EP1590674A2 - Screening for modulators of fat storage - Google Patents

Screening for modulators of fat storage

Info

Publication number
EP1590674A2
EP1590674A2 EP04707568A EP04707568A EP1590674A2 EP 1590674 A2 EP1590674 A2 EP 1590674A2 EP 04707568 A EP04707568 A EP 04707568A EP 04707568 A EP04707568 A EP 04707568A EP 1590674 A2 EP1590674 A2 EP 1590674A2
Authority
EP
European Patent Office
Prior art keywords
rip
target protein
compound
reporter gene
expression
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP04707568A
Other languages
German (de)
English (en)
French (fr)
Inventor
Malcolm George Parker
Roger Imperial College Innovations Ltd WHITE
Goran Leonardsson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ip2ipo Innovations Ltd
Original Assignee
Imperial College Innovations Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Imperial College Innovations Ltd filed Critical Imperial College Innovations Ltd
Publication of EP1590674A2 publication Critical patent/EP1590674A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6875Nucleoproteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • This invention relates to screening methods for identifying compounds that are useful as modulators of fat storage.
  • the invention provides methods of screening for compounds that modulate the function of receptor interacting protein 140 (RIP 140).
  • Obesity is associated with diseases such as diabetes, heart disease and hypertension while anorexia can cause irreversible bone damage and ultimately death.
  • an object of the invention is the provision of a method for identifying compounds that modulate fat storage.
  • the nuclear receptor interacting protein 140 is a co-activator or co-repressor of transcription by a variety of nuclear receptor family members including the estrogen receptors (ER), such as estrogen receptor ⁇ (ER ⁇ ) and estrogen related receptor ⁇ (ERR ⁇ ), retinoic acid receptors (RAR), thyroid hormone receptors (TR), retinoid X receptors (RXR), vitamin D receptor (NDR) and peroxisome proliferator activated receptors (PPAR ⁇ , PPAR ⁇ (also known as PPAR ⁇ ) and PPAR ⁇ ), as well as by the aryl hydrocarbon receptor (AhR).
  • ER estrogen receptor ⁇
  • ERP ⁇ estrogen related receptor ⁇
  • RAR retinoic acid receptors
  • TR thyroid hormone receptors
  • RXR retinoid X receptors
  • NDR peroxisome proliferator activated receptors
  • PPAR ⁇ , PPAR ⁇ also known as PPAR ⁇
  • PPAR ⁇ peroxisome proliferator activated receptors
  • RIP 140 is known to be essential for female fertility.
  • Female mice null for RIP 140 are viable but infertile because of complete failure to release oocytes at ovulation (White et al, 2000, Nature Medicine, 6:1368-1374).
  • RIP 140 is thought to have a secondary role in maintenance of pregnancy (Leonardsson et al, 2002, Endocrinology, 143(2): 700-707).
  • RIP 140 plays a role in controlling fat storage. Mice devoid of the corepressor protein RIP 140 are leaner than wild-type mice and fat in food-deprived RIP 140 null mice appears to be deposited in muscle cells in greater amounts than in food- deprived wild-type mice. RIP 140 null mice are resistant to high-fat diet induced obesity and hepatic steatosis. Fat accumulation in white adipose tissue (WAT) is decreased in RIP 140 null mice compared to wild-type mice.
  • WAT white adipose tissue
  • genes that are expressed only in brown adipose tissue (BAT) in wild-type mice are to be expressed in the WAT of mice null for RIP 140.
  • BAT can be used as an energy source as a result of uncoupling.
  • Genes involved in fat metabolism, energy dissipation and mitochondrial uncoupling are markedly induced in the WAT of RIP 140 null mice, resulting in an increase in oxygen consumption.
  • UCP1 uncoupling protein 1
  • UCP1 uncoupling protein 1
  • the RIP 140 corepressor prevents mitochondrial uncoupling in the WAT of wild-type mice and the leanness of mice null for RIP 140 results from an ability to use WAT as an energy source.
  • Adipogenesis appears to be unaffected in RIP 140 null mice but it is also possible that the leanness of RIP 140 null mice results from RIP 140 having a role in adipocyte function which is inhibited in the null mice.
  • RIP 140 is a co-regulator which exerts its effects by binding to nuclear receptor family members such as ER, RAR, TR, RXR, NDR or PPAR or transcription factors such as AhR, SF-1 and DAX-1. These proteins to which RIP 140 specifically binds are referred to herein as target proteins.
  • Target proteins These proteins to which RIP 140 specifically binds are referred to herein as target proteins.
  • Compounds may modulate the activity of RIP 140 by promoting or inhibiting its ability to form a complex with one or more of these target proteins.
  • Compounds that modulate the ability of RIP 140 to form a complex with one or more of these target proteins may act to decrease or increase fat storage.
  • Compounds that bind to RIP 140 may promote or inhibit its ability to form a complex with a target protein.
  • compounds that bind to a target protein of RIP 140 may promote or inhibit the ability of RIP 140 to form a complex with that target protein.
  • RIP 140 is shown herein to target a regulatory region in the promoter of the UCP-1 gene which is one of the genes upregulated in RIP 140 null mice. This regulatory region contains binding sites for PPAR, TR, RXR and RAR, suggesting that RIP 140 may act as a co-repressor of one or more of these target proteins. Compounds that modulate the ability of RIP 140 to form a complex with one or more of PPAR, TR, RXR and RAR may act to decrease or increase fat storage. Compounds that bind to RIP 140 may promote or inhibit its ability to form a complex with PPAR, TR, RXR and RAR.
  • the invention therefore provides a method of screening for compounds that bind to RIP 140, said method comprising assessing binding of a candidate compound to RIP 140.
  • the invention further provides a method of screening for compounds that bind to a target protein of RIP 140, said method comprising assessing binding of a candidate compound to the target protein.
  • the target protein of RIP 140 is preferably selected from AhR, ER, RAR, TR, RXR, NDR, PPAR, SF-1 and DAX-1.
  • the target protein of RIP 140 is selected from PPAR, TR, RXR and RAR.
  • the target protein of RIP 140 is selected from PPAR ⁇ , PPAR ⁇ and PPAR ⁇ .
  • PPAR ⁇ these three nuclear receptors have already been shown to have a role in fat metabolism.
  • PPAR ⁇ has been shown to be involved in thermogenesis and fatty acid oxidation (Kelly, D.P, 2003, Circ Res, 92: 482-4; Peters, J.M. et al, 1997, JBiol Chem, 272: 27307-12; Barbera, M.J. et al, 2001, JBiol Chem, 276: 1486-93) and PPAR ⁇ has been implicated in lipid homeostasis (Wang, Y.X. et al, 2003, Cell 113: 159- 70).
  • the invention provides a method of screening for compounds that bind to a complex of RIP 140 and a target protein, said method comprising assessing binding of a candidate compound to a complex of RIP 140 and a target protein.
  • the invention further provides a third method of screening for compounds that modulate the formation or maintenance of a complex of RIP 140 and a target protein.
  • a modulator may be either an agonist or an antagnosist of the RIP140:target protein interaction.
  • An agonist is a compound that promotes the formation and/or maintenance of a complex of RIP 140 and a target protein.
  • An antagonist is a compound that inhibits the formation and/or maintenance of a complex of RIP 140 and a target protein.
  • the method of screening for compounds that act to modulate the formation or maintenance of a complex of RIP 140 and a target protein comprises the steps of:
  • the mixing of RIP 140, the target protein and candidate compound in step (a) may be done in any order.
  • the candidate compounds used in the third method may be compounds already identified as binding to RIP 140 or a target protein by the first method of the invention, or compounds identified as binding to a complex of RIP 140 and a target protein by the second method of the invention.
