WO2007130431A2 - FUNCTION OF ESTROGEN RELATED RECEPTOR GAMMA (ERRγ) IN INCREASING MITOCHONDRIAL BIOGENESIS - Google Patents

FUNCTION OF ESTROGEN RELATED RECEPTOR GAMMA (ERRγ) IN INCREASING MITOCHONDRIAL BIOGENESIS Download PDF

Info

Publication number
WO2007130431A2
WO2007130431A2 PCT/US2007/010592 US2007010592W WO2007130431A2 WO 2007130431 A2 WO2007130431 A2 WO 2007130431A2 US 2007010592 W US2007010592 W US 2007010592W WO 2007130431 A2 WO2007130431 A2 WO 2007130431A2
Authority
WO
WIPO (PCT)
Prior art keywords
errγ
polypeptide
pgc
agonist
compound
Prior art date
Application number
PCT/US2007/010592
Other languages
French (fr)
Other versions
WO2007130431A3 (en
Inventor
Shamina M. Rangwala
Susan C. Stevenson
Zhidan Wu
Original Assignee
Novartis Ag
Novartis Pharma Gmbh
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 Novartis Ag, Novartis Pharma Gmbh filed Critical Novartis Ag
Publication of WO2007130431A2 publication Critical patent/WO2007130431A2/en
Publication of WO2007130431A3 publication Critical patent/WO2007130431A3/en

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
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/743Steroid hormones
    • 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
    • G01N33/689Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to pregnancy or the gonads
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/72Assays involving receptors, cell surface antigens or cell surface determinants for hormones
    • G01N2333/723Steroid/thyroid hormone superfamily, e.g. GR, EcR, androgen receptor, oestrogen receptor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/02Screening involving studying the effect of compounds C on the interaction between interacting molecules A and B (e.g. A = enzyme and B = substrate for A, or A = receptor and B = ligand for the receptor)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/04Endocrine or metabolic disorders

Definitions

  • the present invention relates to methods of identifying ERR ⁇ agonists and the obtained agonists in the treatment of diseases whose conditions are improved by activation of ERR ⁇ .
  • the estrogen-related receptors comprise 3 members, ERR ⁇ , ERR ⁇ , and ERR ⁇ , that form a subfamily of orphan nuclear receptors for which natural ligands have yet to be identified.
  • the ERRs share significant amino acid homology with the estrogen receptor (ER) within their DNA binding domains (DBD) and ligand binding domains (LBD), but do not respond to estradiol (Giguere, et al., Trends Endocrinol Metab., 13:220-5 (2002); Heard, et al., MoL Endocrinol, 14:382-92(2000).).
  • ERRs can activate gene transcription in a constitutive manner (Hong, et al., J. Bio. Chem., 21 A: 22618-26 (1999); Chen, et al., J. Bio. Chem., 276:28465-70 (2001).) and their activation ability may be determined by the presence of transcriptional coactivators (Kamei, et al., PNAS USA, 100:12378-83(2003); Schreiber, et al., J. Bio. Chem., 278:9013-8 (2003).).
  • ERR ⁇ is highly expressed in adult tissue such as the brain, skeletal muscle, heart, kidney and retina. It is also expressed in fetal tissue such as the placenta, brain, heart, skeletal muscle, kidney and lung (Bonnelye et al. Mot. Endo., 11: 905-16 (1997).).
  • the present invention provides a method for screening/identifying a compound that binds to a polypeptide containing the ligand-binding domain (LBD) of 50201A
  • ERR ⁇ (a ERR ⁇ LBD-containing polypeptide), and upon binding to ERRy, helps recruiting a PPAR gamma coactivator 1 alpha (PGC- l ⁇ ) polypeptide into contact with the ERR ⁇ LBD-containing polypeptide.
  • the screening is performed by contacting a compound with a biological sample containing the ERR ⁇ LBD-containing polypeptide and the PGC- l ⁇ polypeptide and selecting the compound that binds to the ERR ⁇ LBD- containing polypeptide and recruits the PGC- l ⁇ polypeptide.
  • the ERR ⁇ LBD-containing polypeptide is the ERR ⁇ polypeptide.
  • the screening assay is a cell-based screening assay.
  • a cell expressing an ERR ⁇ LBD-containing polypeptide and a PGC- l ⁇ polypeptide, or functional fragments thereof is contacted with a test compound, and the ability of the test compound to increase the activity of the ERR ⁇ LBD-containing polypeptide is determined.
