiol; a preferred compound of formula 13 is L-(+)-Ergothioneine; a preferred compound of formula 19 is Caffeic Acid Phenol Ester; a preferred compound of formula 20 is MCI-186 and a preferred compound of formula 21 is HBED (Supplementary Table 6).

Also included are pharmaceutically acceptable addition salts and complexes of the compounds of formulas 1-25 and 30. In cases wherein the compounds may have one or more chiral centers, unless specified, the compounds contemplated herein may be a single stereoisomer or racemic mixtures of stereoisomers.

In cases in which the compounds have unsaturated carbon-carbon double bonds, both the cis (Z) and trans (E) isomers are contemplated herein. In cases wherein the compounds may exist in tautomeric forms, such as keto-enol tautomers, such as
and
each tautomeric form is contemplated as being included within the methods presented herein, whether existing in equilibrium or locked in one form by appropriate substitution with R′. The meaning of any substituent at any one occurrence is independent of its meaning, or any other substituent's meaning, at any other occurrence.

Also included in the methods presented herein are prodrugs of the compounds of formulas 1-25 and 30. Prodrugs are considered to be any covalently bonded carriers that release the active parent drug in vivo.

Analogs and derivatives of the above-described compounds can also be used for activating a member of the sirtuin protein family. For example, derivatives or analogs may make the compounds more stable or improve their ability to traverse cell membranes or being phagocytosed or pinocytosed. Exemplary derivatives include glycosylated derivatives, as described, e.g., in U.S. Pat. No. 6,361,815 for resveratrol. Other derivatives of resveratrol include cis- and trans-resveratrol and conjugates thereof with a saccharide, such as to form a glucoside (see, e.g., U.S. Pat. No. 6,414,037). Glucoside polydatin, referred to as piceid or resveratrol 3-O-beta-D-glucopyranoside, can also be used. Saccharides to which compounds may be conjugated include glucose, galactose, maltose, lactose and sucrose. Glycosylated stilbenes are further described in Regev-Shoshani et al. Biochemical J. (published on Apr. 16, 2003 as BJ20030141). Other derivatives of compounds described herein are esters, amides and prodrugs. Esters of resveratrol are described, e.g., in U.S. Pat. No. 6,572,882. Resveratrol and derivatives thereof can be prepared as described in the art, e.g., in U.S. Pat. Nos. 6,414,037; 6,361,815; 6,270,780; 6,572,882; and Brandolini et al. (2002) J. Agric. Food. Chem.50:7407. Derivatives of hydroxyflavones are described, e.g., in U.S. Pat. No. 4,591,600. Resveratrol and other activating compounds can also be obtained commercially, e.g., from Sigma.

In certain embodiments, if an activating compound occurs naturally, it may be at least partially isolated from its natural environment prior to use. For example, a plant polyphenol may be isolated from a plant and partially or significantly purified prior to use in the methods described herein. An activating compound may also be prepared synthetically, in which case it would be free of other compounds with which it is naturally associated. In an illustrative embodiment, an activating composition comprises, or an activating compound is associated with, less than about 50%, 10%, 1%, 0.1%, 10−2% or 10−3% of a compound with which it is naturally associated.

Sirtuin proteins may be activated in vitro, e.g., in a solution or in a cell. In one embodiment, a sirtuin protein is contacted with an activating compound in a solution. A sirtuin is activated by a compound when at least one of its biological activities, e.g., deacetylation activity, is higher in the presence of the compound than in its absence. Activation may be by a factor of at least about 10%, 30%, 50%, 100% (i.e., a factor of two), 3, 10, 30, or 100. The extent of activation can be determined, e.g., by contacting the activated sirtuin with a deacetylation substrate and determining the extent of deacetylation of the substrate, as further described herein. The observation of a lower level of acetylation of the substrate in the presence of a test sirtuin relative to the presence of a non activated control sirtuin indicates that the test sirtuin is activated. The solution may be a reaction mixture. The solution may be in a dish, e.g., a multiwell dish. Sirtuin proteins may be prepared recombinantly or isolated from cells according to methods known in the art.

In another embodiment, a cell comprising a sirtuin deacetylase protein is contacted with an activating compound. The cell may be a eukaryotic cell, e.g., a mammalian cell, such as a human cell, a yeast cell, a non-human primate cell, a bovine cell, an ovine cell, an equine cell, a porcine cell, a sheep cell, a bird (e.g., chicken or fowl) cell, a canine cell, a feline cell or a rodent (mouse or rat) cell. It can also be a non-mammalian cell, e.g., a fish cell. Yeast cells include S. cerevesiae and C. albicans. The cell may also be a prokaryotic cell, e.g., a bacterial cell. The cell may also be a single-celled microorganism, e.g., a protozoan. The cell may also be a metazoan cell, a plant cell or an insect cell. The application of the methods decribed herein to a large number of cell types is based at least on the high convervation of sirtuins from humans to fungi, protozoans, metazoans and plants.

In one embodiment, the cells are in vitro. A cell may be contacted with a solution having a concentration of an activating compound of less than about 0.1 μM; 0.5 μM; less than about 1 μM; less than about 10 μM or less than about 100 μM. The concentration of the activating compound may also be in the range of about 0.1 to 1 μM, about 1 to 10 μM or about 10 to 100 μM. The appropriate concentration may depend on the particular compound and the particular cell used as well as the desired effect. For example, a cell may be contacted with a “sirtuin activating” concentration of an activating compound, e.g., a concentration sufficient for activating the sirtuin by a factor of at least 10%, 30%, 50%, 100%, 3, 10, 30, or 100.

In certain embodiments, a cell is contacted with an activating compound in vivo, such as in a subject. The subject can be a human, a non-human primate, a bovine, an ovine, an equine, a porcine, a sheep, a canine, a feline or a rodent (mouse or rat). For example, an activating compound may be administered to a subject. Administration may be local, e.g., topical, parenteral, oral, or other depending on the desired result of the administration (as further described herein). Administration may be followed by measuring a factor in the subject, such as measuring the activity of the sirtuin. In an illustrative embodiment, a cell is obtained from a subject following administration of an activating compound to the subject, such as by obtaining a biopsy, and the activity of the sirtuin is determined in the biopsy. The cell may be any cell of the subject, but in cases in which an activating compound is administered locally, the cell is preferably a cell that is located in the vicinity of the site of administration.

Also provided are methods for modulating the acetylation level of p53 proteins. As shown herein (see, e.g., the Examples), lysine 382 of p53 proteins in cells is deacetylated following incubation of cells in the presence of low concentrations of resveratrol. Accordingly, “p53 deacetylating concentrations” of compounds include, e.g., concentrations of less than about 0.1 μM, 0.5 μM, 1 μM, 3 μM, 50 μM, 100 μM or 300 μM. It has also been shown herein that p53 proteins in cells are acetylated in the presence of higher concentrations of resveratrol. Accordingly, “p53 acetylating concentrations” of compounds include, e.g., concentrations of at least about 10 μM, 30 μM, 100 μM or 300 μM. The level of acetylation of p53 can be determined by methods known in the art, e.g., as further described in the Examples.

Other methods contemplated are methods for protecting a cell against apoptosis. Without wanting to be limited to a particular mechanism of action, but based at least in part on the fact that acetylation of p53 proteins activates p53 proteins and that activated p53 proteins induce apoptosis, incubating cells comprising p53 proteins in the presence of a p53 deacetylating concentration of an activating compound prevents the induction of apoptosis of the cells. Accordingly, a cell can be protected from apoptosis by activating sirtuins by contacting the cell with an amount of an activating compound sufficient or adequate for protecting against apoptosis, e.g., less than about 0.1 μM, 0.5 μM, 1 μM, 3 μM or 10 μM. An amount sufficient or adequate for protection against apoptosis can also be determined experimentally, such as by incubating a cell with different amounts of an activating compound, subjecting the cell to an agent or condition that induces apoptosis, and comparing the extent of apoptosis in the presence of different concentrations or the absence of an enhancing compound and determining the concentration that provides the desired protection. Determining the level of apoptosis in a population of cells can be performed according to methods known in the art.

Yet other methods contemplated herein are methods for inducing apoptosis in a cell. Without wanting to be limited to a particular mechanism of action, as shown in the Examples, at certain concentrations of compounds, p53 proteins are acetylated rather than deacetylated, thereby activating the p53 proteins, and inducing apoptosis. Apoptosis inducing concentrations of compounds may be, e.g., at least about 10 μM, 30 μM, 100 μM or 300 μM.

Appropriate concentrations for modulating p53 deacetylation and apoptosis can be determined according to methods, e.g., those described herein. Concentrations may vary slightly from one cell to another, from one activating compound to another and whether the cell is isolated or in an organism.

Cells in which p53 acetylation and apoptosis may be modulated can be in vitro, e.g., in cell culture, or in vivo, e.g., in a subject. Administration of an activating compound to a subject can be conducted as further described herein. The level of p53 acetylation and/or apoptosis in cells of the subject can be determined, e.g., by obtaining a sample of cells from the subject and conducting an in vitro analysis of the level of p53 acetylation and/or apoptosis.

Also provided herein are methods for extending the lifespan of a eukaryotic cells and/or increasing their resistance to stress comprising, e.g., contacting the eukaryotic cell with a compound, e.g., a polyphenol compound. Exemplary compounds include the activating compounds described herein, such as compounds of the stilbene, flavone and chalcone families. Although the Examples show that quercetin and piceatannol, which activate sirtuins, were not found to significantly affect the lifespan of eukaryotic cells, it is believed that this may be the result of a lack of entry of the compounds into the cell or potentially the existence of another pathway overriding activation of sirtuins. Derivatives and analogs of these compounds or administration of these compound to other cells or by other methods are expected to activate sirtuins.

In one embodiment, methods for extending the lifespan of a eukaryotic cell and/or increasing its resistance to stress comprise contacting the cell with a stilbene, chalcone, or flavone compound represented by formula 7:

wherein, independently for each occurrence,

M is absent or O;

R1, R2, R3, R4, R5, R′1, R′2, R′3, R′4, and R′5 represent H, alkyl, aryl, heteroary alkaryl, heteroaralkyl, halide, NO2, SR, OR, N(R)2, or carboxyl;

Ra represents H or the two Ra form a bond;

R represents H, alkyl, or aryl; and

n is 0 or 1.

In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein n is 0. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein n is 1. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein M is absent. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein M is O. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein Ra is H. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein M is O and the two Ra form a bond. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein R5 is H. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein R5 is OH. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein R1, R3, and R′3 are OH. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein R2, R4, R′2, and R′3 are OH. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein R2, R′2, and R′3 are OH.

In a further embodiment, methods for extending the lifespan of a eukaryotic cell comprise contacting the cell with a compound represented by formula 7 and the attendant definitions, wherein n is 0; M is absent; Ra is H; R5 is H; R1, R3, and R′3 are OH; and R2, R4, R′1, R′2, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein n is 1; M is absent; Ra is H; R5 is H; R2, R4, R′2, and R′3 are OH; and R1, R3, R′1, R′4, and R′5 are H. In a further embodiment, the methods comprise a compound represented by formula 7 and the attendant definitions, wherein n is 1; M is O; the two Ra form a bond; R5 is OH; R2, R′2, and R′3 are OH; and R1, R3, R4, R′1, R′4, and R′5 are H.

The eukaryotic cell whose lifespan may be extended can be a human, a non-human primate, a bovine, an ovine, an equine, a porcine, a sheep, a canine, a feline, a rodent (mouse or rat) or a yeast cell. A yeast cell may be Saccharomyces cerevisiae or Candida albicans. Concentrations of compounds for this purpose may be about 0.1 μM, 0.3 μM, 0.5 μM, 1 μM, 3 μM, 10 μM, 30 μM, 100 μM or 300 μM. Based at least on the high conservation of Sir2 proteins in various organisms, lifespan can also be prolonged in prokaryotes, protozoans, metazoans, insects and plants.

The cell may be in vitro or in vivo. In some embodiments, a life-extending compound is administered to an organism (e.g., a subject) such as to induce hormesis, i.e., an increasing resistance to mild stress that results in increasing the lifespan of the organism. In fact, it has been shown that SIR2 is essential for the increased longevity provided by calorie restriction, a mild stress, that extends the lifespan of every organism it has been tested on (Lin et al. (2000) Science 249:2126). For example, overexpression of a Caenorhabditis. elegans SIR2 homologue, sir-2.1, increases lifespan via a forkhead transcription factor, DAF-16, and a SIR2 gene has recently been implicated in lifespan regulation in Drosophila melanogaster (Rogina et al. Science (2002) 298:1745). Furthermore, the closest human Sir2 homologue, SIRT1, promotes survival in human cells by down-regulating the activity of the tumor suppressor p53 (Tissenbaum et al. Nature 410, 227-30 (2001); Rogina et al. Science, in press (2002); and Vaziri, H. et al. Cell 107, 149-59. (2001)). The role of SIR2 in stress resistance and cell longevity is further supported by the identification of PNC1 as a calorie restriction- and stress-responsive gene that increases lifespan and stress resistance of cells by depleting intracellular nicotinamide (Anderson et al. (2003) Nature 423:181 and Bitterman et al. (2002) J. Biol. Chem. 277: 45099). Accordingly, compounds may be administered to a subject for protecting the cells of the subject from stresses and thereby extending the lifespan of the cells of the subject.