  • a candidate compound has been identified in vitro as a compound that binds to RIP 140 or to a target protein, as a compound that binds to a complex of RIP 140 and a target protein, or as a modulator of the interaction between a target protein and RIP 140, it may be desirable to perform further experiments to confirm the in vivo function of the compound in modulating fat storage. Any of the above methods may therefore comprise the further steps of administering to a mammal a candidate compound and assessing its effect on fat storage.
  • the invention also provides a method of assessing the in vivo effect on fat storage of a compound obtained or obtainable by any of the methods described above, comprising administering the compound to a mammal and assessing the effect on fat storage.
  • the invention provides a method for screening for a compound that modulate fat storage in a mammal, comprising a first step of identifying a compound which:
  • step (a) binds to RIP 140 or to a RIP 140 target protein; (b) binds to a complex of RIP 140 and a RIP 140 target protein; or (c) modulates the binding interaction between RIP 140 and a RIP 140 target protein, and comprising a second step of administering to a mammal a candidate compound identified in step (a), (b) or (c) and assessing its effect on fat storage in the mammal.
  • the mammal may be any species of mammal including humans but is preferably a monkey, pig, rabbit, guinea pig, rat or mouse. Tests on non-humans may be preferred.
  • the compound identified by a method of the invention may be administered to animal models of obesity and leanness.
  • the compound may be administered to an animal in which a gene or combination of genes known to be involved in fat storage has been knocked-out.
  • the compound may be administered to an animal that is null for RIP 140 or an animal that is null for the ob gene.
  • Other models are widely available (e.g. Pomp (1999) Molecular Medicine Today 5:459-460).
  • the methods of screening for compounds that bind to RIP 140 or to a target protein, or that bind to a complex of RIP 140 and a target protein, or that modulate the interaction between RIP 140 and a target protein may take place in a different geographical location from the method for assessing the effect of the compounds identified on fat storage.
  • the RIP 140 or target protein used in the method of screening for compounds that bind RIP 140 or a target protein may be free in solution, affixed to a solid support, located on a cell surface or located intracellularly.
  • the binding of a candidate compound to RIP 140 or to a target protein is detected by means of a label directly or indirectly associated with the candidate compound.
  • the label may be a fluorophore, radioisotope, or other detectable label.
  • one or both of the candidate compound and RIP 140 may be labelled with a fluorescent label such that the binding between the candidate compound and RIP 140 may be detected by an intrinsic fluorescence change which occurs when the candidate compound binds to RIP 140.
  • the candidate compound may be joined to a fluorescence resonance energy transfer (FRET) donor and RIP 140 to a FRET acceptor (or vice versa) such that, when the candidate compound and RIP 140 interact, stimulation of the FRET donor excites the FRET acceptor causing it to emit photons.
  • FRET fluorescence resonance energy transfer
  • Interaction may be also be detected by fluorescent labelling of the candidate compound and/or RIP 140 such that fluorescence is quenched when they form a complex.
  • Other methods for assessing interaction between the candidate compound and RIP 140 or between a candidate compound and a target protein may include using NMR to determine whether a RIP140:candidate compound complex or a target proteimcandidate compound complex is present.
  • a RIP 140: candidate compound complex or a target proteinxandidate compound complex may also be detected as a band at a particular position when run on a gel.
  • Another method of assessing interaction between RIP 140 and a candidate compound may involve immobilising RIP 140 on a solid surface and assaying for the presence of free candidate compound. If there is no interaction between the candidate compound and RIP 140, free candidate compound will be detected.
  • the candidate compound may be labelled to facilitate detection. This type of assay may also be carried with the candidate compound being immobilised on the solid surface. Interaction between the immobilised RIP 140 and the free candidate compound may also be monitored by a process such as surface plasmon resonance.
  • the methods of screening for compounds that bind to a complex of RIP 140 and a target protein can be carried out using the methods described above but using a complex of RIP 140 and a target protein in place of RIP 140 or in place of the target protein.
  • Direct screening for modulators of a complex of RIP140 and a target protein Modulation of the interaction between RIP 140 and a target protein in the presence of candidate compounds may be assessed directly.
  • Various methods for direct detection of protein/protein interactions are available.
  • the methods described above for assessing whether a candidate compound binds to RIP 140, to a target protein, or to a complex of RIP 140 and a target protein, can also be used to assess whether a candidate compound modulates the interaction between RIP 140 and a target protein.
  • the target protein is already known to bind RIP 140 and the methods described above are hence used to assess whether that interaction is disrupted or promoted by the candidate compound.
  • one of both the target protein and RIP 140 may be labelled with a fluorescent label so that interaction between the target protein and RIP 140 may be detected by an intrinsic fluorescence change that occurs when the RIP140:target protein complex is formed or disrupted in the presence of a candidate compound.
  • Interaction of RIP 140 and a target protein in the presence of candidate compound may also be assessed by detecting the accessibility of peptide sequences (e.g. epitopes) on RIP 140 and/or the target protein that are masked when the two proteins form a complex.
  • peptide sequences e.g. epitopes
  • motifs on RIP140 that interact with nuclear receptors have been identified in WO98/49561, Lee et al (Mol Cell Biol, 1998, 18(11): 6745-44) and in Wei et al (J. Biol. Chem., 2001, 276(19): 16107-12).
  • a lack of interaction between RIP 140 and a target protein that is a nuclear receptor in the presence of a candidate compound may therefore be determined by detection of such motifs, for example, using antibodies.
  • Indirect methods for assessing whether the interaction between a target protein and RIP 140 is modulated in the presence of a candidate compound may also be used.
  • One indirect method of screening for modulation of the interaction between target protein and RIP 140 in the presence of a candidate compound involves using a two-hybrid system.
  • the target protein may be fused to an activation domain of a transcription factor and RIP140 to a DNA-binding domain of a transcription factor (or vice versa), such that interaction between the target protein and RIP 140 promotes the transcription of a reporter gene in a cell.
  • the invention provides a method of screening for compounds that modulate the interaction between the target protein and RIP 140, said method comprising: a) contacting a cell containing a nucleic acid molecule comprising a promoter operatively linked to a reporter gene with: (i) a first fusion protein comprising one of the target protein and RIP 140 fused to the activation domain of a transcription factor, (ii) a second fusion protein comprising the other of the target protein and RIP 140 fused to the DNA- binding domain of a transcription factor; and (iii) a candidate compound; and b) assessing the level of expression of the reporter gene, wherein interaction between the target protein and RIP 140 promotes transcription of the reporter gene by activating said promoter.
  • This method may be used to assess interaction between RIP 140 and the target protein in any eukaryotic cell.
  • the method is used to assess the interaction between RIP 140 and the target protein in a yeast cell or a mammalian cell.
  • the candidate compound is an organic compound and a yeast two-hybrid system is being used