  • the activity is transcriptional activity of the ERR ⁇ LBD-containing polypeptide.
  • the ERR ⁇ LBD-containing polypeptide is ERR ⁇ polypeptide.
  • Figure 1 shows the diagram of FRET assay configuration.
  • Figure 2 shows the agonist-induced FRET response.
  • Figure 3 shows comparison of the ability of ERR ⁇ and ERR ⁇ to transactivate a ERE reporter in the presence and absence of PGC-I ⁇ .
  • Figure 4 shows adenoviral overexpression of PGC-I ⁇ and ERR ⁇ in HeIa cells.
  • Figures 5 A and 5B show the expression of mitochondrial marker genes in HeIa cells over expressing PGC-l ⁇ and ERR ⁇ .
  • Figure 6 shows effects of overexpression of PGC-l ⁇ and ERR ⁇ on fatty acid oxidation in HeIa cells.
  • Figure 7 shows effects of overexpression of PGC-I alpha and ERR gamma on expression of PGC-I alpha and ERR ⁇ in ERR ⁇ null MEFs. 50201A
  • Figure 8 shows effect of overexpression of PGC-I alpha and ERR gamma on expression of mitochondrial markers in ERR alpha null MEFs.
  • Figure 9 shows effect of overexpression of PGC-I alpha and ERR gamma on expression of genes of oxidative stress protection in ERR alpha null MEFs.
  • Figure 10 shows effects of overexpression of ERR gamma and PGC-I alpha on cytochrome c protein levels in ERR alpha null MEFs.
  • Figure 11 shows the human ERR ⁇ polypeptide sequence (SEQ ID NO. 1).
  • Figure 12 shows the LBD domain sequence of human ERR ⁇ sequence (SEQ ID NO. 2).
  • FIG. 13 shows the ERR ⁇ ERE sequence (SEQ ID NO. 3).
  • Figure 14 shows expression levels of genes of oxidative phosphorylation in mouse myotubes treated with Compound 1.
  • Figure 15 shows expression levels of genes of fatty acid oxidation in mouse myotubes treated with Compound 1.
  • Figure 16 shows expression levels of IDH3 ⁇ and ATP-5b in mouse myotubes treated with Compound 1.
  • Figure 17 shows expression levels of UCP2 and UCP3 in mouse myotubes treated with Compound 1.
  • Figure 18 shows expression levels of ERR ⁇ and ERR ⁇ in mouse myotubes treated with Compound 1.
  • Figure 19 shows expression levels of PGC-I ⁇ and PGC- l ⁇ in mouse myotubes treated with Compound 1.
  • Figure 20 shows expression levels of PP ARa, PPAR ⁇ , and PPAR ⁇ in mouse myotubes treated with Compound 1. 50201A
  • Figure 21 shows protein expression levels of cytochrome c in mouse myotubes treated with Compound 1.
  • Figure 22 shows citrate synthase activity in mouse myotubes treated with Compound 1.
  • Figure 23 shows measurement of cellular respiration in primary mouse myotubes treated with Compound 1.
  • agonist of ERR ⁇ refers to a molecule which when bound to the LBD sequence of ERR ⁇ , increases the amount of, or prolongs the duration of, or enhances the activity of ERR ⁇ .
  • Agonists can include polypeptides, nucleic acids, carbohydrates, lipids, or any derivatives thereof, or any other molecules.
  • a sample refers to a whole organism or a subset of its tissues, cells or component parts (e.g. body fluids, including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen).
  • body fluids including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen).
  • a sample further refers to a homogenate, lysate or extract prepared from a whole organism or a subset of its tissues, cells or component parts, or a fraction or portion thereof, including but not limited to, for example, plasma, serum, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs. Most often, the sample has been removed from an animal, but the term “sample” can also refer to cells or tissue analyzed in vivo, i.e., without removal from animal.
  • a sample will contain cells from the animal, but the term can also refer to non-cellular biological material, such as non-cellular fractions of blood, saliva, or urine, that can be used to measure the cancer-associated polynucleotide or polypeptides levels.
  • a sample further refers to a medium, such as a nutrient broth or gel in which an organism has been propagated, which contains cellular components, such as proteins or nucleic acid molecules. 50201 A
  • polypeptide refers to a polymer in which the monomers are amino acids and are joined together through peptide or disulfide bonds. It also refers to either a full-length naturally-occurring amino acid sequence or a fragment thereof between about 8 and about 500 amino acids in length. Additionally, unnatural amino acids, for example, beta-alanine, phenyl glycine and homoarginine may be included. Commonly-encountered amino acids which are not gene-encoded may also be used in the present invention. All of the amino acids used in the present invention may be either the D- or L-optical isomer. The L-isomers are preferred.