Also encompassed are methods for inhibiting sirtuins; inhibiting deacetylation of p53, e.g., for stimulating acetylation of p53; stimulating apoptosis; reducing lifespan and/or rendering cells or organisms more sensitive to stresses. Methods may include contacting a cell or a molecule, such as a sirtuin or a p53 protein, with a compound that inhibits sirtuins, i.e., an “inhibiting compound,” such, a compound having a formula selected from the group of formulas 26-29 and 31:

wherein, independently for each occurrence,

R′ represents H, halogen, NO2, SR, OR, NR2, alkyl, aryl, or carboxy;

R represents H, alkyl, aryl, aralkyl, or heteroaralkyl; and

R″ represents alkyl, alkenyl, or alkynyl.

wherein, independently for each occurrence,

L represents O, NR, or S;

R represents H, alkyl, aryl, aralkyl, or heteroaralkyl;

R′ represents H, halogen, NO2, SR, SO3, OR, NR2, alkyl, aryl, or carboxy;

a represents an integer from 1 to 7 inclusively; and

b represents an integer from 1 to 4 inclusively.

wherein, independently for each occurrence,

L represents O, NR, or S;

R represents H, alkyl, aryl, aralkyl, or heteroaralkyl;

R′ represents H, halogen, NO2, SR, SO3, OR, NR2, alkyl, aryl, or carboxy;

a represents an integer from 1 to 7 inclusively; and

b represents an integer from 1 to 4 inclusively.

wherein, independently for each occurrence,

L represents O, NR, or S;

R represents H, alkyl, aryl, aralkyl, or heteroaralkyl;

R′ represents H, halogen, NO2, SR, SO3, OR, NR2, alkyl, aryl, or carboxy;

a represents an integer from 1 to 7 inclusively; and

b represents an integer from 1 to 4 inclusively.

wherein, independently for each occurrence,

R2, R3, and R4 are H, OH, or O-alkyl;

R′3 is H or NO2; and

A-B is an ethenylene or amido group.

In a further embodiment, the inhibiting compound is represented by formula 31 and the attendant definitions, wherein R3 is OH, A-B is ethenylene, and R′3 is H.

In a further embodiment, the inhibiting compound is represented by formula 31 and the attendant definitions, wherein R2 and R4 are OH, A-B is an amido group, and R′3 is H.

In a further embodiment, the inhibiting compound is represented by formula 31 and the attendant definitions, wherein R2 and R4 are OMe, A-B is ethenylene, and R′3 is NO2.

In a further embodiment, the inhibiting compound is represented by formula 31 and the attendant definitions, wherein R3 is OMe, A-B is ethenylene, and R′3 is H.

Also included are pharmaceutically acceptable addition salts and complexes of the compounds of formulas 26-29 and 31. In cases wherein the compounds may have one or more chiral centers, unless specified, the compounds contemplated herein may be a single stereoisomer or racemic mixtures of stereoisomers.

Exemplary inhibitory compounds are those set forth in the appended Tables for which the “ratio to control rate” is lower than one.

In cases in which the compounds have unsaturated carbon-carbon double bonds, both the cis (Z) and trans (E) isomers are contemplated herein. In cases wherein the compounds may exist in tautomeric forms, such as keto-enol tautomers, such as
and
each tautomeric form is contemplated as being included within the methods presented herein, whether existing in equilibrium or locked in one form by appropriate substitution with R′. The meaning of any substituent at any one occurrence is independent of its meaning, or any other substituent's meaning, at any other occurrence.

Also included in the methods presented herein are prodrugs of the compounds of formulas 26-29 and 31. Prodrugs are considered to be any covalently bonded carriers that release the active parent drug in vivo.

Inhibitory compounds may be contacted with a cell, administered to a subject, or contacted with one or more molecules, such as a sirtuin protein and a p53 protein. Doses of inhibitory compounds may be similar to those of activating compounds.

Whether in vitro or in vivo, a cell may also be contacted with more than one compound (whether an activating compound or an inhibiting compound). A cell may be contacted with at least 2, 3, 5, or 10 different compounds. A cell may be contacted simultaneously or sequentially with different compounds.

Also encompassed are compositions comprising one or more activating or inhibiting compounds having a formula selected from the group of formulas 1-31. Compounds may be in a pharmaceutical composition, such as a pill or other formulation for oral administration, further described herein. Compositions may also comprise or consist of extracts of plants, red wine or other source of the compounds.

Yet other methods contemplated herein include sceening methods for identifying compounds that modulate sirtuins. Assays may be conducted in a cell based or cell free format. For example, an assay may comprise incubating (or contacting) a sirtuin with a test compound under conditions in which a sirtuin can be activated by an agent known to activate the sirtuin, and monitoring or determining the level of activation of the sirtuin in the presence of the test compound relative to the absence of the test compound. The level of activation of a sirtuin can be determined by determining its ability to deacetylate a substrate. Exemplary substrates are acetylated peptides, e.g., those set forth in FIG. 5, which can be obtained from BIOMOL (Plymouth Meeting, Pa.). Preferred substrates include peptides of p53, such as those comprising an acetylated K382. A particularly preferred substrate is the Fluor de Lys-SIRT1 (BIOMOL), i.e., the acetylated peptide Arg-His-Lys-Lys. Other substrates are peptides from human histones H3 and H4 or an acetylated amino acid (see FIG. 5). Substrates may be fluorogenic. The sirtuin may be SIRT1 or Sir2 or a portion thereof. For example, recombinant SIRT1 can be obtained from BIOMOL. The reaction may be conducted for about 30 minutes and stopped, e.g., with nicotinamide. The HDAC fluorescent activity assay/drug discovery kit (AK-500, BIOMOL Research Laboratories) may be used to determine the level of acetylation. Similar assays are described in Bitterman et al. (2002) J. Biol. Chem. 277:45099. The level of activation of the sirtuin in an assay may be compared to the level of activation of the sirtuin in the presence of one or more (separately or simultaneously) compounds described herein, which may serve as positive or negative controls. Sirtuins for use in the assays may be full length sirtuin proteins or portions thereof. Since it has been shown herein that activating compounds appear to interact with the N-terminus of SIRT1, proteins for use in the assays include N-terminal portions of sirtuins, e.g., about amino acids 1-176 or 1-255 of SIRT1; about amino acids 1-174 or 1-252 of Sir2.

In one embodiment, a screening assay comprises (i) contacting a sirtuin with a test compound and an acetylated substrate under conditions appropriate for the sirtuin to deacetylate the substrate in the absence of the test compound; and (ii) determining the level of acetylation of the substrate, wherein a lower level of acetylation of the substrate in the presence of the test compound relative to the absence of the test compound indicates that the test compound stimulates deacetylation by the sirtuin, whereas a higher level of acetylation of the substrate in the presence of the test compound relative to the absence of the test compound indicates that the test compound inhibits deacetylation by the sirtuin.

Methods for identifying compounds that modulate, e.g., stimulate or inhibit, sirtuins in vivo may comprise (i) contacting a cell with a test compound and a substrate that is capable of entering a cell in the presence of an inhibitor of class I and class II HDACs under conditions appropriate for the sirtuin to deacetylate the substrate in the absence of the test compound; and (ii) determining the level of acetylation of the substrate, wherein a lower level of acetylation of the substrate in the presence of the test compound relative to the absence of the test compound indicates that the test compound stimulates deacetylation by the sirtuin, whereas a higher level of acetylation of the substrate in the presence of the test compound relative to the absence of the test compound indicates that the test compound inhibits deacetylation by the sirtuin. A preferred substrate is an acetylated peptide, which is also prefeably fluorogenic, as further described herein (Examples). The method may further comprise lysing the cells to determine the level of acetylation of the substrate. Substrates may be added to cells at a concentration ranging from about 1 μM to about 10 mM, preferably from about 10 μM to 1 mM, even more preferably from about 100 μM to 1 mM, such as about 200 μM. A preferred substrate is an acetylated lysine, e.g., ε-acetyl lysine (Fluor de Lys, FdL) or Fluor de Lys-SIRT1. A preferred inhibitor of class I and class II HDACs is trichostatin A (TSA), which may be used at concentrations ranging from about 0.01 to 100 μM, preferably from about 0.1 to 10 μM, such as 1 μM. Incubation of cells with the test compound and the substrate may be conducted for about 10 minutes to 5 hours, preferably for about 1-3 hours. Since TSA inhibits all class I and class II HDACs, and that certain substrates, e.g., Fluor de Lys, is a poor substrate for SIRT2 and even less a substrate for SIRT3-7, such an assay may be used to identify modulators of SIRT1 in vivo. An exemplary assay is further described in the Examples and shown in FIG. 4a.

Also provided herein are assays for identifying agents that are capable of extending or reducing the lifespan of cells and/or increasing or decreasing their resistance to stress. A method may comprise incubating cells with a test compound and determining the effect of the test compound on rDNA silencing and rDNA recombination, wherein an increase in the frequency of rDNA recombination and an absence of effect on rDNA silencing in the presence of the test compound relative to the absence of the test compound indicates that the test compound extends lifespan. This assay is based at least on the observation that resveratrol reduced the frequency of rDNA recombination by about 60% in a SIR2 dependent manner, but did not increasing rDNA silencing.

Also provided herein are methods for identifying the binding site of activating or inhibitory compounds in sirtuin proteins. In one embodiment, BML-232 (Table 10) is used. BML-232, has very similar SIRT1 activating properties to resveratrol and contains a phenylazide function. Phenylazide groups may be activated by the absorption of ultraviolet light to form reactive nitrenes. When a protein-bound phenylazide is light-activated it can react to form covalent adducts with various protein functional groups in the site to which it is bound. The photo cross-linked protein may then be analyzed by proteolysis/mass spectrometry to identify amino acid residues which may form part of the binding site for the compound. This information, in combination with published three dimensional structural information on SIRT1 homologs could be used to aid the design of new, possibly higher affinity, SIRT1 activating ligands.

Exemplary Uses

In one embodiment, cells are treated in vitro as described herein to extend their lifespan, e.g., to keep them proliferating longer and/or increasing its resistance to stress or prevent apoptosis. That compounds described herein may increase resistance to stress is based at least on the observation that Sir2 provides stress resistance and that PNC1 modulates Sir2 activity in response to cell stress (Anderson et al. (2003) Nature 423:181). This is particularly useful for primary cell cultures (i.e., cells obtained from an organism, e.g., a human), which are known to have only a limited lifespan in culture. Treating such cells according to methods described herein, e.g., by contacting them with an activating or lifespan extending compound, will result in increasing the amount of time that the cells are kept alive in culture. Embryonic stem (ES) cells and pluripotent cells, and cells differentiated therefrom, can also be treated according to the methods described herein such as to keep the cells or progeny thereof in culture for longer periods of time. Primary cultures of cells, ES cells, pluripotent cells and progeny thereof can be used, e.g., to identify compounds having particular biological effects on the cells or for testing the toxicity of compounds on the cells (i.e., cytotoxicity assays). Such cells can also be used for transplantation into a subject, e.g., after ex vivo modification.

In other embodiments, cells that are intended to be preserved for long periods of time are treated as described herein. The cells can be cells in suspension, e.g., blood cells, serum, biological growth media, or tissues or organs. For example, blood collected from an individual for administering to an individual can be treated as described herein, such as to preserve the blood cells for longer periods of time, such as for forensic purposes. Other cells that one may treat for extending their lifespan or protect against apoptosis include cells for consumption, e.g., cells from non-human mammals (such as meat), or plant cells (such as vegetables).

Compounds may also be applied during developmental and growth phases in mammals, plants, insects or microorganisms, in order to, e.g., alter, retard or accelerate the developmental and/or growth process.

In another embodiment, cells obtained from a subject, e.g., a human or other mammal, are treated according to methods described herein and then administered to the same or a different subject. Accordingly, cells or tissues obtained from a donor for use as a graft can be treated as described herein prior to administering to the recipient of the graft. For example, bone marrow cells can be obtained from a subject, treated ex vivo, e.g., to extend their lifespan, and then administered to a recipient. The graft can be an organ, a tissue or loose cells.

In yet other embodiments, cells are treated in vivo, e.g., to increase their lifespan or prevent apoptosis. For example, skin can be protected from aging, e.g., developing wrinkles, by treating skin, e.g., epithelial cells, as described herein. In an exemplary embodiment, skin is contacted with a pharmaceutical or cosmetic composition comprising a compound described herein. Exemplary skin afflictions or skin conditions include disorders or diseases associated with or caused by inflammation, sun damage or natural aging. For example, the compositions find utility in the prevention or treatment of contact dermatitis (including irritant contact dermatitis and allergic contact dermatitis), atopic dermatitis (also known as allergic eczema), actinic keratosis, keratinization disorders (including eczema), epidermolysis bullosa diseases (including penfigus), exfoliative dermatitis, seborrheic dermatitis, erythemas (including erythema multiforme and erythema nodosum), damage caused by the sun or other light sources, discoid lupus erythematosus, dermatomyositis, skin cancer and the effects of natural aging. The formulations may be administered topically, to the skin or mucosal tissue, as an ointment, lotion, cream, microemulsion, gel, solution or the like, as described in the preceding section, within the context of a dosing regimen effective to bring about the desired result. A dose of active agent may be in the range of about 0.005 to about 1 micromoles per kg per day, preferably about 0.05 to about 0.75 micromoles per kg per day, more typically about 0.075 to about 0.5 micromoles per kg per day. It will be recognized by those skilled in the art that the optimal quantity and spacing of individual dosages will be determined by the nature and extent of the condition being treated, the site of administration, and the particular individual undergoing treatment, and that such optimums can be determined by conventional techniques. That is, an optimal dosing regimen for any particular patient, i.e., the number and frequency of doses, can be ascertained using conventional course of treatment determination tests. Generally, a dosing regimen herein involves administration of the topical formulation at least once daily, and preferably one to four times daily, until symptoms have subsided.

Topical formulations may also be used as chemopreventive compositions. When used in a chemopreventive method, susceptible skin is treated prior to any visible condition in a particular individual.