  • the permeability of the yeast cell wall is preferably enhanced e.g. by using chemicals such as polymyxin B.
  • the level of expression of a reporter gene in the two-hybrid system is indicative of the level of interaction between the target protein and RIP 140.
  • a candidate compound that inhibits the interaction between the target protein and RIP 140 decreases or abolishes the level of expression of the reporter gene.
  • a candidate compound that promotes the interaction between the target protein and RIP140 maintains or increases the level of expression of the reporter gene.
  • the reporter gene is easily assayed.
  • the reporter gene may give a detectable signal, such as a visible signal.
  • the reporter gene may encode a protein which gives a visible signal itself, or which catalyses a reaction which gives a visible signal or change e.g. a fluorescent protein or an enzyme.
  • the reporter gene may encode an enzyme such as a beta- galactosidase or a peroxidase, both of which are commonly used with coloured substrates and/or products.
  • the reporter gene may encode a green fluorescent protein (GFP) or a fluorescent derivative thereof such as YFP or CFP (see Prasher et al, 1995, Trends Genet 11(8): 320).
  • the reporter gene may encode a luminescent protein, such as luciferase.
  • the reporter gene may drive DNA replication (Nasavada et al, 1991, PNAS, 88:10686-10690) in the cell or may encode a drug resistance marker (Fearon et al, 1992, PNAS 89: 7958-7962).
  • the reporter gene may encode a protein that enables positive selection of cells in which the interaction between the target cell and RIP 140 is inhibited.
  • the reporter gene may encode a protein that is toxic or cytostatic so that only cells that do not express the reporter are able to survive or grow. As a result, the only cells to survive are those in which a candidate compound inhibits the interaction between the target protein and RIP 140 so that the reporter gene is not expressed.
  • reporter genes of this type that may be used in yeast include URA3, LYS2 and CYH2 (see Nidal et al, 1996, PNAS, 93: 10315-10320).
  • the protein encoded by the reporter gene may also prevent cell growth in the absence or presence of a particular amino acid or other component in cell media.
  • the reporter gene may encode a D ⁇ A-binding protein, TnlO tetracycline, which represses transcription of a TetRop-HIS3 gene so that yeast cells in which the reporter gene is expressed do not grow in the absence of histidine (see Shih et al, 1996, PNAS, 93: 13896-13901).
  • yeast cells in which the interaction between RIP 140 and the target protein has been disrupted do not express TN10 tetracycline and are consequently able to grow in the absence of histidine.
  • the proteins encoded by the reporter genes may be in the form of fusion proteins. Methods for the generation of fusion proteins are standard in the art and will be known to the skilled reader.
  • SF-1 is a transcription factor which is inhibited by RIP 140 while transcription of genes by AhR is activated by RIP 140 (Kumar et al, J. Biol. Chem. (1999) 274:22155-22164).
  • Interaction between RIP140 and a target protein of this type may also be assessed indirectly by means of a reporter gene under the control of a promoter which is regulated by the target protein. Where binding of RIP 140 to the target protein inhibits transcription of the reporter gene, disruption of the RIP140:target protein complex in the presence of a candidate compound will result in expression of the reporter gene. Conversely, where binding of RIP 140 to the target protein promotes transcription of the reporter gene, disruption of the RIP140:target protein interaction will inhibit transcription of the reporter gene.
  • the invention provides a method of screening for compounds that modulate the interaction between RIP 140 and a target protein which is a transcription factor, said method comprising: a) contacting a nucleic acid molecule, comprising a target protein-regulated promoter operatively linked to a reporter gene, with one or more candidate compounds, in the presence of RIP 140 and the target protein; and b) assessing the level of expression of the reporter gene.
  • This method employs a nucleic acid molecule comprising a promoter operatively linked to a reporter gene, such that transcription of the reporter gene is under the control of the promoter and is regualted by the target protein.
  • These nucleic acids are referred to as reporter constructs.
  • the promoter in the construct is a target protein-regulated promoter from which the target protein is able to promote transcription of the reporter gene.
  • the nature of the promoter will depend on the identity of the target protein.
  • the target protein is a nuclear hormone receptor, such as ER or TR
  • the promoter contains a hormone response element to which the nuclear hormone receptor binds to initiate transcription.
  • SF-1 regulated promoters contain at least one gonadotroph specific element (GSE) to which SF-1 binds to initiate transcription (Bryan et al, 1999, J Molec. Endocrin, 22:241-249).
  • the target protein-regulated promoter may be derived from the region upstream of any gene whose transcription is regulated by the target protein.
  • the reporter gene controlled by the promoter may be a gene which is regulated by the target protein in nature.
  • the reporter construct preferably includes the reporter gene and its natural upstream regulatory sequences.
  • the promoter controls transcription of a heterologous reporter gene which is easily assayed, as described above for two- hybrid methods.
  • compounds that inhibit the interaction between that target protein and RIP 140 are always detected as a result of a decrease in the expression of the reporter gene.
  • the result depend on whether RIP 140 acts to activate or inhibit the target protein's activity: compounds that inhibit the interaction between RIP 140 and the target protein give an increase in the expression of the reporter gene when RIP 140 inhibits the activity of the target protein, but give a decrease in expression of the reporter gene when RIP 140 activates the activity of the target protein.
  • the reporter gene is a fluorescent protein
  • inhibition of the interaction between RIP 140 and a target protein that is inhibited by RIP 140 may be detected by an increase in fluorescent protein expression (cf. two-hybrid system).
  • inhibition of the interaction between RIP 140 and a target protein that is activated by RIP 140 may be detected by a decrease in fluorescent protein expression.
  • the reporter gene is a toxic gene
  • inhibition of the interaction between RIP 140 and a target protein that is inhibited by RIP 140 may be detected by an increase in cell death.
  • inhibition of the interaction between RIP 140 and a target protein that is activated by RIP 140 may be detected by an increase in cell survival.
  • the proteins encoded by the reporter genes may be in the form of fusion proteins as described above.
  • genes that give a visible signal may be fused downstream of a gene that is linked in nature to the promoter in the construct.
  • Reporter constructs used in the indirect screening methods of the invention may be in the form of a viral vector or a non- viral vector.
  • the nucleic acid molecules used in these methods of the invention are in the form of a conventional non- viral vector, such as a plasmid.
  • the introduction of the non-viral vector into the animal cells may be carried out by any method known in the art including dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of polynucleotides in liposomes or direct microinjection of the DNA into nuclei, etc.
  • the invention provides the use of a reporter construct, as described above, in a method of screening for compounds that modulate fat storage.
  • the methods of the invention may be carried out in cell-free systems or in cells or tissues.
  • the indirect screening methods described above may be carried out in a cell-free system, in a cell or in a tissue.