  • a polypeptide sequence also encompasses naturally-occurring allelic variants of said polypeptide.
  • detectable polypeptide refers to a polypeptide whose presence or absence can be measured directly or indirectly, quantitatively or qualitatively with standard methods well known in the art.
  • mitochondrial marker gene refers to gene product (RNA or protein) of a nuclear or mitochondrial encoded DNA sequence, that forms a component of the mitochondria, and the measurement of which provides an indicator of mitochondrial function.
  • a significant increase in the level or “a significant increase in the expression level” refers to either an increase of the level or the expression level from the control level by an amount greater than the standard error of the assay employed.
  • the term also refers to an increase by at least about 5%, about 10%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, or about 90%, about 100%, about 150%, or about 200%, or greater.
  • the present invention is based on the surprising discovery that ERR ⁇ agonists result in enhance mitochondriogenesis.
  • the present invention provides a method for screening/identifying a compound that binds to a polypeptide containing the ligand-binding domain (LBD) of ERR ⁇ (a ERR ⁇ LBD-containing polypeptide), and upon binding to ERR ⁇ , helps recruiting a PPAR gamma coactivator 1 alpha (PGC- l ⁇ ) polypeptide into contact with the ERR ⁇ LBD-containing polypeptide.
  • the screening is performed by contacting a compound with a biological sample containing the ERR ⁇ LBD-containing polypeptide and the PGC-I ⁇ polypeptide and selecting the compound that binds to the ERR ⁇ LBD- containing polypeptide and recruits the PGC- l ⁇ polypeptide.
  • the ERR ⁇ LBD-containing polypeptide is the ERR ⁇ polypeptide.
  • the binding of the compound can be tested by detecting a direct binding to the ERR ⁇ LBD-containing polypeptide and the close contact of the ERR ⁇ -containing polypeptide with the PGC-I ⁇ polypeptide, or by detecting the signal that indicates the direct binding to ERR ⁇ LBD-containing polypeptide or the close contact of ERR ⁇ LBD- containing polypeptide with the PGC- l ⁇ polypeptide.
  • competition assays provide a suitable format for identifying test compounds that specifically bind to the ERR ⁇ LBD-containing polypeptide.
  • test compounds are screened in competition with a compound already known to bind to the ERR ⁇ LBD-containing polypeptide. If the test compounds inhibit binding of the compound known to bind the ERR ⁇ LBD-containing polypeptide, then the test compounds also bind to the ERR ⁇ LBD-containing polypeptide. 50201A
  • RIA solid phase direct or indirect radioimmunoassay
  • ElA solid phase direct or indirect enzyme immunoassay
  • sandwich competition assay see Stahli et al., Methods in En ⁇ ymology 9:242-253 (1983)
  • solid phase direct biotin-avidin EIA see Kirkland et al., J. Immunol 137:3614-3619 (1986)
  • solid phase direct labeled assay solid phase direct labeled sandwich assay
  • solid phase direct labeled sandwich assay see Harlow and Lane, "Antibodies, A Laboratory Manual '," Cold Spring Harbor Press (1988)
  • solid phase direct label RIA using 125 I label see Morel et al., MoI.
  • Test compounds identified by competition assay include compounds binding to the same epitope as the reference compound and compounds binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference compound for steric hindrance to occur. Usually, when a competing agent is present in excess, it will inhibit specific binding of a reference compound to a common target polypeptide by at least 50 or 75%.
  • FRET fluorescence resonance energy transfer
  • D fluorescence donor
  • A fluorescence acceptor
  • the fluorescence emitted upon excitation of the donor fluorophore will have a different wavelength than that emitted in response to that excitation wavelength when the text 50201 A
  • the screening assay is a ceil-based screening assay.
  • a cell expressing an ERR ⁇ LBD-containing polypeptide and a PGC- l ⁇ polypeptide, or functional fragments thereof is contacted with a test compound, and the ability of the test compound to increase the activity of the ERR ⁇ LBD-containing polypeptide is determined.
  • the activity is transcriptional activity of the ERR ⁇ LBD-containing polypeptide.
  • the ERR ⁇ LBD-containing polypeptide is ERR ⁇ polypeptide.