Compounds can also be delivered locally, e.g., to a tissue or organ within a subject, such as by injection, e.g., to extend the lifespan of the cells; protect against apoptosis or induce apoptosis.

In yet another embodiment, a compound is administered to a subject, such as to generally increase the lifespan of its cells and to protect its cells against stress and/or against apoptosis. It is believed that treating a subject with a compound described herein is similar to subjecting the subject to hormesis, i.e., mild stress that is beneficial to organisms and may extend their lifespan. For example, a compound can be taken by subjects as a food or dietary supplement. In one embodiment, such a compound is a component of a multi-vitamin complex. Compounds can also be added to existing formulations that are taken on a daily basis, e.g., statins and aspirin. Compounds may also be used as food additives.

Compounds described herein could also be taken as one component of a multi-drug complex or as a supplement in addition to a multi-drug regimen. In one embodiment, this multi-drug complex or regimen would include drugs or compounds for the treatment or prevention of aging-related diseases, e.g., stroke, heart disease, arthritis, high blood pressure, Alzheimer's. In another embodiment, this multi-drug regimen would include chemotherapeutic drugs for the treatment of cancer. In a specific embodiment, a polyphenol compound could be used to protect non-cancerous cells from the effects of chemotherapy.

Compounds may be administered to subject to prevent aging and aging-related consequences or diseases, such as stroke, heart disease, arthritis, high blood pressure, and Alzheimer's disease. Compounds described herein can also be administered to subjects for treatment of diseases, e.g., chronic diseases, associated with cell death, such as to protect the cells from cell death. Exemplary diseases include those associated with neural cell death or muscular cell death, such as Parkinson's disease, Alzheimer's disease, multiple sclerosis, amniotropic lateral sclerosis, and muscular dystrophy; AIDS; fulminant hepatitis; diseases linked to degeneration of the brain, such as Creutzfeld-Jakob disease, retinitis pigmentosa and cerebellar degeneration; myelodysplasis such as aplastic anemia; ischemic diseases such as myocardial infarction and stroke; hepatic diseases such as alcoholic hepatitis, hepatitis B and hepatitis C; joint-diseases such as osteoarthritis; atherosclerosis; alopecia; damage to the skin due to UV light; lichen planus; atrophy of the skin; cataract; graft rejections; and etc.

Compounds described herein can also be administered to a subject suffering from an acute disease, e.g., damage to an organ or tissue, e.g., a subject suffering from stroke or myocardial infarction or a subject suffering from a spinal cord injury. Compounds can also be used to repair an alcoholic's liver.

Compounds can also be administered to subjects who have recently received or are likely to receive a dose of radiation. In one embodiment, the dose of radiation is received as part of a work-related or medical procedure, e.g., working in a nuclear power plant, flying an airplane, an X-ray, CAT scan, or the administration of a radioactive dye for medical imaging; in such an embodiment, the compound is administered as a prophylactic measure. In another embodiment, the radiation exposure is received unintentionally, e.g., as a result of an industrial accident, terrorist act, or act of war involving radioactive material. In such a case, the compound is preferably administered as soon as possible after the exposure to inhibit apoptosis and the subsequent development of acute radiation syndrome.

Based at least on the discovery that certain concentrations of activating compounds prevent deacetylation of p53 in cells and thereby may induce apoptosis in cells, the activating compounds can also be administed to a subject in conditions in which apoptosis of certain cells is desired. For example, cancer may be treated or prevented. Exemplary cancers are those of the brain and kidney; hormone-dependent cancers including breast, prostate, testicular, and ovarian cancers; lymphomas, and leukemias. In cancers associated with solid tumors, a activating compound may be administered directly into the tumor. Cancer of blood cells, e.g., leukemia can be treated by administering a activating compound into the blood stream or into the bone marrow. Benign cell growth can also be treated, e.g., warts. Other diseases that can be treated include autoimmune diseases, e.g., systemic lupus erythematosus, scleroderma, and arthritis, in which autoimmune cells should be removed. Viral infections such as herpes, HIV, adenovirus, and HTLV-1 associated malignant and benign disorders can also be treated by administration of compounds. Alternatively, cells can be obtained from a subject, treated ex vivo to remove certain undesirable cells, e.g., cancer cells, and administered back to the same or a different subject.

In other embodiments, methods described herein are applied to yeast cells. Situations in which it may be desirable to extend the lifespan of yeast cells include any process in which yeast is used, e.g., the making of beer, yogurt, and bakery items, e.g., bread. Use of yeast having an extended lifespan can result in using less yeast or in having the yeast be active for longer periods of time. Yeast or other mammalian cells used for recombinantly producing proteins may also be treated as described herein.

Subjects that may be treated as described herein include eukaryotes, such as mammals, e.g., humans, ovines, bovines, equines, porcines, canines, felines, non-human primate, mice, and rats. Cells that may be treated include eukaryotic cells, e.g., from a subject described above, or plant cells, yeast cells and prokaryotic cells, e.g., bacterial cells. For example, activating compounds may be administered to form animals to improve their ability to withstand farming conditions longer.

Compounds may also be used to increase lifespan, stress resistance, and resistance to apoptosis in plants. In one embodiment, a compound is applied to plants, either on a periodic basis or in fungi. In another embodiment, plants are genetically modified to produce a compound. In another embodiment, plants and fruits are treated with a compound prior to picking and shipping to increase resistance to damage during shipping.

Compounds may also be used to increase lifespan, stress resistance and resistance to apoptosis in insects. In this embodiment, compounds would be applied to useful insects, e.g., bees and other insects that are involved in pollination of plants. In a specific embodiment, a compound would be applied to bees involved in the production of honey. Generally, the methods described herein may be applied to any organism, e.g., eukaryote, that may have commercial importance. For example, they can be applied to fish (aquaculture) and birds (e.g., chicken and fowl).

Higher doses of compounds may also be used as a pesticide by interfering with the regulation of silenced genes and the regulation of apoptosis during development. In this embodiment, a compound may be applied to plants using a method known in the art that ensures the compound is bio-available to insect larvae, and not to plants.

Activated sirtuin proteins that are in vitro outside of a cell may be used, e.g., for deacetylating target proteins, thereby, e.g., activating the target proteins. Activated sirtuins may be used, e.g., for the identification, in vitro, of previously unknown targets of sirtuin deacetylation, for example using 2D electrophoresis of acetyl labeled proteins.

At least in view of the link between reproduction and longevity (Longo and Finch, Science, 2002), the compounds can be applied to affect the reproduction of organisms such as insects, animals and microorganisms.

Inhibitory compounds may be used for similar purposes as high concentrations of activating compounds can be used for. For example, inhibitory compounds may be used to stimulate acetylation of substrates such as p53 and thereby increase apoptosis, as well as to reduce the lifespan of cells and organisms and/or rendering them more sensitive to stress.

Pharmaceutical Compositions and Methods

Pharmaceutical compositions for use in accordance with the present methods may be formulated in conventional manner using one or more physiologically acceptable carriers or excipients. Thus, activating compounds and their physiologically acceptable salts and solvates may be formulated for administration by, for example, injection, inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration. In one embodiment, the compound is administered locally, at the site where the target cells, e.g., diseased cells, are present, i.e., in the blood or in a joint.

Compounds can be formulated for a variety of loads of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remmington's Pharmaceutical Sciences, Meade Publishing Co., Easton, Pa. For systemic administration, injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous. For injection, the compounds can be formulated in liquid solutions, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution. In addition, the compounds may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included.

For oral administration, the pharmaceutical compositions may take the form of, for example, tablets, lozanges, or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., ationd oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate. Preparations for oral administration may be suitably formulated to give controlled release of the active compound.

For administration by inhalation, the compounds may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g., gelatin, for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.

The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.

The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.

In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.

Pharmaceutical compositions (including cosmetic preparations) may comprise from about 0.00001 to 100% such as from 0.001 to 10% or from 0.1% to 5% by weight of one or more compounds described herein.

In one embodiment, a compound described herein, is incorporated into a topical formulation containing a topical carrier that is generally suited to topical drug administration and comprising any such material known in the art. The topical carrier may be selected so as to provide the composition in the desired form, e.g., as an ointment, lotion, cream, microemulsion, gel, oil, solution, or the like, and may be comprised of a material of either naturally occurring or synthetic origin. It is preferable that the selected carrier not adversely affect the active agent or other components of the topical formulation. Examples of suitable topical carriers for use herein include water, alcohols and other nontoxic organic solvents, glycerin, mineral oil, silicone, petroleum jelly, lanolin, fatty acids, vegetable oils, parabens, waxes, and the like.

Formulations may be colorless, odorless ointments, lotions, creams, microemulsions and gels.

Compounds may be incorporated into ointments, which generally are semisolid preparations which are typically based on petrolatum or other petroleum derivatives. The specific ointment base to be used, as will be appreciated by those skilled in the art, is one that will provide for optimum drug delivery, and, preferably, will provide for other desired characteristics as well, e.g., emolliency or the like. As with other carriers or vehicles, an ointment base should be inert, stable, nonirritating and nonsensitizing. As explained in Remington's, cited in the preceding section, ointment bases may be grouped in four classes: oleaginous bases; emulsifiable bases; emulsion bases; and water-soluble bases. Oleaginous ointment bases include, for example, vegetable oils, fats obtained from animals, and semisolid hydrocarbons obtained from petroleum. Emulsifiable ointment bases, also known as absorbent ointment bases, contain little or no water and include, for example, hydroxystearin sulfate, anhydrous lanolin and hydrophilic petrolatum. Emulsion ointment bases are either water-in-oil (W/O) emulsions or oil-in-water (O/W) emulsions, and include, for example, cetyl alcohol, glyceryl monostearate, lanolin and stearic acid. Exemplary water-soluble ointment bases are prepared from polyethylene glycols (PEGs) of varying molecular weight; again, reference may be had to Remington's, supra, for further information.

Compounds may be incorporated into lotions, which generally are preparations to be applied to the skin surface without friction, and are typically liquid or semiliquid preparations in which solid particles, including the active agent, are present in a water or alcohol base. Lotions are usually suspensions of solids, and may comprise a liquid oily emulsion of the oil-in-water type. Lotions are preferred formulations for treating large body areas, because of the ease of applying a more fluid composition. It is generally necessary that the insoluble matter in a lotion be finely divided. Lotions will typically contain suspending agents to produce better dispersions as well as compounds useful for localizing and holding the active agent in contact with the skin, e.g., methylcellulose, sodium carboxymethylcellulose, or the like. An exemplary lotion formulation for use in conjunction with the present method contains propylene glycol mixed with a hydrophilic petrolatum such as that which may be obtained under the trademark Aquaphor® from Beiersdorf, Inc. (Norwalk, Conn.).

Compounds may be incorporated into creams, which generally are viscous liquid or semisolid emulsions, either oil-in-water or water-in-oil. Cream bases are water-washable, and contain an oil phase, an emulsifier and an aqueous phase. The oil phase is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation, as explained in Remington's, supra, is generally a nonionic, anionic, cationic or amphoteric surfactant.

Compounds may be incorporated into microemulsions, which generally are thermodynamically stable, isotropically clear dispersions of two immiscible liquids, such as oil and water, stabilized by an interfacial film of surfactant molecules (Encyclopedia of Pharmaceutical Technology (New York: Marcel Dekker, 1992), volume 9). For the preparation of microemulsions, surfactant (emulsifier), co-surfactant (co-emulsifier), an oil phase and a water phase are necessary. Suitable surfactants include any surfactants that are useful in the preparation of emulsions, e.g., emulsifiers that are typically used in the preparation of creams. The co-surfactant (or “co-emulsifer”) is generally selected from the group of polyglycerol derivatives, glycerol derivatives and fatty alcohols. Preferred emulsifier/co-emulsifier combinations are generally although not necessarily selected from the group consisting of: glyceryl monostearate and polyoxyethylene stearate; polyethylene glycol and ethylene glycol palmitostearate; and caprilic and capric triglycerides and oleoyl macrogolglycerides. The water phase includes not only water but also, typically, buffers, glucose, propylene glycol, polyethylene glycols, preferably lower molecular weight polyethylene glycols (e.g., PEG 300 and PEG 400), and/or glycerol, and the like, while the oil phase will generally comprise, for example, fatty acid esters, modified vegetable oils, silicone oils, mixtures of mono- di- and triglycerides, mono- and di-esters of PEG (e.g., oleoyl macrogol glycerides), etc.

Compounds may be incorporated into gel formulations, which generally are semisolid systems consisting of either suspensions made up of small inorganic particles (two-phase systems) or large organic molecules distributed substantially uniformly throughout a carrier liquid (single phase gels). Single phase gels can be made, for example, by combining the active agent, a carrier liquid and a suitable gelling agent such as tragacanth (at 2 to 5%), sodium alginate (at 2-10%), gelatin (at 2-15%), methylcellulose (at 3-5%), sodium carboxymethylcellulose (at 2-5%), carbomer (at 0.3-5%) or polyvinyl alcohol (at 10-20%) together and mixing until a characteristic semisolid product is produced. Other suitable gelling agents include methylhydroxycellulose, polyoxyethylene-polyoxypropylene, hydroxyethylcellulose and gelatin. Although gels commonly employ aqueous carrier liquid, alcohols and oils can be used as the carrier liquid as well.