  • the cell-free system must contain all the necessary components for transcription of the reporter gene where the level of expression is detected by measuring mRNA levels, and all the necessary components for transcription and translation of the reporter gene where the level of expression is assessed by measuring protein levels. It is preferred that the methods of screening of the invention be conducted in cell-free systems since this facilitates high-throughput screening of candidate compounds.
  • Indirect screening methods of the invention are preferably carried out in eukaryotic cells, such as mammalian (e.g. human) cells or tissues, or yeast cells.
  • eukaryotic cells such as mammalian (e.g. human) cells or tissues, or yeast cells.
  • the cell should preferably express both RIP 140 and the target protein endogenously. If RIP 140 and the target protein are not endogenously expressed, they may be introduced into the cell using a viral or non- viral vector encoding the RIP 140 or target protein. Preferably, RIP140 and the target protein are introduced into the cell in the form of plasmids. Assessing level of expression of reporter gene
  • the level of expression of a reporter gene may be assessed by measuring the level of a mRNA transcribed from the reporter gene or the level of protein translated after its transcription.
  • the measurement methods may be qualitative or quantitative.
  • the level of mRNA transcribed from a reporter gene can be assessed, for example, by traditional blotting techniques described in Sambrook et al [supra].
  • Messenger RNA can be purified and separated using gel electrophoresis. The nucleic acids on the gel are then blotted onto a solid support, such as nitrocellulose. The solid support is exposed to a labelled probe and then washed to remove any unhybridized probe. Next, the duplexes containing the labelled probe are detected. Typically, the probe is labelled with a radioactive moiety.
  • the level of mRNA transcribed from the reporter gene may be detected by PCR- based methods.
  • the mRNA transcribed from the reporter gene may be specifically amplified using primers that only bind to the mRNA with the amplified mRNA being detected using the blotting methods described above.
  • the level of transcription of the reporter gene could be also detected using fluorescence resonance energy transfer (FRET) through fluorophores coupled to two oligonucleotides that are complementary to the mRNA transcribed from the reporter gene, (see Wouters et al, 2001, Trends in Cell Biology 11, 203-211).
  • FRET fluorescence resonance energy transfer
  • RNA-specific detection technique As the cell-free system, cell or tissue will contain DNA from which the mRNA is transcribed, it is preferred to use a RNA-specific detection technique or to focus on sequence present in the mRNA transcription but not in the DNA (e.g. splice junctions, polyA tail etc.).
  • the methods of the invention may comprise an initial step of: extracting mRNA from the cell-free system, cell or tissue; removing DNA from the cell-free system, cell or tissue; and/or disrupting DNA but not mRNA in the cell-free system, cell or tissue.
  • Methods for selectively extracting RNA from biological samples are well known and include methods based on guanidinium buffers, lithium chloride, acid phenol: chloroform extraction, SDS/potassium acetate, etc.
  • RNA may be enriched for example using oligo-dT techniques.
  • Methods for removing DNA from biological samples include DNase digestion.
  • Methods for removing DNA encoding the reporter gene but not the RNA transcribed from it will use an agent which is specific to a sequence within the DNA.
  • Measuring level of protein Measurement of mRNA levels is not ideal in high throughput screening methods, so it is preferred that reporter gene expression is assessed by measuring protein levels.
  • the level of protein expressed from the reporter gene can be conveniently measured by using an antibody which binds to the protein encoded by the reporter gene. Following removal of unbound antibody, the level of the protein encoded by the reporter gene can be determined by assessing the level of the antibody bound to it. This may be done by labelling the antibody that binds to the protein or by using a second labelled antibody which binds to the first antibody.
  • the level of expression of the reporter gene is preferably assessed by detecting the visible signal.
  • the reporter gene encodes a fluorescent protein such as GFP, or an enzyme such as luciferase
  • the level of expression may be assessed by fluorescence/luminescence detection.
  • the reporter gene encodes a protein that is toxic or cytostatic
  • the level of expression may be assessed by looking at cell survival or cell growth.
  • Reference standards A reference standard (e.g. a control), is typically needed in order to detect whether the interaction between the target protein and RIP 140 is modulated in the method of the invention.
  • the interaction between target protein and RIP 140 in the presence of a candidate compound may be compared with the interaction between the target protein and RIP 140 in the absence of a candidate compound.
  • the reference may have been determined before performing the method of the invention, or may be determined during (e.g. in parallel) or after the method has been performed. It may be an absolute standard derived from previous work.
  • Proteins for use in methods of the invention may use target proteins and RIP 140 derived from any eukaryote.
  • they use target proteins and RIP 140 derived from an animal, such as a mammal.
  • RIP 140 and the target protein used in a method of the invention are both derived from the same mammal.
  • the RIP 140 and target protein are preferably both human proteins.
  • the RIP 140 gene has been cloned in a number of mammalian species including humans (Cavailles et al, 1995, EMBO , 14:3741-3751) and mouse (Lee et al, 1998, Mol Cell Biol, 18: 6745-55), and variants including natural biological variants, allelic variants and mutants containing amino acid substitutions, insertions or deletions from the wild-type sequence are in the NCBI database.
  • references in the methods of the invention to the use of RIP 140 include these and other variants, provided that, where the method involves a RIP140/target protein interaction, the variants should retain the ability to interact with the target protein of interest. Similarly, variants of target proteins may be used if they retain the ability to interact with RIP 140.
  • the methods of the invention may focus on single modules within the protein, and in particular on modules which possess a particular binding activity (e.g. on the region of ER which is involved in the ER/RIP140 interaction).
  • Motifs in RIP 140 that interact with nuclear receptors are disclosed in WO98/49561, Lee et al (Mol Cell Biol, 1998, 18(11): 6745-44) and Wei et al (J. Biol. Chem., 2001, 276(19): 16107-12).
  • fragments of target proteins that interact with RIP 140 and fragments of RIP 140 that interact with target proteins may used in the methods of the invention.
  • Other suitable variants of RIP 140 and target proteins are known from the literature.
  • Polypeptides that are structurally similar to target proteins and RIP 140, or to fragments of RIP 140 and target proteins that retain the ability to interact may also be used in the methods of the invention. These may be derived from natural target proteins or RIP 140 or they may be prepared synthetically or using techniques of genetic engineering. In particular, synthetic molecules that are designed to mimic the tertiary structure of target proteins or RIP 140 and in particular the domains of the target protein and RIP 140 that interact may be used in the methods of the invention. References to the use of target proteins and RIP 140 in the methods of the invention include the use of polypeptides that are structurally similar to target proteins and RIP 140, or to fragments thereof.
  • references to the use of target proteins and RIP 140 in the methods of the invention also include the use of fusion proteins comprising target proteins or RIP 140, fusion proteins comprising variants or fragments thereof, or fusion proteins comprising polypeptides that are structurally similar to target proteins or RIP 140 or to fragments of target proteins or RIP 140.
  • Such fusion proteins are particularly useful in two-hybrid methods.