  • Determining the ability of the test compound to increase the activity of the ERR ⁇ LBD-containing polypeptide can be accomplished by assessing induction of a reporter gene expressed in the cell and comprising a sequence that is recognized by the ERR ⁇ DNA-binding domain (DBD) wherein the reporter gene encodes a detectable polypeptide.
  • the level of the detectable polypeptide is significantly increased in the presence of the test compound relative to that in the absence of the test compound.
  • the detectable polypeptide can be any kind of polypeptide that is detected through standard methods.
  • the detectable polypeptide is selected from chloramphenicol acetyl transferase, green fluorescent protein, or yellow fluorescent protein, or luciferase, ⁇ - galactosidase, secreted alkaline phosphatase, etc.
  • the cell can be a mammalian cell, an insect cell, a bacterial cell, or a yeast cell, etc..
  • determining the ability of the test compound to increase the activity of the ERR ⁇ LBD-containing polypeptide can be accomplished by assessing the increase in the expression levels of the mitochondrial marker genes in cells/tissues.
  • the increased expression levels of mitochondrial marker genes in cells/tissues indicate increased mitochondrial functions, which positively correlate with insulin sensitivity and improvement of anti-diabetic properties of the cells/tissues. See Goodpaster BH et al, 50201A
  • the compound which is ERR ⁇ agonist can be a chemical compound or a polypeptide.
  • the compound is useful for the prevention, delay of progression or the treatment of diabetes preferably type 2 diabetes, insulin resistance, metabolic disease/metabolic syndrome, dyslipidemia, obesity/weight loss, Neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease or Huntington's disease. Furthermore, the compound is useful for improving exercise endurance capacity in a subject.
  • the FRET assay is designed to detect agonist-induced activation of ERR ⁇ in the presence of the coactivator peptide PGC- l ⁇ .
  • the following components were added in a final volume of 50 ⁇ L: (His) 6 -hERR ⁇ LBD or GST-hERRy LBD, Europium-labeled anti- (His) ⁇ antibody or Europium-labeled anti-GST antibody and Cy5 -labeled PGC-I ⁇ peptide (Cy5-RPCSELLKYLTT (SEQ ID NO. 4) with C-terminal acid, custom made and labeled at AnaSpec).
  • Mix 1 contained antibody, hERR ⁇ LBD in buffer in a volume of 19 ⁇ L and was added to 30 ⁇ L of Mix 2 which contained Cy5-peptide in buffer (1. 6xHis- ERR ⁇ -LBD, 3.6mg/ml, MW 27KDa; 2. Cy5-PGC-I peptide from AnaSpec; 3. Other buffer components from Sigma; 4. EU-antiHis ⁇ -Ab from Perkin Elmer; and 5. Test compounds.).
  • the assays were carried out in black 384-well plates and incubated at room temperature for 3hrs before FRET signals were measured using a Wallac Victor 2 (Perkin Elmer) plate reader. The ratio of emission signals (665 nm/615 ran) was used to determine the FRET assay response. In some cases, the FRET signal to background ratio 50201A
  • test compounds were dissolved in DMSO at 10 mM and used as indicated.
  • the FRET response is dose-dependent and an EC 5 0 of 2.1 ⁇ M was derived from the data.
  • Compound 2 also induced a dose-dependent FRET response having a maximal response of 70% at saturating concentrations of compound with an EC5 0 of 0.54 ⁇ M. Both EC 50 's were similar to the reported values of 1.3 ⁇ M and 0.13 ⁇ M for the Compound 1 and Compound 2 equivalents, respectively.
  • Mouse embryonic fibroblast cells were isolated from ERR ⁇ null mice. Briefly, timed heterozygote matings were set up. On day 13 and a half post coiturn, pregnant females was euthanized and the embryos isolated into PBS. Each embryo was individually dissected to remove liver and eye tissue. These tissues were saved for genotyping. The embryos were then individually homogenized in PBS using 3 ml syringes and 18 and a half gauge needles.
  • HeLa cells were transfected as follows. A pre-made transfection reaction mixture was prepared according to manufacturer's instructions. For each well of cells to be transfected, 40 ng of ERE-luciferase reporter (firefly luminescence measurement, Promega), 3 ng of phRL-SV40 (for Renilla luminescence measurement, Promega), 120 ng of total receptor cDNA plasmid (mouse PGC- l ⁇ or human ERR ⁇ or control plasmid, 50201 A
  • Luciferase activity was measured with the Dual-Glo luciferase assay system (Promega catalog number PRE2940) according to manufacturer's instructions. Briefly, 72 hours after transfection, one volume of Dual-Glo luciferase reagent equal to the culture medium volume (90ul per cell for 96 well plate ) was added to each well. After 10 minutes of gently mixing, the firefly luminescence was measured with En Vision (Envision TM 2100 multilabel reader, Perkin Elmer, Wellesley, MA) at 0.1 sec/well. Subsequently, one volume of Dual-Glo Stop Sc GIo reagent was added to each well. Renilla luminescence was measured at the same speed after 10 minutes mixing. The ratio of firefly luminescence over renilla luminescence (for transfection efficiency control) was calculated.