Various additives, known to those skilled in the art, may be included in formulations, e.g., topical formulations. Examples of additives include, but are not limited to, solubilizers, skin permeation enhancers, opacifiers, preservatives (e.g., anti-oxidants), gelling agents, buffering agents, surfactants (particularly nonionic and amphoteric surfactants), emulsifiers, emollients, thickening agents, stabilizers, humectants, colorants, fragrance, and the like. Inclusion of solubilizers and/or skin permeation enhancers is particularly preferred, along with emulsifiers, emollients and preservatives. An optimum topical formulation comprises approximately: 2 wt. % to 60 wt. %, preferably 2 wt. % to 50 wt. %, solubilizer and/or skin permeation enhancer; 2 wt. % to 50 wt. %, preferably 2 wt. % to 20 wt. %, emulsifiers; 2 wt. % to 20 wt. % emollient; and 0.01 to 0.2 wt. % preservative, with the active agent and carrier (e.g., water) making of the remainder of the formulation.

A skin permeation enhancer serves to facilitate passage of therapeutic levels of active agent to pass through a reasonably sized area of unbroken skin. Suitable enhancers are well known in the art and include, for example: lower alkanols such as methanol ethanol and 2-propanol; alkyl methyl sulfoxides such as dimethylsulfoxide (DMSO), decylmethylsulfoxide (C.sub.10 MSO) and tetradecylmethyl sulfboxide; pyrrolidones such as 2-pyrrolidone, N-methyl-2-pyrrolidone and N-(-hydroxyethyl)pyrrolidone; urea; N,N-diethyl-m-toluamide; C.sub.2 -C.sub.6 alkanediols; miscellaneous solvents such as dimethyl formamide (DMF), N,N-dimethylacetamide (DMA) and tetrahydrofurfuryl alcohol; and the 1-substituted azacycloheptan-2-ones, particularly 1-n-dodecylcyclazacycloheptan-2-one (laurocapram; available under the trademark Azone® from Whitby Research Incorporated, Richmond, Va.).

Examples of solubilizers include, but are not limited to, the following: hydrophilic ethers such as diethylene glycol monoethyl ether (ethoxydiglycol, available commercially as Transcutol®) and diethylene glycol monoethyl ether oleate (available commercially as Softcutol®); polyethylene castor oil derivatives such as polyoxy 35 castor oil, polyoxy 40 hydrogenated castor oil, etc.; polyethylene glycol, particularly lower molecular weight polyethylene glycols such as PEG 300 and PEG 400, and polyethylene glycol derivatives such as PEG-8 caprylic/capric glycerides (available commercially as Labrasol®); alkyl methyl sulfoxides such as DMSO; pyrrolidones such as 2-pyrrolidone and N-methyl-2-pyrrolidone; and DMA. Many solubilizers can also act as absorption enhancers. A single solubilizer may be incorporated into the formulation, or a mixture of solubilizers may be incorporated therein.

Suitable emulsifiers and co-emulsifiers include, without limitation, those emulsifiers and co-emulsifiers described with respect to microemulsion formulations. Emollients include, for example, propylene glycol, glycerol, isopropyl myristate, polypropylene glycol-2 (PPG-2) myristyl ether propionate, and the like.

Other active agents may also be included in formulations, e.g., other anti-inflammatory agents, analgesics, antimicrobial agents, antifungal agents, antibiotics, vitamins, antioxidants, and sunblock agents commonly found in sunscreen formulations including, but not limited to, anthranilates, benzophenones (particularly benzophenone-3), camphor derivatives, cinnamates (e.g., octyl methoxycinnamate), dibenzoyl methanes (e.g., butyl methoxydibenzoyl methane), p-aminobenzoic acid (PABA) and derivatives thereof, and salicylates (e.g., octyl salicylate).

In certain topical formulations, the active agent is present in an amount in the range of approximately 0.25 wt. % to 75 wt. % of the formulation, preferably in the range of approximately 0.25 wt. % to 30 wt. % of the formulation, more preferably in the range of approximately 0.5 wt. % to 15 wt. % of the formulation, and most preferably in the range of approximately 1.0 wt. % to 10 wt. % of the formulation.

Topical skin treatment compositions can be packaged in a suitable container to suit its viscosity and intended use by the consumer. For example, a lotion or cream can be packaged in a bottle or a roll-ball applicator, or a propellant-driven aerosol device or a container fitted with a pump suitable for finger operation. When the composition is a cream, it can simply be stored in a non-deformable bottle or squeeze container, such as a tube or a lidded jar. The composition may also be included in capsules such as those described in U.S. Pat. No. 5,063,507. Accordingly, also provided are closed containers containing a cosmetically acceptable composition as herein defined.

In an alternative embodiment, a pharmaceutical formulation is provided for oral or parenteral administration, in which case the formulation may comprises an activating compound-containing microemulsion as described above, but may contain alternative pharmaceutically acceptable carriers, vehicles, additives, etc. particularly suited to oral or parenteral drug administration. Alternatively, an activating compound-containing microemulsion may be administered orally or parenterally substantially as described above, without modification.

Compounds described herein may be stored in oxygen free environment according to methods in the art. For example, resveratrol or analog thereof can be prepared in an airtight capusule for oral administration, such as Capsugel from Pfizer, Inc.

Cells, e.g., treated ex vivo with a compound described herein, can be administered according to methods for administering a graft to a subject, which may be accompanied, e.g., by administration of an immunosuppressant drug, e.g., cyclosporin A. For general principles in medicinal formulation, the reader is referred to Cell Therapy: Stem Cell Transplantation, Gene Therapy, and Cellular Immunotherapy, by G. Morstyn & W. Sheridan eds, Cambridge University Press, 1996; and Hematopoietic Stem Cell Therapy, E. D. Ball, J. Lister & P. Law, Churchill Livingstone, 2000.

Kits

Also provided herein are kits, e.g., kits for therapeutic purposes or kits for modulating the lifespan of cells or modulating apoptosis. A kit may comprise one or more activating or inhibitory compounds described herein, and optionally devices for contacting cells with the compounds. Devices include syringes, stents and other devices for introducing a compound into a subject or applying it to the skin of a subject.

The present description is further illustrated by the following examples, which should not be construed as limiting in any way. The contents of all cited references (including literature references, issued patents, published patent applications as cited throughout this application) are hereby expressly incorporated by reference.

The practice of the present methods will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, for example, Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989); DNA Cloning, Volumes I and II (D. N. Glover ed., 1985); Oligonucleotide Synthesis (M. J. Gait ed., 1984); Mullis et al. U.S. Pat. No. 4,683,195; Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. 1984); Transcription And Translation (B. D. Hames & S. J. Higgins eds. 1984); Culture Of Animal Cells (R. I. Freshney, Alan R. Liss, Inc., 1987); Immobilized Cells And Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide To Molecular Cloning (1984); the treatise, Methods In Enzymology (Academic Press, Inc., N.Y.); Gene Transfer Vectors For Mammalian Cells (J. H. Miller and M. P. Calos eds., 1987, Cold Spring Harbor Laboratory); Methods In Enzymology, Vols. 154 and 155 (Wu et al. eds.), Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds., Academic Press, London, 1987); Handbook Of Experimental Immunology, Volumes I-IV (D. M. Weir and C. C. Blackwell, eds., 1986); Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986).

EXAMPLES Example 1 Small Molecule Activators of SIRT1

To identify compounds that modulate SIRT1 activity, we screened a number of small molecule libraries using a fluorescent deacetylation assay in 96-well plates (Bitterman et al. J Biol Chem 277, 45099-107 (2002)). The substrate used in the assay was a fluorogenic peptide based on the sequence encompassing the p53-K382 acetylation site, a known target of SIRT1 in vivo (Vaziri et al. Cell 107, 149-59 (2001); Luo et al. Cell 107, 137-48 (2001); Langley et al. EMBO J 21, 2383-2396 (2002)). This substrate was preferred over a variety of other fluorogenic peptide substrates that were based on other known HDAC targets (FIG. 5). The small molecule libraries included analogues of nicotinamide, ε-acetyl lysine, NAD+, nucleotides, nucleotide analogues and purinergic ligands. From the initial screen, several sirtuin inhibitors were found (Supplementary Table 7). However, the most striking outcome was the identification of two compounds, quercetin and piceatannol, that stimulated SIRT1 activity five and eight-fold, respectively (Table 1). Both quercetin and piceatannol have been previously identified as protein kinase inhibitors (Glossmann et al. Naunyn Schmiedebergs Arch Pharmacol 317, 100-2 (1981); Oliver et al. J Biol Chem 269, 29697-703 (1994)).

Comparison of the structures of the two activating compounds suggested a possible structure-activity relationship. Piceatannol comprises two phenyl groups trans to one another across a linking ethylene moiety. The trans-stilbene ring structures of piceatannol are superimposable on the flavonoid A and B rings of quercetin, with the ether oxygen and carbon-2 of the C ring aligning with the ethylene carbons in piceatannol (see structures, Table 1). Further, the 5,7,3′ and 4′ hydroxyl group positions in quercetin can be aligned, respectively, with the 3,5,3′ and 4′ hydroxyls of piceatannol.

Given the demonstrated longevity-enhancing effects of sirtuin activity in S. cerevisiae( Kaeberlein et al. Genes Dev 13, 2570-80 (1999)) and C. elegans(Tissenbaum, H. A. and Guarente, L. Nature 410, 227-30. (2001)), it was naturally of interest to further explore the structure-activity relationship among compounds that stimulate SIRT1. Both quercetin and piceatannol are polyphenols, members of a large and diverse group of plant secondary metabolites that includes flavones, stilbenes, flavanones, isoflavones, catechins (flavan-3-ols), chalcones, tannins and anthocyanidins (Ferguson, L. R. Mutat Res 475, 89-111 (2001); Middleton et al. Pharmacol Rev 52, 673-751 (2000)). Polyphenols noteworthy with respect to potential longevity-enhancing effects include resveratrol, a stilbene found in red wine and epigallocatechin gallate (EGCG) from green tea. Both have been suggested on the basis of epidemiological and mechanistic investigations to exert cancer chemopreventive and cardioprotective effects (Ferguson, L. R. Mutat Res 475, 89-111 (2001); Middleton et al. Pharmacol Rev 52, 673-751 (2000); and Jang et al. Science 275, 218-20 (1997)). We therefore performed a secondary screen encompassing resveratrol, EGCG and additional representatives from a number of the polyphenol classes listed above. The screen emphasized flavones due to the great number of hydroxyl position variants available in this group (Middleton et al. Pharmacol Rev 52, 673-751 (2000). The results of this screen are summarized in Supplementary Tables 1-6. In the tables, a “ratio to control rate” above 1 indicates that a compound with such a rate is an activator of the sirtuin tested and a number under 1 indicates that a compound is an inhibitor.

Additional potent SIRT1 activators were found among the stilbenes, chalcones and flavones (Table 1, Supplementary Tables 1 and 2). The six most active flavones had 3′ and 4′ hydroxyls (Supplementary Table 2), although it should be noted that the most active compound overall, resveratrol (3,5,4′-trihydroxystilbene), was more active than piceatannol, which differs only by its additional 3′-hydroxyl (Table 1). The importance of the 4′-hydroxyl to activity is underscored by the fact that each of the 12 most stimulatory flavones share this feature (Supplementary Tables 1 and 2).

Many, but not all of the most active compounds include hydroxyls in the two meta positions (e.g. 5,7-dihydroxylated flavones) of the ring (A ring), trans to that with the 4′ or 3′,4′ pattern (B ring, see Table 1, Supplementary Tables 1 and 2). A potentially coplanar orientation of the trans phenyl rings may be important for activity since catechins and flavanones, which lack the 2,3 double-bond, have weak activity despite having equivalent hydroxylation patterns to various stimulatory flavones (compare Supplementary Tables 2 and 3 with 4 and 5). The absence of activity in the isoflavone genistein, although hydroxylated in an equivalent way to the stimulatory compounds apigenin and resveratrol (see Supplementary Tables 1, 2 and 4), is consistent with the idea that the trans positioning and spacing of the hydroxylated rings contributes strongly to activity.

The biological effects of polyphenols are frequently attributed to antioxidant, metal ion chelating and/or free-radical scavenging activity (Ferguson, L. R. Mutat Res 475, 89-111 (2001); Jang et al. Science 275, 218-20 (1997)). We considered the possibility that the apparent polyphenol stimulation of SIRT1 might simply represent the repair of oxidative and/or metal-ion induced damage incurred during preparation of the recombinant protein. Two features of our results argue against this being the case. First, a variety of free-radical protective compounds, including antioxidants, chelators and radical scavengers, failed to stimulate SIRT1 (see Supplementary Table 6.). Second, among various polyphenols of equivalent antioxidant capacity we observed diverse SIRT1 stimulating activity (e.g. compare resveratrol, quercetin and the epicatechins in Supplementary Tables 1, 2 and 5 and see Stojanovic et al. Arch Biochem Biophys 391, 79-89 (2001)).

Example 2 Resveratrol's Effects on SIRT1 Kinetics

Detailed enzyme kinetic investigations were performed using the most potent activator, resveratrol. Dose-response experiments performed under the conditions of the polyphenol screening assays (25 μM NAD+, 25 μM p53-382 acetylated peptide), showed that the activating effect doubled the rate at ˜11 μM and was essentially saturated at 100 μM resveratrol (FIG. 1a). Initial enzyme rates, in the presence or absence of 100 μM resveratrol, were determined either as a function of acetyl-peptide concentration with high NAD+ (3 mM NAD+, FIG. 1b) or as a function of NAD+ concentration with high acetyl-peptide (1 mM p53-382 acetylated peptide, FIG. 1c). Although resveratrol had no significant effect on the two Vmax determinations (FIGS. 1b, 1c), it had pronounced effects on the two apparent Kms. Its effect on the acetylated peptide Km was particularly striking, amounting to a 35-fold decrease (FIG. 1b). Resveratrol also lowered the Km for NAD+ over 5-fold (FIG. 1c). Since resveratrol acts only on Km, it could be classified as an allosteric effector of ‘K system’ type (Monod et al. J. Mol. Biol. 12, 88-118 (1965)). This can imply that only the substrate binding affinity of the enzyme has been altered, rather than a rate-limiting catalytic step.