  • Typical candidate compounds for use in all the screening methods of the invention include, but are not restricted to, peptides, peptoids, proteins, lipids, metals, small organic molecules, RNA aptamers, antibiotics and other known pharmaceuticals, polyamines, antibodies or antibody derivatives (e.g. antigen-binding fragments, single chain antibodies including scFvs, etc), and combinations or derivatives thereof.
  • Small organic molecules have a molecular weight of about more than 50 and less than about 2,500 daltons, and most preferably between about 300 and about 800 daltons.
  • Candidate compounds may be derived from large libraries of synthetic or natural compounds. For instance, synthetic compound libraries are commercially available from MayBridge Chemical Co.
  • libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts may be used.
  • candidate compounds may be synthetically produced using combinatorial chemistry either as individual compounds or as mixtures.
  • RIP 140 binding of RIP 140 to nuclear receptors may be inhibited by acetylation of the domains in RIP 140 that interacts with the receptor (Vo et al, 2001, Mol Cell Biol, 21(18): 6181-8).
  • Candidate compounds may therefore be compounds identified as acetylating RIP 140 in a pre-screen.
  • the invention further provides a compound that binds to RIP 140 or to a target protein, or that binds to a complex of RIP 140 and a target protein, or that modulates the interaction between RIP 140 and a target protein, obtained or obtainable by any of the methods described above.
  • the compounds of the invention are organic compounds.
  • compositions comprising a compound binds to RIP 140 or to a target protein, or that binds to a complex of RIP 140 and a target protein, or that modulates the interaction between RIP 140 and a target protein, obtained or obtainable by any of the methods described above.
  • Compounds that are found to modulate fat storage may be useful for the treatment of disorders associated with deviation in fat storage in their own right or may be lead compounds for the development of new drugs for the treatment of such disorders. They may also be useful in research into the regulation of fat storage.
  • compositions comprising those compounds.
  • the invention further provides compounds obtained or obtainable using the methods of the invention, and compositions comprising those compounds, for use as a medicament e.g. for increasing or decreasing fat storage.
  • the invention also provides the use of compounds obtained or obtainable using the methods of the invention, or compositions comprising those compounds in the manufacture of a medicament to increase or decrease fat storage.
  • the compounds may be used to treat or prevent disorders associated with increased or decreased fat storage, such as obesity or anorexia.
  • a method of increasing or decreasing fat storage comprising administering a compound obtained or obtainable by any one of the methods of the invention, or a composition comprising such a compound, to a mammal, preferably a human, is also provided.
  • Figure 2 A) Weight gain and adipose tissue deposition in wild-type (WT) and RIP 140 knockout (KO) mice; B) MRI scans and whole body spectra of wild-type and RIP 140 knockout mice.
  • FIG 4 Comparison of glucose tolerance tests in wild-type and RIP 140 null (Nripl null) mice.
  • Figure 5 Comparison of white adipose tissue (WAT) histology and cell size in wild-type (WT) and RIP 140 knockout (KO) mice.
  • WAT white adipose tissue
  • FIG. 6 Comparison of brown adipose tissue (BAT) and skeletal muscle histology in wild-type (WT) and RIP140 knockout (KO).
  • Figure 7 A) Comparison of differentiation to adipocytes in wild-type (WT) and RIP 140 knockout (KO) cells; B) Expression analysis of RIP 140 mRNA.
  • Figure 8 Relative expression of RIP140 in different tissues of a wild-type mouse.
  • FIG. 9A compares expression of PPAR ⁇ , C/EBPa, C/EBPb and SREBP.
  • Figure 9B compares expression of Glut4, aP2, leptin and adipolectin.
  • Figure 9C compares expression of G6Pase, PEPCK and 24p3. Expression in wild-type mice is shown in left-hand column of each pair of columns and expression in RIP 140 null mice in the right hand column of each pair of columns.
  • FIG 10 Expression of C/EBP ⁇ in white adipose tissue (WAT), brown adipose tissue (BAT), muscle and liver of faster and fed wild-type mice (WT) and RIP 140 knockout mice (KO). Expression in wild-type mice is shown in left-hand column of each pair of columns and expression in RIP 140 null mice in the right hand column of each pair of columns.
  • FIG 11 Expression of PPAR ⁇ in white adipose tissue (WAT), brown adipose tissue (BAT), muscle and liver of faster and fed wild-type mice (WT) and RIP 140 knockout mice (KO). Expression in wild-type mice is shown in left-hand column of each pair of columns and expression in RIP 140 null mice in the right hand column of each pair of columns.
  • FIG. 12 Expression of SREBPlc in white adipose tissue (WAT), brown adipose tissue (BAT), muscle and liver of faster and fed wild-type mice (WT) and RIP140 knockout mice (KO). Expression in wild-type mice is shown in left-hand column of each pair of columns and expression in RIP 140 null mice in the right hand column of each pair of columns.
  • Figure 13 Expression of Glut4 in white adipose tissue (WAT), brown adipose tissue (BAT), muscle and liver of faster and fed wild-type mice (WT) and RIP 140 knockout mice (KO). Expression in wild-type mice is shown in left-hand column of each pair of columns and expression in RIP 140 null mice in the right hand column of each pair of columns.
  • FIG 14 Expression of leptin in white adipose tissue (WAT), brown adipose tissue (BAT), muscle and liver of faster and fed wild-type mice (WT) and RIP 140 knockout mice (KO). Expression in wild-type mice is shown in left-hand column of each pair of columns and expression in RIP 140 null mice in the right hand column of each pair of columns.
  • FIG. 15 Expression of CPTlb in white adipose tissue (WAT), brown adipose tissue (BAT), muscle and liver of faster and fed wild-type mice (WT) and RIP 140 knockout mice (KO). Expression in wild-type mice is shown in left-hand column of each pair of columns and expression in RIP 140 null mice in the right hand column of each pair of columns.
  • FIG 16 Expression of UCP1 in white adipose tissue (WAT), brown adipose tissue (BAT), muscle and liver of faster and fed wild-type mice (WT) and RIP 140 knockout mice (KO). Expression in wild-type mice is shown in left-hand column of each pair of columns and expression in RIP 140 null mice in the right hand column of each pair of columns.
  • Figure 17 Expression of PPAR ⁇ in white adipose tissue (WAT), brown adipose tissue (BAT), muscle and liver of faster and fed wild-type mice (WT) and RIP 140 knockout mice (KO).
  • MRI Magnetic resonance Imaging
  • MRS Spectroscopy
  • h Morphology of inguinal white adipose tissue (WAT), brown adipose tissue (BAT), and liver from wild type (WT) and RIP140 null mice (KO), fed a high fat diet (35% w/w) for 10 days.
  • WAT white adipose tissue
  • BAT brown adipose tissue
  • KO liver from wild type (WT) and RIP140 null mice (KO) fed a high fat diet (35% w/w) for 10 days.
  • White and brown adipose tissues were stained with hematoxylin and eosin (H/E). Liver tissues were stained with oil Red O to demonstrate lipid accumulation and counterstained with H/E. Scale bars correspond to 50 ⁇ m.
  • Figure 20 RIP140 expression in metabolic tissues and differentiated adipocytes in vitro.
  • Regulation of RIP 140 mRNA levels during differentiation of 3T3-L1 adipocyte cells Differentiation was induced in 2-day confluent cells using standard hormone cocktail of insulin, dexamethasone and IBMX.
  • pc preconfluent cells
  • Cells treated identically were also subject to ⁇ -galactosidase activity analysis to demonstrate RIP140 promoter activity (right panel). Note the lack of ⁇ -galactosidase activity in wild type cells and the correlation of fat droplet containing cells staining with oil red O and ⁇ - galactosidase.
  • Figure 23 UCP-1 gene expression is elevated in cells devoid of RIP 140.