Our previous kinetic analysis of SIRT1 and Sir2 (Bitterman et al. J Biol Chem 277, 45099-107 (2002)) and our genetic analysis of Sir2's role in yeast lifespan extension (Anderson et al. Nature 423, 181-5 (2003); Anderson et al. J Biol Chem 277, 18881-90. (2002)) have implicated nicotinamide (a product of the sirtuin reaction) as a physiologically important inhibitor of sirtuin activity. Therefore the effects of resveratrol on nicotinamide inhibition were tested. In experiments similar to those of FIGS. 1b and 1c, kinetic constants in the presence of 50 μM nicotinamide were determined either by varying the concentration of NAD+ or that of the p53-382 acetylated peptide (FIG. 1d). Nicotinamide, in contrast to resveratrol, affects the SIRT1 Vmax (note 30% and 36% Vmax decreases in absence of resveratrol, FIG. 1d and see Bitterman et al. J Biol Chem 277, 45099-107 (2002)). In the presence of 50 μM nicotinamide, resveratrol appears to have complex, concentration-dependent effects on the kinetics of SIRT1 (FIG. 1d). Apparent Km for NAD+ and acetylated substrate appear to actually be raised by 5 μM resveratrol when nicotinamide is present. At 20 and 100 μM, in the presence of 50 μM nicotinamide, resveratrol lowers the Km for both NAD+ and acetylated peptide, without reversing the nicotinamide-induced Vmax decrease. It has been proposed that sirtuins may bind nicotinamide at a second site, known as “the C pocket”, distinct from the “B” site that interacts with the nicotinamide moiety of NAD+ (Bitterman et al. J Biol Chem 277, 45099-107 (2002)). In light of this potential complexity, further kinetic studies, supplemented by structural/crystallographic information, will likely be necessary to fully elucidate the interplay between the effects of nicotinamide and polyphenols.

Example 3 Activating Compounds Extend Yeast Lifespan

To investigate whether these compounds could stimulate sirtuins in vivo, we utilized S. cerevisiae, an organism in which the upstream regulators and downstream targets of Sir2 are relatively well understood. A resveratrol dose-response study of Sir2 deacetylation rates (FIG. 2a) indeed reveals that resveratrol stimulates Sir2 in vitro, with the optimum concentration of activator being 2-5 μM. Levels of activation were somewhat lower than those for SIRT1, and unlike SIRT1, inhibition was seen at concentrations greater than ˜100 μM.

Resveratrol and four other potent sirtuin activators, representatives of the stilbene, flavone, and chalcone families, were tested for their effect on yeast lifespan. Due to the potential impediment by the yeast cell wall or plasma membrane and suspected slow oxidation of the compound in the medium, we chose to use a concentration (10 μM) slightly higher than the optimal resveratrol concentration in vitro. As shown in FIG. 2b, quercetin and piceatannol had no significant effect on lifespan. In contrast, butein, fisetin and resveratrol increased average lifespan by 31, 55 and 70%, respectively, and all three significantly increased maximum lifespan (FIG. 2c). Concentrations of resveratrol higher than 10 μM provided no added lifespan benefit and there was no lasting effect of the compound on the lifespan of pre-treated young cells (FIG. 2d).

For subsequent yeast genetic experiments we focused on resveratrol because it was the most potent SIRT1 activator and provided the greatest lifespan extension. Glucose restriction, a form of CR in yeast, resulted in no significant extension of the long-lived resveratrol-treated cells (FIG. 3a), indicating that resveratrol likely acts via the same pathway as CR. Consistent with this, resveratrol had no effect on the lifespan of a sir2 null mutant (FIG. 3b). Given that resveratrol is reported to have fungicidal properties at high concentrations (Pont, V. and Pezet, R. J Phytopathol 130, 1-8 (1990)), and that mild stress can extend yeast lifespan by activating PNC1 (Anderson et al. Nature 423, 181-5 (2003)), it was plausible that resveratrol was extending lifespan by inducing PNC1, rather than acting on Sir2 directly. However, resveratrol extended the lifespan of a pnc1 null mutant nearly as well as it did wild type cells (FIG. 3b). Together these data show that resveratrol acts downstream of PNC1 and requires SIR2 for its effect. Thus, the simplest explanation for our observations is that resveratrol increases lifespan by directly stimulating Sir2 activity.

A major cause of yeast aging is thought to stem from the inherent instability of the repetitive rDNA locus (Sinclair, D. A. Mech Ageing Dev 123, 857-67 (2002); Lin et al. Science 289, 2126-8 (2000); Sinclair, D. A. and Guarente, L. Cell 91, 1033-42 (1997); Defossez et al. Mol Cell 3, 447-55 (1999); Park et al. Mol Cell Biol 19, 3848-56 (1999)). Homologous recombination between rDNA repeats can generate an extrachromosomal circular form of rDNA (ERC) that is replicated until it reaches toxic levels in old cells. Sir2 is thought to extend lifespan by suppressing recombination at the replication fork barrier of rDNA( Benguria et al. Nucleic Acids Res 31, 893-8 (2003)). Consistent with the lifespan extension we observed for resveratrol, this compound reduced the frequency of rDNA recombination by ˜60% (FIG. 3c), in a SIR2-dependent manner (FIG. 3d). In the presence of the Sir2 inhibitor nicotinamide, recombination was also decreased by resveratrol (FIG. 3c), in agreement with the kinetic data (see FIG. 1d). Interestingly, we found that resveratrol and other sirtuin activators had only minor effects on rDNA silencing (FIG. 3e and f).

Another measure of lifespan in S. cerevisiae is the length of time cells can survive in a metabolically active but nutrient deprived state. Aging under these conditions (i.e. chronological aging) is primarily due to oxidative damage (Longo, V. D. and Finch, C. E. Science 299, 1342-6 (2003)). Resveratrol (10 μM or 100 μM) failed to extend chronological lifespan (not shown), indicating that the sirtuin-stimulatory effect of resveratrol may be more relevant in vivo than its antioxidant activity (Ferguson, L. R. Mutat Res 475, 89-111 (2001); Middleton et al. Pharmacol Rev 52, 673-751 (2000)).

Example 4 Effects of Activators in Human Cells

To test whether these compounds could stimulate human SIRT1 in vivo, we first employed a cellular deacetylase assay that we had developed. A schematic of the assay procedure is depicted in FIG. 4a. Cells are incubated with media containing the fluorogenic ε-acetyl-lysine substrate, ‘Fluor de Lys’ (FdL). This substrate, neutral when acetylated, becomes positively charged upon deacetylation and accumulates within cells (see FIG. 6a). Lysis of the cells and addition of the non-cell-permeable ‘Developer’ reagent releases a fluorophor specifically from those substrate molecules that have been deacetylated (FIG. 4a and see Methods). With HeLa cells growing adherently, 5-10% of the signal produced in this assay is insensitive to 1 μM trichostatin A (TSA), a potent inhibitor of class I and II HDACs but not sirtuins (class III) (Denu, J. M. Trends Biochem Sci 28, 41-8 (2003)). (FIGS. 6b and 6c).

A selection of SIRT1-stimulatory and non-stimulatory polyphenols were tested for their effects on this TSA-insensitive signal (FIG. 4b). Cellular deacetylation signals in the presence of each compound (y-axis, FIG. 4b) were plotted against their fold-stimulations of SIRT1 in vitro (x-axis, FIG. 4b, data from Supplementary Tables 1-3). For most of the compounds, the in vitro activity roughly corresponded to the cellular signal. Compounds with little or no in vitro activity clustered around the negative control (Group A, FIG. 4b). Another grouping, of strong in vitro activators is clearly distanced from the low activity cluster in both dimensions (Group B, FIG. 4b). A notable outlier was butein, a potent activator of SIRT1 in vitro which had no effect on the cellular signal. With allowances for possible variation among these compounds in properties unrelated to direct sirtuin stimulation, such as cell-permeability and rates of metabolism, these data are consistent with the idea that certain polyphenols can activate native sirtuins in vivo.

One known target of SIRT1 in vivo is lysine 382 of p53. Deacetylation of this residue by SIRT1 decreases the activity and half-life of p53 (Vaziri et al. Cell 107, 149-59 (2001); Luo et al. Cell 107, 137-48. (2001); Langley et al. EMBO J 21, 2383-2396 (2002)). To follow the acetylation status of K382 we generated a rabbit polyclonal antibody that recognizes the acetylated form of K382 (Ac-K382) on Western blots of whole cell lysates. As a control we showed that the signal was specifically detected in extracts from cells exposed to ionizing radiation (FIG. 4c), but not in extracts from cells lacking p53 or where arginine had been substituted for lysine 382 (data not shown). U2OS osteosarcoma cells were pre-treated for 4 hours with resveratrol (0.5 and 50 μM) and exposed to UV radiation. We consistently observed a marked decrease in the level of Ac-K382 in the presence of 0.5 μM resveratrol, compared to untreated cells (FIG. 4d). At higher concentrations of resveratrol (>50 μM) the effect was reversed (FIG. 4d and data not shown), consistent with previous reports of increased p53 activity at such concentrations (Dong, Z. Mutat Res 523-524, 145-50 (2003)). The ability of low concentrations of resveratrol to promote deacetylation of p53 was diminished in cells expressing a dominant-negative SIRT1 allele (H363Y) (FIG. 4e), demonstrating that SIRT1 is necessary for this effect. This biphasic dose-response of resveratrol could explain the dichotomy in the literature regarding the effects of resveratrol on cell survival (Ferguson, L. R. Mutat Res 475, 89-111 (2001); Dong, Z. Mutat Res 523-524, 145-50 (2003); Nicolini et al. Neurosci Lett 302, 41-4 (2001)).

Thus, we have discovered the first known class of small molecule sirtuin activators, all of which are plant polyphenols. These compounds can dramatically stimulate sirtuin activity in vitro and promote effects consistent with increased sirtuin activity in vivo. In human cells, resveratrol promotes SIRT1-mediated p53 deacetylation of K382. In yeast, the effect of resveratrol on lifespan is as great as any longevity-promoting genetic manipulation (Anderson et al. Nature 423, 181-5 (2003)) and has been linked convincingly to the direct activation of Sir2. The correlation between lifespan and rDNA recombination, but not silencing, adds to the body of evidence that yeast aging is due to DNA instability (Sinclair, D. A. Mech Ageing Dev 123, 857-67 (2002); Lin et al. Science 289, 2126-8 (2000); Sinclair, D. A. and Guarente, L. Cell 91, 1033-42. (1997); Defossez et al. Mol Cell 3, 447-55 (1999); Park et al. Mol Cell Biol 19, 3848-56 (1999)) not gene dysregulation (Jazwinski, S. M. Ann N Y Acad Sci 908, 21-30 (2000)).

Sirtuins have been found in diverse eukaryotes, including fungi, protozoans, metazoans and plants (Pandey et al. Nucleic Acids Res 30, 5036-55 (2002); Frye, R. A. Biochem Biophys Res Commun 273, 793-8 (2000)), and likely evolved early in life's history (Kenyon, C. A conserved regulatory mechanism for aging. Cell 105, 165-168 (2001)). Plants are known to produce a variety of polyphenols, including resveratrol, in response to stresses such as dehydration, nutrient deprivation, UV radiation and pathogens (Soleas et al. Clin Biochem 30, 91-113 (1997); Coronado et al. Plant Physiol 108, 533-542 (1995)). Therefore it is believed that these compounds may be synthesized to regulate a sirtuin-mediated plant stress response. This would be consistent with the recently discovered relationship between environmental stress and Sir2 activity in yeast (Anderson et al. Nature 423, 181-5 (2003)). Perhaps these compounds have stimulatory activity on sirtuins from fungi and animals because they mimic an endogenous activator, as is the case for the opiates/endorphins, cannabinols/endocannabinoids and various polyphenols with estrogen-like activity (Ferguson, L. R. Mutat Res 475, 89-111 (2001); Middleton et al. Pharmacol Rev 52, 673-751 (2000)). Alternatively, animal and fungal sirtuins may have retained or developed an ability to respond to these plant metabolites because they are a useful indicator of a deteriorating environment and/or food supply.

Example 5 Materials and Methods for Examples 1-4

Compound Libraries and Deacetylation Assays

His6-tagged recombinant SIRT1 and GST-tagged recombinant Sir2 were prepared as described by (Bitterman et al. J Biol Chem 277, 45099-107. (2002). From 0.1 to 1 μg of SIRT1 and 1.5 μg of Sir2 were used per deacetylation assay (in 50 μl total reaction). SIRT1 assays and certain of those for Sir2 employed the p53-382 acetylated substrate (‘Fluor de Lys-SIRT1′, BIOMOL) rather than FdL.

Themed compound libraries (BIOMOL) were used for primary and secondary screening. Most polyphenol compounds were dissolved at 10 mM in dimethylsulfoxide (DMSO) on the day of the assay. For water soluble compounds and negative controls, 1% v/v DMSO was added to the assay. In vitro fluorescence assay results were read in white ½-volume 96-well microplates (Corning Costar 3693) with a CytoFluor™II fluorescence plate reader (PerSeptive Biosystems, Ex. 360 nm, Em. 460 nm, gain=85). HeLa cells were grown and the cellular deacetylation assays were performed and read, as above, but in full-volume 96-well microplates (Corning Costar 3595). Unless otherwise indicated all initial rate measurements were means of three or more replicates, obtained with single incubation times, at which point 5% or less of the substrate initially present had been deacetylated. Calculation of net fluorescence increases included subtraction of a blank value, which in the case of Sir2 was obtained by omitting the enzyme from the reaction and in the case of SIRT1 by adding an inhibitor (200 μM suramin or 1 mM nicotinamide) to the reaction prior to the acetylated substrate. A number of the polyphenols partially quenched the fluorescence produced in the assay and correction factors were obtained by determining the fluorescence increase due to a 3 μM spike of an FdL deacetylated standard (BIOMOL, catalog number KI-142). All error bars represent the standard error of the mean.