  • a Taqman real-time PCR analysis of UCP-1 mRNA levels in 3T3L1 and RIPKO-1 cells, pre-confluent (PC), and following treatment with standard diffentiation cocktail including rosiglitazone on days 0-5 as indicated
  • b Schematic representation of the murine UCP-1 enhancer element including the regulatory elements cAMP response element (CRE), peroxisome proliferator response element (PPRE), retinoid acid response element (RARE), thyroid response element (TRE), and brown adipocyte regulatory element (BRE).
  • CRE cAMP response element
  • PPRE peroxisome proliferator response element
  • RARE retinoid acid response element
  • TRE thyroid response element
  • BRE brown adipocyte regulatory element
  • RIPKO-1 cells were transfected with UCP-1 promoter luciferase reporter constructs, as indicated, and incubated in the absence and presence of differential cocktail including rosiglitazone (D+Ro) for 1 , 3 or 5 days.
  • D+Ro rosiglitazone
  • FIG. 24 Expression of RIP 140 inhibits PPAR dependent transcription of a CPTlb promoter and a PPRE regulated reporter gene, a, PPAR ⁇ , b, PPAR ⁇ and c, PPAR ⁇ were analysed using a mouse CPT1 genomic fragment consisting of 2.0kb of promoter sequence cloned into the pGL3 reporter (Promega) and a 3x PPRE-TK promoter in the same vector. Transfection experiments were carried out in COS cells (PPAR ⁇ and PPAR ⁇ ) and in HEK 293 cells (PPAR ⁇ ) in the presence of specific ligands for each receptor as indicated. MODES FOR CARRYING OUT THE INVENTION
  • mice were found to be viable but to have impaired growth, as demonstrated by an approximately 20-25% reduction in adult body weight. This reduction is primarily a consequence of considerably less accumulation of total body fat (approx. 75% reduction vs. wild-type), manifested in almost negligible appearance of subcutaneous fat and about half the amount of inguinal fat depots ( Figures 1 and 2).
  • the reduction of body fat in RIP 140 knockout mice appears to be due to reduced adipose cell size.
  • Expression analysis and in vitro differentiation suggest that the reduced adipose cell size may be due to impaired WAT function rather than impaired adipocyte differentiation.
  • Possible mechanism(s) for the observed phenotype is an increased fatty acid transport (up-regulation of CTPlb) and/or increased uncoupling (up-regulation of UCP- 1) in mitochondria of WAT.
  • Example 1 The experiments in Example 1 are described in more detail, along with additional results, in Example 2.
  • RIP 140 null mice are lean with a 20% reduction in body weight in both males and females compared to wild type mice, which increases with age (Fig. 19a and data not shown).
  • Magnetic resonance imaging (MRI) and spectroscopy (MRS) of whole body fat content revealed the almost complete absence of subcutaneous fat and a marked decrease in other fat depots (Fig. 19b).
  • Total body fat content was reduced by approximately 70% as analysed by whole body proton MRS while the weight of epididymal fat was reduced by 40-60%, according to the age of the mice (Fig. 19c). The reduction in body weight and fat content was not due to increased physical activity as judged by open field activity measurements (data not shown).
  • RIP 140 null mice The response of RIP 140 null mice to the effects of feeding a high fat diet for 10 days were determined.
  • the average weight gain in wild-type mice was 5.3 ⁇ 0.29 g, corresponding to a 14.5% increase of bodyweight whereas RIP140 null mice gained only 1.0 ⁇ 0.15 g, corresponding to 3.8% of bodyweight (Fig. 19e).
  • Serum biochemistry did not reveal increased free fatty acids and hypertriglyceridemia (Fig 19f).
  • the reduced fat content in adipose tissue leads to a reduction in circulating leptin levels (Fig. 19g).
  • RIP 140 mRNA is widely expressed, with highest levels in WAT followed by skeletal muscle and with lower levels in BAT and liver (Fig. 2a).
  • An expression profiling analysis of 3T3-L1 cells indicated that there was a marked progressive increase in RIP 140 expression during differentiation into adipocytes (Soukas, A. et al, 2001, J Biol Chem 276: 34167-74) and this was confirmed by quantitative real-time PCR analysis which showed that maximum levels occurred 3-6 days after induction of differentiation (Fig. 20a). Given these observations and the lean phenotype exhibited by RIP 140 null mice the role of RIP 140 in the differentiation and function of adipocytes was investigated.
  • RIP 140 was required for adipocyte differentiation was tested by comparing the ability of fibroblasts from wild type and RIP 140 null embryos to differentiate in vitro. It was found that confluent fibroblasts incubated in a standard hormone differentiation cocktail (with or without the PPAR ⁇ agonist rosiglitazone) were fully capable of differentiating into adipocytes, as judged by oil red O staining, even when cells were devoid of the RIP 140 gene (Fig. 20b and data not shown).
  • a link between adipocyte differentiation and RIP 140 gene transcription was demonstrated by analysing the expression of the ⁇ -galactosidase gene, which has been inserted in place of the RIP140 coding sequence in the null mice (White, R. et al., 2000 Nat Med 6: 1368-74).
  • Fig. 20b indicates that oil red O positive cells and ⁇ -galactosidase activity were detected only in differentiated cells suggesting that the adipose phenotype is required for RIP 140 promoter activity.
  • the delay in the increase in RIP 140 expression following hormone treatment and the ability of embryonic fibroblasts to differentiate in the absence of RIP 140 demonstrates that adipogenesis does not depend on this nuclear receptor corepressor.
  • the in vivo role of RIP 140 in maintaining the function of adipose and other metabolic tissues was analysed.
  • Fig. 22b the levels of SRC1 and TIF2, which have been found to control energy balance between white and brown adipose tissue, were not altered (Fig. 22b).
  • certain transcription factors required for adipocyte function such as CEBP ⁇ and SREBPlc, were reduced by 50-60% in WAT from RIP 140 null mice but not in other tissues (data not shown). Consistent with these reductions, there was a decrease in the expression of corresponding target genes including for example, adiponectin, leptin, glucose transporter 4 (Glut4) and aP2, (Fig. 22c, Fig. 19g and data not shown).
  • RIP 140 may regulate gene expression by examining the promoters for the UCP1 and CPTl genes in transfected cell lines.
  • a RIP 140 null cell line termed RIPKO-1 cells
  • RIPKO-1 cells was generated from mouse embryonic fibroblasts that could be induced to undergo adipocyte differentiation in vitro as judged by its ability to express a number of marker genes and to accumulate triglyceride (data not shown).
  • treatment of confluent RIPKO-1 cells with hormone cocktail to induce adipocyte differentiation results in the induction of UCP 1 (Fig. 23 a) and increased expression of CPTl (data not shown).
  • UCP1 transcription is subject to suppression by RIP 140 and that this is mediated by the 220bp regulatory region found 2.5 kb upstream of the start site of transcription, presumably by targeting transcription factors that bind to elements within this region of the promoter.
  • the UCP-1 enhancer element has been shown to be a target for a number of nuclear receptor signalling pathways, the best characterised of which is PPAR ⁇ .
  • the lean phenotype of RIP 140 null mice and the level of upregulation of UCP 1 resembles that found in mice in which an activated form of PPAR ⁇ is overexpressed in adipose tissue, raising the possibility that this PPAR isoform might also be a target for repression.
  • RIP 140 The ability of RIP 140 to repress the transcriptional activity of the three individual PPAR isoforms in cells devoid of these receptors was exmined. Since the UCP1 promoter exhibited minimal activity in heterologous cell lines compared with that in adipocytes, luciferase reporter genes containing the CPTl promoter sequences and a PPRE consensus sequences as a control were analysed. It was found that RIP 140 was able to repress the ability of PPAR ⁇ (Fig. 24a) PPAR ⁇ (Fig. 24b) and PPAR ⁇ (Fig. 24c) to stimulate transcription from both the CPTlb promoter and the PPRE containing reporter gene.