Media and Strains

All yeast strains were grown at 30° C. in complete yeast extract/bactopeptone, 2.0% (w/v) glucose (YPD) medium except where stated otherwise. Calorie restriction was induced in 0.5% glucose. Synthetic complete (SC) medium consisted of 1.67% yeast nitrogen base, 2% glucose, 40 mg/liter each of auxotrophic markers. SIR2 was integrated in extra copy and disrupted as described by Lin et al. (Science 289, 2126-8 (2000)). Other strains are described elsewhere (Bitterman et al. J Biol Chem 277, 45099-107 (2002)). For cellular deacetylation assays, HeLa S3 cells were used. U2OS osteosarcoma and human embryonic kidney (HEK 293) cells were cultured adherently in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% fetal calf serum (FCS) with 1.0% glutamine and 1.0% penecillin/streptomycin. HEK 293 overexpressing dominant negative SIRT1 H363Y was a gift of R. Frye (U. Pittsburgh).

Lifespan Determinations

Lifespan measurements were performed using PSY316AT MATα as previously described by Anderson et al. (J Biol Chem 277, 18881-90. (2002). All compounds for lifespan analyses were dissolved in 95% ethanol and plates were dried and used within 24 hours. Prior to lifespan analysis, cells were pre-incubated on their respective media for at least 15 hours. Following transfer to a new plate, cells were equilibrated on the medium for a minimum of 4 hours prior to micro-manipulating them. At least 30 cells were examined per experiment and each experiment was performed at least twice. Statistical significance of lifespan differences was determined using the Wilcoxon rank sum test. Differences are stated to be significant when the confidence is higher than 95%.

Silencing and Recombination Assays

Ribosomal DNA silencing assays using the URA3 reporters were performed as previously described by Bitterman et al. (J Biol Chem 277, 45099-107 (2002)). Ribosomal DNA recombination frequencies were determined by plating W303AR cells (Sinclair, D. A. and Guarente, L. Cell 91, 1033-42 (1997)) on YPD medium with low adenine/histidine and counting the fraction of half-red sectored colonies using Bio-Rad Quantity One software as described by Anderson et al. (J Biol Chem 277, 18881-90. (2002)). At least 6000 cells were analyzed per experiment and all experiments were performed in triplicate. All strains were pre-grown for 15 hours with the relevant compound prior to plating.

Proteins and Western Analyses

Recombinant Sir2-GST was expressed and purified from E. coli as previously described except that lysates were prepared using sonication (Bitterman et al. J Biol Chem 277, 45099-107 (2002). Recombinant SIRT1 from E. coli was prepared as previously described (Bitterman et al. J Biol Chem 277, 45099-107 (2002). Polyclonal antiserum against p53-AcK382 was generated using an acetylated peptide antigen as previously described (Vaziri et al. Cell 107, 149-59 (2001) with the following modifications. Anti-Ac-K382 antibody was affinity purified using non-acetylated p53-K382 peptides and stored in PBS at −70° C. and recognized an acetylated but not a non-acetylated p53 peptide. Western hybridizations using anti-acetylated K382 or anti-actin (Chemicon) antibody were performed at 1:1000 dilution of antibody. Hybridizations with polyclonal p53 antibody (Santa Cruz Biotech.) used 1:500 dilution of antibody. Whole cell extracts were prepared by lysing cells in buffer containing 150 mM NaCl, 1 mM MgCl2, 10% glycerol, 1% NP40, 1 mM DTT and anti-protease cocktail (Roche).

Example 6 Localization of the Activation Domain of Sirtuins to their N-Terminus

Yeast Sir2 and human SIRT1 are very homologous and differ from human SIRT2 by the addition of an N-terminal domain that is absent in SIRT2. The effect of resveratrol was assayed on human recombinant SIRT2 as follows. Human recombinant SIRT2 was incubated at a concentration of 1.25 μg/well with 25 μM of Fluor de Lys-SIRT2 (BIOMOL cat. # KI-179) and 25 μM NAD+ for 20 minutes at 37° C., as described above. Results, indicate that, in contrast to SIRT1, increasing concentrations of resveratrol decrease SIRT2 activity. Thus, based on the difference in structure of SIRT1 and SIRT2, i.e., the absence of an N-terminal domain, it is believed that the N-terminal domain of SIRT1 and Sir2 is necessary for activation by the compounds described herein. In particular, it is likely that the activator compounds described herein interact with the N-terminal portion of sirtuins. The N-terminal portion of SIRT1 that is necessary for the action of the compounds is from about amino acid 1 to about amino acid 176, and that of Sir2 is from about amino acid 1 to about amino acid 175.

Example 7 Resveratrol Extends the Lifespan of C. elegans

50 C. elegans worms (strain N2) were grown in the presence or absence of 100 μM resveratrol for 17 days. On day 17, only 5 worms in the control group without resveratrol were alive, whereas 17 worms were alive in the group that was treated with resveratrol. Thus, the presence of resveratrol in the growth media of C. elegans extends their lifespan.

Example 8 Identification of Additional Activators of Sirtuins

Using the screening assay described in Example 1, five more sirtuin activators have been identified. These are set forth in supplementary Table 8.

Example 9 Identification of Inhibitors of Sirtuins

Using the screening assay described in Example 1, more inhibitors were identified. These are set forth in the appended supplementary Table 8, and correspond to the compounds having a ratio to control rate of less than 1.

Example 10 Identification of Further Activators and Inhibitors of Sirtuins

Additional activators and inhibitors of sirtuins were identified, and are listed in Tables 9-13. In these Tables, “SE” stands for Standard error of the mean and N is the number of replicates used to calculate mean ratio to the control rate and standard error.

All SIRT1 rate measurements used in the calculation of “Ratio to Control Rate” were obtained with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described above and in K. T. Howitz et al. Nature (2003) 425: 191. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Stability determinations (t1/2) derived from SIRT1 rate measurements performed in a similar way to those described above, except that 5 μM p53-382 acetylated peptide substrate was used rather than 25 μM. The fold-stimulation (ratio to control) obtained with a compound diluted from an aged stock solution was compared to an identical dilution from a stock solution freshly prepared from the solid compound. “t1/2” is defined as the time required for the SIRT1 fold-stimulation of the compound from the aged solution to decay to one-half of that obtained from a freshly prepared solution. Ethanol stocks of resveratrol, BML-212 and BML-221 were prepared at 2.5 mM and the compounds were assayed at a final concentration of 50 μM. The water stock of resveratrol was 100 μM and the assay performed at 10 μM. Stocks were aged by storage at room 5 temperature, in glass vials, under a nitrogen atmosphere.

The effect of some of these compounds on lifespan was determined in yeast, C. elegans and D. melanogaster, as described above. The results are set forth below in Table A:

% change in yeast % change in C. elegans % change in D. replicative lifespan lifespan relative to melanogaster lifespan relative to untreated untreated organisms relative to untreated Compound organisms (10 μM)a (100/500 μM)b (100 μM)c untreated 100% 100% 100% Resveratrol 170-180% 110% 130% 3,5,4′-Trihydroxy-trans-stilbene (from M. Tatar) Pinosylvin 114% ? ? 3,5-Dihydroxy-trans-stilbene BML-212  98% ? ? 3,5-Dihydroxy-4′-fluoro-trans-stilbene BML-217  90% ? ? 3,5-Dihydroxy-4′-chloro-trans-stilbene BML-221 165% >100% (ongoing) ? 3,4′-Dihydroxy-5-acetoxy-trans-stilbene BML-233 ? 70% (10)  ? 3,5-Dihydroxy-4′-methoxy-trans-stilbene 50% (500)
aReplicative lifespans performed using 2% (w/v) glucose standard yeast compete medium (YPD) under standard conditions.

bLifespan assays performed on N2 worms using E. coli as food under standard conditions.

cLifespan assays preformed using 1.5% yeast as food supply under otherwise standard conditions.

The results indicate that resveratrol significantly extends lifespan in yeast, C. elegans and in D. melanogaster. Since BML-233 was shown to be a strong activator of 15 sirtuins (see above), the results obtained in C. elegans may indicate that the compound is toxic to the cells.

Without wanting to be limited to particular structures, it appears that the following structure/activity relationships exist. SIRT1 activation results from several of these new analogs confirmed the importance of planarity, or at least the potential for planarity, between and within the two rings of the active compounds. Reduction of the double bond of the ethylene function, between, the two rings essentially abolishes activity (compare Resveratrol, Table A and Dihydroresveratrol, Table E). Replacement of a phenyl moiety with a cyclohexyl group is nearly as detrimental to SIRT1 stimulating activity (compare Pinosylvin, Table 9 and BML-224, Table 12). Amide bonds are thought to have a partially double bond character. However, replacement of the ethylene function with a carboxamide abolished activity (compare Pinosylvin, Table 9, with BML-219, Table 13). It is possible that this effect could be due in part to the position that carbonyl oxygen must assume in the conformation that places the two rings trans to one another. If so, a compound in which the positions of the amide nitrogen and carbonyl are reversed might be expected to have greater activity.

In twelve of the analogs resveratrol's 4′-hydroxy was replaced with various functionalities (see Tables 9 and 10, BML-221 in Table 11, BML-222 in Table 12). Although none of the replacements tried led to substantial increases in SIRT1 stimulating activity, this parameter was, in general, remarkably tolerant of substitutions at this position. Small groups (H— in Pinosylvin, Cl— in BML-217, —CH3 in BML-228) did the least to decrease activity. There is some evidence of a preference in the enzyme's stilbene binding/activation site for unbranched (ethyl in BML-225, azido in BML-232, —SCH3 in BML-230) and hydrophobic functions (compare isopropyl in BML-231 to acetoxy in BML-221, acetamide in BML-222). Solution stability relative to resveratrol was strongly increased by one of the two 4′-substitutions (acetoxy, BML-221) tested for this so far.

Resveratrol is currently the most potent known activator of SIRT1. The collection of analogs described above, particularly the group entailing substitutions at the 4′ position, may be instrumental in informing the design of new SIRT1 ligands with improved pharmacological properties. One parameter that may be of interest in this regard is stability. One 4′-substituted analog, BML-221, displays a vast improvement in solution stability relative to resveratrol and although diminished in vitro SIRT1 activating ability, retains much of resveratrol's biological activity (see lifespan data). The 4′-hydroxyl of resveratrol is thought to be of primary importance to resveratrol's free-radical scavenging reactivity (S. Stojanovic et al. Arch. Biochem. Biophys. 2001 391 79). Most of the 4′-substituted analogs have yet to be tested for solution stability, but if resveratrol's instability in solution is due to redox reactivity, many of the other analogs would be expected to also exhibit improved stability.

The results obtained with 4′-substituted analogs may indicate promising routes to explore while seeking to increase SIRT1 binding affinity. For example, the efficacy of the 4′-ethyl compound (BML-225) might indicate the presence of a narrow, hydrophobic binding pocket at the SIRT1 site corresponding to the 4′ end of resveratrol. Several new series of 4′-substituted analogs are planned, the simplest comprising straight-chain aliphatic groups of various lengths.

Example 11 Methods of Synthesis of the Compounds in Tables 9-13

Most of the resveratrol analogs were synthesized by the same general procedure, from a pair of intermediates, a benzylphosphonate and an aldehyde. The synthesis or sources of these intermediates are described in section II. Section III. describes the procedures for synthesizing the final compounds from any of the benzylphosphonate/aldehyde pairs. The coupling reaction (Section III. A.) is followed by one of two deprotection reactions depending on whether the intermediates contained methoxymethyl (Section III. B.) or methoxy (Section III. C.) protecting groups. Section IV corresponds to Tables 14-18, which list the particular benzylphosphonate and aldehyde used in the synthesis of particular final compounds. Seven of the compounds—Resveratrol, 3,5-Dihydroxy-4′-methoxy-trans-stilbene, Rhapontin aglycone, BML-227, BML-221, Dihydroresveratrol, BML-219—were not synthesized by the general procedure and “N/A” appears next to their entries in the table. Resveratrol was from BIOMOL and the syntheses of the remaining compounds are described in Section V.

II. Synthetic Intermediates

A. Benzylphosphonates (Synthesized)

Synthesis of Diethyl 4-Acetamidobenzylphosphonate: To diethyl 4-aminobenzylphosphonate in 1:1 methylene chloride/pyridine was added catalytic DMAP and acetic anhydride (1.1 eq.). After 3 hours, the reaction was evaporated to dryness and purified via flash chromatography (silica gel).

Synthesis of Diethyl 4-Methylthiobenzylphosphonate: 4-Methylthiobenzyl chloride was heated with triethylphosphite (as solvent) at 120° C. overnight. Excess triethyl phosphite was distilled off under high vacuum and heat. Flash chromatography (silica gel) yielded the desired product.

Synthesis of Diethyl 3,5-Dimethoxybenzylphosphonate: From 3-5-Dimethoxybenzyl bromide. See synthesis of Diethyl 4-Methylthiobenzylphosphonate.

Synthesis of Diethyl 4-Fluorobenzylphosphonate: From 4-Fluorobenzylphosphonate. See synthesis of Diethyl 4-Methylthiobenzylphosphonate.