  • the present work demonstrates an essential role for the corepressor RIP 140 in the maintenance of energy homeostasis with ligand dependent transcriptional repression of nuclear receptors playing a fundamental role in preventing the expression of key metabolic genes in WAT.
  • the upregulation of UCPl is an intrinsic property of WAT devoid of RIP 140 since this was also observed when null cells were differentiated into adipocytes in cell culture. It is proposed that the UCPl gene is normally repressed in WAT as a consequence of RIP 140 targeting a 220 bp enhancer element 2.5 kb upstream of the start site of transcription.
  • the UCPl enhancer may be regulated by all three subtypes of PPAR, by retinoids and thyroid hormone, as well as through activation of ⁇ -adrenergic receptor intracellular signalling pathways.
  • Expression of UCPl has been shown to be induced in WAT in transgenic mice by introduction of an activated form of PPAR ⁇ and also in human white adipocytes in culture as a result of expression of PGCl ⁇ and activation by ligands for PPAR ⁇ (Tiraby, C. et al, 2003, JBiol Chem, 278: 33370-6).
  • RIP140 is capable of interacting with all of these receptors in a ligand-dependent manner and of repressing their transcriptional activity, as shown for PPARs in this study (Cavailles, N. et al, 1995, Embo J, 14: 3741-51; L ⁇ orset, F. et al, 1996, Mol Cell Biol, 16: 6029-36; Treuter, E. et al, 1998, Mol Endocrinol 12: 864-81; Tazawa, H. et al, 2003, Mol Cell Biol, 23: 4187-98). Recent studies have demonstrated that RIP 140 has intrinsic repressive activity contained within four repression domains within the protein No, ⁇ .
  • the temporal and cell type specific control of RIP 140 expression provides an additional level of regulation of repressor function.
  • the delayed onset of RIP 140 expression may be necessary to avoid interference with the action of PPAR ⁇ during the process of adipogenesis while facilitating a role in regulating the activity of the differentiated adipocyte.
  • RIP 140 is essential to prevent the upregulation of genes involved in energy dissipation and mitochondrial uncoupling, processes that compromise the function of the adipocyte as a site of energy storage in the form of triglycerides.
  • RIP 140 plays a crucial role in energy homeostasis and demonstrate a novel role for corepressor action in addition to coactivator recruitment in determining adipocyte function. Since the interaction of RIP 140 with nuclear receptors is a ligand-dependent process, specific recruitment of this corepressor provides a novel therapeutic target for the treatment of obesity and related disorders.
  • mice The generation of RIP 140 null mice have previously been described (White, R. et al, 2000, Nat Med, 6: 1368-74). Mice used in this study were backcrossed 6 generations to C57BL/6J background. Mice were maintained under standard conditions, with controlled light and temperature and fed a chow diet ad libitum, except for high fat diet experiments where mice were fed a 35% w/w diet (Lillico). All experiments were performed according to Home Office guidelines.
  • MRT Magnetic resonance imaging
  • MRS spectroscopy
  • Leptin measurements were determined using Linco Research Inc Mouse Leptin RIA kit. Triglycerides were measured using Triglyceride GPO-Trinder reagent (Sigma) and free fatty acids determined using ADIFAB free fatty acid indicator (Molecular Probes). All assays were carried out according to the manufacturers protocols. Morphological and immuno-histochemical analysis
  • Tissues were fixed in neutral buffered formalin, embedded in paraffin and sectioned at 5 ⁇ m onto poly-L-lysine coated slides. For histology, sections were stained with haematoxylin and eosin. For immunohistochemistry, deparaffinised sections were incubated in 0.3% hydrogen peroxide (Sigma) in methanol for 30 min to inactivate endogenous peroxidase, rinsed in PBS and incubated in 1:75 normal goat serum/PBS for 30 min to reduce non-specific background staining. Sections were incubated overnight at 4°C with polyclonal rabbit anti-mouse primary antibody to UCPl (AB3038, Chemicon International) diluted 1:800 in PBS.
  • Oxygen consumption was measured in wild type and RIP 140 null mice fed a chow diet (7 mice per group) using the OXYMAX System v4.66 (Columbus Instruments, Columbus, OH) and conditions of a settling time of 180 s, measuring time of 60 s, and room air as a reference. Animals were placed individually in 4 0.3-liter chambers. Results are expressed as ml/kg/min. Cell culture of mouse embryonic fibroblasts and 3T3-L1 cells.
  • Mouse embryonic fibroblasts were isolated and cultured using standard protocol in DMEM/F12 media supplemented with 10% foetal bovine serum. For differentiation experiments, MEFs cultured for 4-6 passages were used. 3T3-L1 cells were cultured in DMEM/F12 media supplemented with 10% newborn bovine serum. Differentiation of 2 day post-confluent MEFs and 3T3-L1 cells was performed as previously described (Soukas, A. et al, 2001, J Biol Chem 276: 34167 - A) with the modification that MEFs which were also supplemented with rosiglitazone.
  • ⁇ - galactosidase activity was analysed as previously described (White, R. et al, 2000, Nat Med, 6: 1368-74).
  • the cell line RIPKO-1 was generated by culturing RIP 140 null MEFs for greater than 20 passages.
  • UCP-1 promoter luciferase reporter constructs were generated by cloning a 4Kb fragment or 220bp enhancer element (-2530 to -2310 bp relative to ATG) of the 5' flanking region of the murine UCP-1 gene into pGL3 /basic vector.
  • RIPKO-1 cells were transfected, immediately following plating into 24-well plates, using Fugene ⁇ with l ⁇ g reporter gene, 250ng pRL-CMN and/or 500ng pCI-RIP. Twenty-four hours following transfection the medium was replenished with either control medium or medium containing differentiation cocktail. Cells were harvested for luciferase assay 1, 3, and 5 days following the addition of the differentiation cocktail.
  • Total R ⁇ A was isolated using TRIzol according to the manufacturer's instructions. To obtain first strand cD ⁇ A for further analysis, 1 ⁇ g total R ⁇ A was treated with deoxyribonuclease, and cD ⁇ A was prepared using the Superscript First-Strand Synthesis System for RT-PCR according to the manufacturer's instructions. Real-time PCR was performed with the ABI PRISM 7700 Sequence Detection System. RIP 140 and LI 9 expression was determined using specific primers and TaqMan probes. Expression of all other genes was determined with SYBR-green reagent using specific primers according to the manufacturer's instructions. Expression levels for all genes were correlated to mean value of two independent internal controls, the ribosomal coding gene LI 9 and cyclophilin. Primer and probe sequences are available on request.
  • HEK293 or Cos7 cells were transfected using FuGE ⁇ E 6 with 20ng luciferase reporter, 5ng pRLCMV control, 0.5ng pcD ⁇ A3 PPAR ⁇ , ⁇ or ⁇ and/or lOng pCI RIP 140 where indicated. 24h after transfection cells were treated with ligands WY14,643 (lO ⁇ M), Rosiglitazone (5 ⁇ M), or GW501516 (5nM).
  • Cells were harvested 24hr after addition of ligands and luciferase activity measured using a Victor 2 luminometer.
  • the reporter firefly luciferase activity was measured using the LucLiteTM kit, subsequently the Renilla luciferase activity used as internal control was determined by the addition of EDTA (8 mM final concentration) and Coelenterazine substrate (4.7 ⁇ M (250ng/well)) to the firefly luciferase reaction.
  • the Renilla luciferase activity was used to correct for differences in transfection efficiency.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Immunology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Rheumatology (AREA)
  • Emergency Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Child & Adolescent Psychology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Microbiology (AREA)
  • Endocrinology (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
EP04707568A 2003-02-03 2004-02-03 Screening for modulators of fat storage Withdrawn EP1590674A2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB0302446.0A GB0302446D0 (en) 2003-02-03 2003-02-03 Screening for modulators of fat storage
GB0302446 2003-02-03
PCT/GB2004/000413 WO2004070347A2 (en) 2003-02-03 2004-02-03 Screening for modulators of fat storage

Publications (1)

Publication Number Publication Date
EP1590674A2 true EP1590674A2 (en) 2005-11-02

Family

ID=9952335

Family Applications (1)

Application Number Title Priority Date Filing Date
EP04707568A Withdrawn EP1590674A2 (en) 2003-02-03 2004-02-03 Screening for modulators of fat storage

Country Status (6)

Country Link
EP (1) EP1590674A2 (ja)
JP (1) JP2006518591A (ja)
AU (1) AU2004209335A1 (ja)
CA (1) CA2515347A1 (ja)
GB (1) GB0302446D0 (ja)
WO (1) WO2004070347A2 (ja)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7691823B2 (en) * 2004-03-05 2010-04-06 University Of Massachusetts RIP140 regulation of glucose transport
JP2006090948A (ja) * 2004-09-27 2006-04-06 Fuji Photo Film Co Ltd バイオセンサーを用いたスクリーニング方法

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6266622B1 (en) * 1995-12-13 2001-07-24 Regents Of The University Of California Nuclear receptor ligands and ligand binding domains
JP4296608B2 (ja) * 1997-08-27 2009-07-15 田辺三菱製薬株式会社 Pparのアゴニスト及びアンタゴニストのスクリーニング方法
GB9917405D0 (en) * 1999-07-23 1999-09-22 Univ Dundee Methods of treatment and drug screening methods
AU1758401A (en) * 1999-11-12 2001-06-06 Merck & Co., Inc. Melanocortin-3 receptor deficient cells, non-human transgenic animals and methods of selecting compounds which regulate body weight
US20020004205A1 (en) * 2000-03-30 2002-01-10 Consler Thomas G. Method of investigating functional molecular interactions and reagents for use therein
US20030104975A1 (en) * 2001-06-14 2003-06-05 Johan Auwerx Cofactor-based screening method for nuclear receptor modulators and related modulators

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2004070347A2 *

Also Published As

Publication number Publication date
WO2004070347A2 (en) 2004-08-19
WO2004070347A3 (en) 2004-12-16
GB0302446D0 (en) 2003-03-05
AU2004209335A1 (en) 2004-08-19
CA2515347A1 (en) 2004-08-19
JP2006518591A (ja) 2006-08-17

Similar Documents

Publication Publication Date Title
Koh et al. Lipin1 is a key factor for the maturation and maintenance of adipocytes in the regulatory network with CCAAT/enhancer-binding protein α and peroxisome proliferator-activated receptor γ2
Stayrook et al. Regulation of carbohydrate metabolism by the farnesoid X receptor
Ludbrook et al. Excess DAX1 leads to XY ovotesticular disorder of sex development (DSD) in mice by inhibiting steroidogenic factor-1 (SF1) activation of the testis enhancer of SRY-box-9 (Sox9)
Sasaki et al. Induction of hypothalamic Sirt1 leads to cessation of feeding via agouti-related peptide
de Souza et al. The estrogen receptor α colocalizes with proopiomelanocortin in hypothalamic neurons and binds to a conserved motif present in the neuron-specific enhancer nPE2
Teijido et al. Localization and functional analyses of the MLC1 protein involved in megalencephalic leukoencephalopathy with subcortical cysts
Luiro et al. Batten disease (JNCL) is linked to disturbances in mitochondrial, cytoskeletal, and synaptic compartments
Schmidt et al. SORLA facilitates insulin receptor signaling in adipocytes and exacerbates obesity
US20050136429A1 (en) SIRT1 modulation of adipogenesis and adipose function
Subkhangulova et al. SORCS 1 and SORCS 3 control energy balance and orexigenic peptide production
Kubota et al. Lrp6 hypomorphic mutation affects bone mass through bone resorption in mice and impairs interaction with Mesd
Boutet et al. Taf1 regulates Pax3 protein by monoubiquitination in skeletal muscle progenitors
Szabo et al. Calreticulin inhibits commitment to adipocyte differentiation
Oyabu et al. FOXO1 cooperates with C/EBPδ and ATF4 to regulate skeletal muscle atrophy transcriptional program during fasting
Ito et al. Arginine methyltransferase CARM1/PRMT4 regulates endochondral ossification
Hayashi et al. Hypoxia up-regulates hypoxia-inducible factor-1α expression through RhoA activation in trophoblast cells
Smith et al. Mutant huntingtin interacts with β-tubulin and disrupts vesicular transport and insulin secretion
Arena et al. Somatostatin receptor subtype-dependent regulation of nitric oxide release: involvement of different intracellular pathways
WO2003027634A2 (en) Identification of modulatory molecules using inducible promoters
Lau et al. Dexras1 interacts with FE65 to regulate FE65-amyloid precursor protein-dependent transcription
Debril et al. Scaffold attachment factor B1 directly interacts with nuclear receptors in living cells and represses transcriptional activity
Bland et al. USP8 deubiquitinates the leptin receptor and is necessary for leptin-mediated synapse formation
Al-Massadi et al. Pharmacological and genetic manipulation of p53 in brown fat at adult but not embryonic stages regulates thermogenesis and body weight in male mice
Li et al. TMEM132A, a novel Wnt signaling pathway regulator through Wntless (WLS) interaction
US7981615B2 (en) Compositions and methods for treating diseases associated with T-box and N-Myc

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20050811

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: IMPERIAL COLLEGE INNOVATIONS LIMITED

DAX Request for extension of the european patent (deleted)
RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: IMPERIAL INNOVATIONS LIMITED

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1086063

Country of ref document: HK

17Q First examination report despatched

Effective date: 20060915

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20100408

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1086063

Country of ref document: HK