Synthesis of Diethyl 4-azidobenzylphosphonate: To diethyl 4-aminobenzylphosphonate in acetonitrile (2.5 mL) at 0° C. was added 6M HCl (1 mL). Sodium nitrite (1.12 eq.) in water (1 mL) was added drop wise and the resulting solution stirred at 0° C. for 30 mins. Sodium azide (8 eq.) in water (1 mL) added drop wise (bubbling) and the solution stirred at 0° C. for 30 mins., then at room temperature for 1 hour. The reaction was diluted with ethyl acetate and washed with water and brine and dried over sodium sulfate. Flash chromatography (silica gel) yielded the desired product.

B. Aldehydes (Synthesized)

Synthesis of 3,5-Dimethoxymethoxybenzaldehyde: To 3,5-dihydroxybenzaldehyde in DMF at 0° C. was added sodium hydride (2.2 eq.). The reaction was stirred for 30 min. at 0° C. Chloromethylmethyl ether (2.2 eq.) was added neat, drop wise and the reaction allowed to warm to room temperature over 1.5 hrs. The reaction mixture was diluted with diethyl ether and washed with water (2×) and brine (1×) and dried over sodium sulfate. Flash chromatography (silica gel) yielded the desired product.

C. Purchased Intermediates: Unless Listed Above, all Synthetic Intermediates were Purchase from Sigma-Aldrich.

III. General Procedure for the Synthesis of Resveratrol Analogues

A. Benzylphosphonate/Aldehyde Coupling Procedure

To the appropriate benzylphosphonate (1.2 eq.) in dimethylformamide (DMF) at room temperature was added sodium methoxide (1.2 eq.). This solution was allowed to stir at room temperature for approximately 45 minutes. The appropriate aldehyde (1 eq.) was then added (neat or in a solution of dimethylformamide). The resulting solution was then allowed to stir overnight at room temperature. Thin layer chromatography (TLC) was used to determine completeness of the reaction. If the reaction was not complete, the solution was heated at 45-50° C. until complete. The reaction mixture was poured into water and extracted with ethyl acetate (2×). The combined organic layers were washed with brine and dried over sodium sulfate. Flash chromatography (silica gel) yielded the desired products.

B. General Procedure for the Deprotection of Methoxymethylresveratrol Analogues

To the appropriate methoxymethylstilbene derivative in methanol was added two drops of concentrated HCl. The resulting solution was heated overnight at 50° C. The solution was evaporated to dryness upon completion of the reaction. Flash chromatography (silica gel) yielded the desired product.

C. General Procedure for the Deprotection of Methoxyresveratrol Analogues

To the appropriate methoxystilbene derivative in methylene chloride was added tetrabutylammonium iodide (1.95 eq. per methoxy group). The reaction was cooled to 0° C. and boron trichloride (1 M in methylene chloride; 2 eq. per methoxy group) was added dropwise. Following the addition of boron trichloride, the cooling bath was removed and the reaction allowed to stir at room temperature until complete (as indicated by TLC). Saturated sodium bicarbonate solution was added and the reaction vigorously stirred for 1 hour. The reaction was poured into cold 1M HCl and extracted with ethyl acetate (3×). The combined organic layers were washed with water (1×) and brine (1×) and dried over sodium sulfate. Flash chromatography (silica gel) yielded the desired products.

V. Special Syntheses

Synthesis of BML-219 (N-(3,5-Dihydroxyphenyl)benzamide): To benzoyl chloride (1 eq.) in dry methylene chloride at room temperature was added triethylamine (1.5 eq.) and a catalytic amount of DMAP followed by 3,5-dimethoxyaniline (1 eq.). The reaction was allowed to stir overnight at room temperature. Upon completion, the reaction was diluted with ethyl acetate and washed with 1M HCl, water and brine and dried over sodium sulfate. Flash chromatography (silica gel) yielded the methoxystilbene derivative. To the methoxystilbene in dry methylene chloride at 0° C. was added tetrabutylammonium iodide (3.95 eq.) followed by boron trichloride (4 eq.; 1M in methylene chloride). Upon completion of the reaction (TLC), saturated sodium bicarbonate was added and the mixture was vigorously stirred for 1 hour. The reaction was diluted with ethyl acetate and washed with 1M HCl and brine and dried over sodium sulfate. Flash chromatography (silica gel) yielded the desired product.

Synthesis of BML-220 (3,3′,5-trihydroxy-4′-methoxystilbene): To Rhapontin in methanol was added catalytic p-toluenesulfonic acid. The reaction was refluxed overnight. Upon completion of the reaction (TLC), the reaction mixture was evaporated to dryness and taken up in ethyl acetate. The organics were washed with water and brine and dried over sodium sulfate. Flash chromatography (silica gel) yielded the desired product.

Synthesis of BML-233 (3,5-Dihydroxy-4′-methoxystilbene): To deoxyrhapontin in methanol was added catalytic p-toluenesulfonic acid. The reaction was refluxed overnight. Upon completion of the reaction (TLC), the reaction mixture was evaporated to dryness and taken up in ethyl acetate. The organics were washed with water and brine and dried over sodium sulfate. Flash chromatography (silica gel) yielded the desired product.

Synthesis of BML-221 and 227 (4′ and 3 monoacetylresveratrols): To resveratrol in tetrahydrofuran at room temperature was added pyridine (1 eq.) followed by acetic anhydride (1 eq.). After stirring for 48 hrs., another 0.25 eq. acetic anhydride added followed by 24 hrs. of stirring. The reaction was diluted with methylene chloride (reaction was not complete) and washed with cold 0.5M HCl, water and brine. Organics were dried over sodium sulfate. Flash chromatography yielded a mixture of 4′- and 3-acetyl resveratrols. Preparative HPLC yielded both monoacetyl resveratrols.

Synthesis of Dihydroresveratrol: To resveratrol in argon-purged ethyl acetate in a Parr shaker was added 10% palladium on carbon (10 wt %). The mixture was shaken under an atmosphere of hydrogen (30 psi) for 5 hours. Filtration through a pad of celite yielded the desired material.

Equivalents

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments described herein. Such equivalents are intended to be encompassed by the following claims.

TABLE 1 APPENDIX OF TABLES: Stimulation of SIRT1 Catalytic Rate by Plant Polyphenols (100 μM). Ratio to Control Rate Compound Mean ± SE Structure Resveratrol (3,5,4′-Trihydroxy-trans-stilbene) 13.4 ± 1.0  Butein (3,4,2′,4′-Tetrahydroxychalcone) 8.53 ± 0.89 Piceatannol (3,5,3′,4′-Tetrahydroxy-trans-stilbene) 7.90 ± 0.50 Isoliquiritigenin (4,2′,4′-Trihydroxychalcone) 7.57 ± 0.84 Fisetin (3,7,3′,4′-Tetrahydroxyflavone) 6.58 ± 0.69 Quercetin (3,5,7,3′,4′-Pentahydroxyflavone) 4.59 ± 0.47
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Supplementary Table 1. Effects of Stilbenes and Chalcones (100 μM) on SIRT1 Rate. Ratio to Control Rate Compound Mean ± SE Replicates Structure Skeleton Resveratrol (3,5,4′-Trihydroxy- trans-stilbene) Piceatannol # (3,5,3,′4′- Tetrahydroxy-trans-stilbene) Deoxyrhapontin (3,5-Dihydroxy-4′-methoxystilbene 3-O-β-D-glucoside) trans-Stilbene Rhapontin 3,3′,5-Trihydroxy-4′-methoxystilbene 3-O-β-D-glucoside cis-Stilbene 13.4 ± # 1.0   7.90 ± 0.50  1.94 ± 0.21   1.48 ± 0.15  1.40 ± 0.37−  1.14 ± 0.29 10   7  6   6 6   6 Butein (3,4,2′,4′- Tetrahydroxychalcone) 4,2′,4′- Trihydroxychalcone 3,4,2′,4′,6′- Pentahydroxychalcone Chalcone 8.53 ± 0.89  7.57 ± 0.84  2.80 ± 0.32  1.34 ± 0.17 6  6  6  6
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All radio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Supplementary Table 2. Effects of Flavones (100 μM) on SIRT1 Rate (Part I). Ratio to Control Rate Compound Mean ± SE Replicates Structure Skeleton Fisetin (3,7,3′,4′- Tetrahydroxyflavone) 5,7,3′4′,5′- Pentahydroxyflavone Luteolin (5,7,3′,4′- Tetrahydroxyflavone) 3,6,3′,4′- Tetrahydroxyflavone Quercetin (3,5,7,3′,4′- Pentahydroxyflavone) 7,3′,4′,5′- Tetrahydroxyflavone 6.58 ± # 0.69   6.05 ± 0.98  5.66 ± 0.80   5.45 ± 0.57  4.59 ± 0.47   3.62 ± 0.56 9   6  6   12  16   6 Kaempferol 3.55 ± 0.56 6 (3,5,7,4′- Tetrahydroxyflavone) 6-Hydroxyapigenin 3.06 ± 0.29 6 (5,6,7,4′- Tetrahydroxyflavone; Scutellarein) Apigenin 2.77 ± 0.40 6 (5,7,4′- Trihydroxyflavone) 3,6,2′,4′- 2.10 ± 0.22 6 Tetrahydroxyflavone 7,4′-Dihydroxyflavone 1.91 ± 0.17 6
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Supplementary Table 3. Effects of Flavones (100 μM) on SIRT1 Rate (Part II). Ratio to Control Rate Compound Mean ± SE Replicates Structure Skeleton 7,8,3,4′- Tetrahydroxyflavone 3,6,2′,3′- Tetrahydroxyflavone 4′-Hydroxyflavone 5,4′-Dihydroxyflavone 5,7-Dihydroxyflavone Morin (3,5,7,2′,4′- Pentahydroxyflavone) Flavone 1.91 ± # 0.39  1.74 ± 0.27  1.73 ± 0.12 1.56 ± 0.15 1.51 ± 0.18 1.461 ± 0.071   1.41 ± 0.23 6  6  6 6 6 6   6 5-Hydroxyflavone 1.22 ± 0.19 6 Myricetin 0.898 ± 0.070 12  (Cannabiscetin; 3,5,7,3′,4′,5′- Hexahydroxyflavone) 3,7,3′,4′,5′- 0.826 ± 0.074 12  Pentahydroxyflavone Gossypetin 0.723 ± 0.062 6 (3,5,7,8,3′,4′- Hexahydroxyflavone)
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Supplementary Table 4. Effects of isoflavones, Flavanones and Anthocyanidins (100 μM) on SIRT1 Rate Ratio to Control Rate Compound Mean ± SE Replicates Structure Skeleton Daidzein (7,4′-Dihydroxyisoflavone) (5,7,4′- Trihydroxyisoflavone) 2.28 ± 0.74  1.109 ± 0.026 2  2 Naringenin (5,7,4′- Trihydroxyflavanone) 3,5,7,3′,4′- Pentahydroxyflavanone Flavanone 2.10 ± 0.23   1.97 ± 0.22  1.92 ± 0.24 6   5  6 Pelargonidin chloride (3,5,7.4′- Tetrahydroxyflavylium chloride) Cyanidin chloride (3,5,7,3′,4′- Pentahydroxyflavylium chloride) Delphinidin chloride (3,5,7,3′,4′,5′- Hexahydroxyflavylium chloride) 1.586 ± # 0.037    0.451 ± 0.015    0.4473 ± 0.0071 2    2    2
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Supplementary Table 5. Effects of Catechins (Flavan-3-ols) (100 μM) on SIRT1 Rate. Ratio to Control Rate Compound Mean ± SE Replicates Structure Skeleton/Structure (−)-Epicatechin (Hydroxy Sites: 3,5,7,3′,4′) (−)-Catechin (Hydroxy Sites: 3,5,7,3′,4′) (−)-Gallocatechin (Hydroxy Sites: 3,5,7,3′,4′,5′) (+)-Catechin (Hydroxy Sites: 3,5,7,3′,4′) (+)-Epicatechin (Hydroxy Sites: 3,5,7,3′,4′) (−)-Epigallocatechin (Hydroxy Sites: 3,5,7,3′,4′,5′) 1.53 ± # 0.31  1.41 ± 0.21  1.35 ± 0.25  1.31 ± 0.19  1.26 ± 0.20  0.41 ± 0.11 4  4  4  4  4  4 (−)-Epigallocatechin Gallate (Hydroxy Sites: 3*,5,7,3′,4′,5′; *Position of gallate ester) 0.32 ± 0.12 4
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Supplementary Table 6. Effects of Free Radical Protective Compounds (100 μM) on SIRT1 Rate. Ratio to Control Rate Protective Compound Mean ± SE Replicates Mechanism Hinokitiol 2.48 ± 0.15 2 Iron Chelatol (b-Thujaplicin; 2- hydroxy-4-isopropyl-2,4,6- cycloheptatrien-1-one) L-(+)-Ergothioneine 2.06 ± 0.48 2 Antioxidant, ((S)-a-Carboxy-2,3- Peroxynitrite dihydro-N,N,N-trimethyl-2- Scavenger thioxo-1H-imidazole- 4-ethanaminium inner salt) Caffeic Acid Phenyl Ester 1.80 ± 0.16 2 Iron Chelator MCI-186 1.2513 ± 0.0080 2 Radical (3-Methyl-1-phenyl-2- Scavenger pyrazolin-5-one) and Antioxidant HBED 1.150 ± 0.090 2 Iron Chelator (N,N′-Di-(2-hydroxy- benzyl)ethylenediamine- N,N′-diacetic acid.HCl.H2O) Ambroxol  1.075 ± 0.0026 2 Radical (trans-4-(2-Amino-3,5- Scavenger dibromobenzylamino) cyclohexane.HCl) U-83836E 1.030 ± 0.055 2 “Lazaroid” ((−)-2-((4-(2,6-di-1- amino- Pyrrolidinyl-4-pyrimidinyl)- steroid, 1-piperazinyl)methyl)-3,4- Peroxidation dihydro-2,5,7,8- inhibitor tetramethyl-2H-1- benzopyran-6-ol-2HCl) Trolox 0.995 ± 0.019 2 Antioxidant (6-Hydroxy-2,5,7,8- tetramethylchroman-2- carboxylic acid)
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Supplementary Table 7. Effects of Miscellaneous Compounds (100 μM) on SIRT1 Catalytic Rate. Ratio to Control Rate Mean ± Repli- Compound SE cates Structure & Activities Dipyridamole (2,6-bis- (Diethano- lamino)- 4,8-di- piperidino- pyrimido[5,4- d]pyrimidine) 3.54 ±0.20 2 Inhibitor of Adenosine Transport, Phosphodiesterase, 5-Lipoxygenase Nicotinamide 0.428 ± 42 0.019 Sirtuin Reaction Product/Inhibitor NF279 0.0035 ± 3 0.0011 Purinergic Receptor Antagonist NF023 −0.0016 ± 3 0.0015 G-protein Antagonist Suramin −0.0002 ± 3 0.0010 G-protein Antagonist, Reverse Transcriptase Inhibitor
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

Supplementary Table 8. Effects of Various Modulators on SIRT1 Rate. Ratio to Compound, Control Rate (Concentration) Mean ± SE Replicates Structure ZM 336372, (100 μM) 3.5 ± 1.1 3 Camptothecin, (10 μM) 2.92 ± 0.41 3 Coumestrol, (10 μM) 2.30 ± 0.31 2 NDGA, (100 μM) 1.738 ± 0.088 3 Esculetin, (10 μM) 1.737 ± 0.082 3 Sphingosine 0.069 ± 0.028 3
Abbreviation: SE, Standard error of the mean. Rate measurements with 25 μM NAD+ and 25 μM p53-382 acetylated peptide substrate were performed as described in Methods. All ratio data were calculated from experiments in which the total deacetylation in the control reaction was 0.25-1.25 μM peptide or 1-5% of the initial concentration of acetylated peptide.

TABLE 9 SIRT1 Rate Effects of New Resveratrol Analogs (100 μM). Ratio to Stability in Control Rate Solution Compound Mean ± SE N Structure t1/2, hrs. BML-217 (3,5-Dihydroxy- 4′-chloro-trans- stilbene) 10.6 ± 0.4  3 Resveratrol (3,5,4′- Trihydroxy-trans- stilbene) 10.4 ± 0.5  43 59 (ethanol), 20 (water) Pinosylvin (3,5-Dihydroxy- trans-stilbene) 9.95 ± 0.45 3 BML-225 (3,5-Dihydroxy- 4′-ethyl-trans- stilbene) 9.373 ± 0.014 3 BML-212 (3,5-Dihydroxy- 4′-fluoro-trans- stilbene) 8.20 ± 0.69 3 66 (ethanol) BML-228 (3,5-Dihydroxy- 4′-methyl-trans- stilbene) 7.72 ± 0.12 3

TABLE 10 SIRT1 Rate Effects of New Resveratrol Analogs (100 μM). Ratio to Stability in Control Rate Solution Compound Mean ± SE N Structure t1/2, hrs. BML-232 (3,5-Dihydroxy- 4′-azido-trans- stilbene) 7.24 ± 0.12 3 BML-230 (3,5-Dihydroxy- 4′-thiomethyl- trans-stilbene) 6.84 ± 1.26 6 BML-229 (3,5-Dihydroxy- 4′-nitro-trans- stilbene) 6.78 ± 0.22 3 BML-231 (3,5-Dihydroxy- 4′-isopropyl- trans-stilbene) 6.01 ± 0.15 3 BML-233 3,5-Dihydroxy-4′- methoxy-trans- stilbene 5.48 ± 0.33 6

TABLE II SIRT1 Rate Effects of New Resveratrol Analogs (100 μM). Ratio to Stability in Control Rate Solution Compound Mean ± SE N Structure t1/2, hrs. Rhapontin aglycone (3,5,3′Trihydroxy- 4′-methoxy-trans- stilbene) 4.060 ± 0.069 3 BML-227 (3,4′-Dihydroxy-5- acetoxy-trans- stilbene) 3.340 ± 0.093 3 BML-221 (3,5-Dihydroxy-4′- acetoxy-trans- stilbene) 3.05 ± 0.54 6 504 (ethanol) BML-218 (E)-1-(3,5- Dihydroxyphenyl)- 2-(2-napthyl) ethene 3.05 ± 0.37 6 3-Hydroxystilbene 2.357 ± 0.074 3

TABLE 12 SIRT1 Rate Effects of New Resveratrol Analogs (100 μM). Ratio to Stability in Control Rate Solution Compound Mean ± SE N Structure t1/2, hrs. BML-226 (3,5-Dimethoxymethoxy- 4′-thiomethyl-trans- stilbene) 2.316 ± 0.087 3 BML-222 (3,5-Dihydroxy-4′- acetamide-trans- stilbene) 1.88 ± 0.11 3 3,4-Dihydroxy- trans-stilbene 1.64 ± 0.10 6 BML-224 (E)-1-(3,5- Dihydroxyphenyl)- 2-(cyclohexyl) ethene 1.297 ± 0.042 3 3,4-Dimethoxy- trans-stilbene 1.127 ± 0.019 3

TABLE 13 SIRT1 Rate Effects of New Resveratrol Analogs (100 μM). Ratio to Stability in Control Rate Solution Compound Mean ± SE N Structure t1/2, hrs. Dihydroresveratrol (1-(3,5-Dihydroxyphenyl)- 2-(4-hydroxyphenyl) ethane) 1.08 ± 0.14 4 4-Hydroxy-trans- stilbene 0.943 ± 0.039 3 BML-219 N-phenyl-(3,5- dihydroxy)benzamide 0.902 ± 0.014 3 3,5-Dihydroxy-4′- nitro-trans-stilbene 0.870 ± 0.019 3 4-Methoxy-trans-stilbene 0.840 ± 0.089 3

TABLE 14 Resveratrol Analog Synthetic Intermediates Compound Benzylphosphonate Aldehyde Structure BML-217 (3,5-Dihydroxy- 4′-chloro-trans- stilbene) Diethyl 3-5- dimethoxybenzyl phosphonate 4-Chlorobenzaldehyde Resveratrol (3,5,4′- Trihydroxy-trans- stilbene) N/A N/A Pinosylvin (3,5-Dihydroxy- trans-stilbene) Diethyl benzyl phosphonate 3,5-Dimethoxy benzaldehyde BML-225 (3,5-Dihydroxy- 4′-ethyl-trans- stilbene) Diethyl 3-5- dimethoxybenzyl phosphonate 4-Ethylbenzaldehyde BML-212 (3,5-Dihydroxy- 4′-fluoro-trans- stilbene) Diethyl 4-fluoro benzylphosphonate 3,5-Dimethoxy benzaldehyde BML-228 (3,5-Dihydroxy- 4′-methyl-trans- stilbene) Diethyl 3-5- dimethoxybenzyl phosphonate 4-Methylbenzaldehyde

TABLE 15 Resveratrol Analog Synthetic Intermediates Compound Benzylphosphonate Aldehyde Structure BML-232 (3,5-Dihydroxy- 4′-azido-trans- stilbene) Diethyl 4-azido benzylphosphonate 3,5-Dimethoxymethoxy benzaldehyde BML-230 (3,5-Dihydroxy- 4′-thiomethyl- trans-stilbene) Diethyl 4-methylthio benzylphosphonate 3,5-Dimethoxymethoxy benzaldehyde BML-229 (3,5-Dihydroxy- 4-nitro-trans- stilbene) Diethyl 3-5- dimethoxybenzyl phosphonate 4-Nitrobenzaldehyde BML-231 (3,5-Dihydroxy- 4′-isopropyl- trans-stilbene) Diethyl 3-5- dimethoxybenzyl phosphonate 4-Isopropyl benzaldehyde 3,5-Dihydroxy- 4′-methoxy- trans-stilbene N/A N/A

TABLE 16 Resveratrol Analog Synthetic Intermediates Compound Benzylphosphonate Aldehyde Structure Rhapontin aglycone (3,5,3′Trihydroxy- 4′-methoxy-trans- stilbene) N/A N/A BML-227 (3,4′-Dihydroxy-5- acetoxy-trans- stilbene) N/A N/A BML-221 (3,5-Dihydroxy-4′- acetoxy-trans- stilbene) N/A N/A BML-218 (E)-1-(3,5- Dihydroxyphenyl)- 2-(2-napthyl) ethene Diethyl 3-5- dimethoxybenzyl phosphonate 2-Naphthaldehyde BML-216 3-Hydroxystilbene Benzylphosphonate 3-Methoxy benzaldehyde

TABLE 17 Resveratrol Analog Synthetic Intermediates Compound Benzylphosphonate Aldehyde Structure BML-226 (3,5-Dimethoxymethoxy- 4′-thiomethyl- trans-stilbene) Diethyl 4-methylthio benzylphosphonate 3,5dimethoxymethoxy benzaldehyde BML-222 (3,5-Dihydroxy-4′- acetamide-trans- stilbene) Diethyl 4-acetamido benzylphosphonate 3,5-dimethoxymethoxy benzaldehyde BML-215 3,4-Dihydroxy- trans-stilbene Benzylphosphonate 3,4-Dimethoxy benzaldehyde BML-224 (E)-1-(3,5- Dihydroxyphenyl)- 2-(cyclohexyl) ethene 3,5-Dimethoxy benzylphosphonate Cyclohexane carboxaldehyde 3,4-Dimethoxy- trans-stilbene Benzylphosphonate 3,4-Dimethoxy benzaldehyde

TABLE 18 Resveratrol Analog Synthetic Intermediates Compound Benzylphosphonate Aldehyde Structure Dihydroresveratrol (1-(3,5-Dihydroxyphenyl)- 2-(4-hydroxyphenyl) ethane) N/A N/A BML-214 4-Hydroxy-trans- stilbene Benzylphosphonate 4-Methoxy benzaldehyde BML-219 N-phenyl-(3,5- dihydroxy)benzamide N/A N/A 3,5-Dihydroxy-4-nitro- trans-stilbene 3,5-Dimethoxy benzylphosphonate 4-Nitrobenzaldehdye 4-Methoxy-trans- stilbene Benzylphosphonate 4-Methoxy benzaldehyde

Claims

1. A method for activating a sirtuin deacetylase protein family member, comprising contacting the sirtuin deacetylase protein family member with an activating compound having a formula selected from the group consisting of formulas 1-25 and 30.

2. The method of claim 1, wherein the compound is a polyphenol compound or analog or derivative thereof.

3. The method of claim 1, wherein the plant compound is selected from the group consisting of flavones, stilbenes, flavanones, isoflavones, catechins, chalcones, tannins and anthocyanidins or analog or derivative thereof.

4. The method of claim 1, wherein, if the compound is a naturally occurring compound, it is not in a form in which it is naturally occurring.

5. The method of claim 3, wherein the compound is selected from the group consisting of resveratrol, butein, piceatannol, isoliquiritgenin, fisetin, luteolin, 3,6,3′,4′-tetrahydroxyflavone, quercetin, and analogs and derivatives thereof.

6. The method of claim 1, wherein the sirtuin deacetylase protein family member is SIRT1.

7. The method of claim 1, wherein the sirtuin deacetylase protein family member is in a cell, and the method comprises contacting the cell with the compound.

8. The method of claim 7, wherein the cell is in vitro.

9. The method of claim 7, wherein the cell is a cell of a subject.

10. The method of claim 7, wherein the cell is in a subject and the method comprises administering the compound to the subject.

11. The method of claim 1, further comprising determining the activity of the sirtuin deacetylase protein family member.

12. The method of claim 10, further comprising determining the activity of the sirtuin deacetylase protein family member.

13. The method of claim 7, wherein the cell is contacted with the compound at a concentration of 0.1-100 μM.

14. The method of claim 1, further contacting the cell with an additional activating compound having a formula selected from the group consisting of formulas 1-25 and 30.

15. The method of claim 14, comprising contacting the cell with a least three different activating compounds having a formula selected from the group consisting of formulas 1-25 and 30.

16. A method for inhibiting the activity of a sirtuin protein family member, comprising contacting the sirtuin deacetylase protein family member with an inhibiting compound having a formula selected from the group consisting of formulas 26-29 and 31.

17. A method for shortening the lifespan of a cell or rendering it resistant to stress, comprising contacting the cell with an inhibiting compound having a formula selected from the group consisting of formulas 26-29 and 31.

18. A composition comprising two compounds each having a formula selected from the group of formulas 1-31.

19. A method for identifying a compound that modulates SIRT1 in vivo, comprising (i) contacting a cell comprising a SIRT1 protein with a peptide of p53 comprising an acetylated residue 382 in the presence of an inhibitor of class I and class II HDAC under conditions appropriate for SIRT1 to deacetylate the peptide and (ii) determining the level of acetylation of the peptide, wherein a different level of acetylation of the peptide in the presence of the test compound relative to the absence of the test compound indicates that the test compound modulates SIRT1 in vivo.

Patent History
Publication number: 20060084135
Type: Application
Filed: Jul 1, 2004
Publication Date: Apr 20, 2006
Inventors: Konrad Howitz (Allentown, PA), Robert Zipkin (Wynnewood, PA)
Application Number: 10/884,062
Classifications
Current U.S. Class: 435/29.000
International Classification: C12Q 1/02 (20060101);