  • HeLa cells or MEFs were trypsinized and counted using a hemocytometer and plated in a 6 well plate at 0.5 x 10 6 cells per well. Cells were then transduced with adenovirus expressing 1) GFP, 2) mouse PGC- l ⁇ , 3) human ERR ⁇ or 4) human ERR ⁇ with mouse PGC- l ⁇ .
  • the ERR ⁇ and PGC-I ⁇ adenoviruses expressed GFP as a secondary transcript through an internal ribosomal entry site in the adenoviral vector.
  • the transduction medium consisted of DMEM (Gibco, catalog number 11965- 092) and 2% FBS (Gibco, catalog number 10082-147).
  • DMEM Gibco, catalog number 11965- 092
  • FBS Gibco, catalog number 10082-147.
  • HeLa cells were transduced 4000 viral particles/cell, while MEFs were infected at a viral titer of 30,000 viral particles/cell.
  • the adenoviral 50201 A When using two adenoviruses in combination, the adenoviral 50201 A
  • titer was doubled, resulting in 8000 viral particles/cell in the case of HeLa cells and 60,000 viral particles/cell in the case of MEFs.
  • the media was replenished 24 hrs after infection.
  • the percent of GFP-positive cells in each well was determined visually with a fluorescence microscope and served as an index for transduction efficiency.
  • Relative mRNA expression levels were calculated comparing the level of expression of target genes to that of B2M of the infected cells to the cells infected with adenoviral GFP. Samples were assayed in quadruplicate and expressed as the mean ⁇ SEM of the fold change relative to the control (set at 100%).
  • Infected cells were lysed using Cell Extraction Buffer (Biosource, catalog number FNNOOl 1) supplemented with Complete Mini protease inhibitor cocktail (Roche, catalog number 1836153) for 30 minutes on ice.
  • the cell lysate was centrifuged at 14,000 rpm for 20 minutes. The supernatant was transferred to a fresh tube and stored at -20 0 C until use.
  • the protein was quantified using the Bio-RAD DC protein Assay kit (Biorad, Hercules, CA, Catalog number 500-0116) according to the manufacturer's instructions. 50201A
  • Cytochrome C protein level was measured by ELISA using an immunoassay kit from R&D Systems (catalog number DCTCO) according to the manufacturer's instructions. Samples were assayed in triplicate and expressed as the mean ⁇ SEM.
  • ERR ⁇ could increase transactivation of an ERE- luciferase reporter in HeLa cells (Figure 3).
  • ERR ⁇ is a stronger inducer of transcription on the ERE in this cell line, with an approximate 50-fold induction with ERR ⁇ as compared to 6 fold induction with ERR ⁇ .
  • PGC-l ⁇ induced ERR ⁇ more strongly than ERR ⁇ (174 fold as compared to 34 fold).
  • Rates of fatty acid oxidation were also increased in the presence of either PGC-I ⁇ alone or both ERR ⁇ and PGC- l ⁇ , implying that the changes in gene expression can also translate into an improvement in mitochondrial function (Figure 6).
  • mice Primary mouse myoblasts from FVB mice were isolated and maintained as described in.. Megeney, L.A., B. Kablar, K. Garrett, J.E. Anderson, and M.A. Rudnicki. 1996. MyoD is required for myogenic stem cell function in adult skeletal muscle. Genes Dev. 10: 1173-1183
  • mouse myoblasts were grown to 80% confluence in F-10/Ham's media containing 20% fetal bovine serum, 1% Penicillin/Streptomycin and2.5 ng/mL bFGF (human recombinant). The cells were then plated to 700,000 cells per well in 6-well plates, and allowed to differentiate into myo tubes for 36 to 48 hours in DMEM containing 5% equine serum and 50201A
  • RNA samples were treated with the ERR ⁇ / ⁇ agonist Compound 1 for 24 hours.
  • Assays performed included analysis of gene expression by real time quantitative PCR (RT-PCR), cytochrome c ELISA, citrate synthase assay, fatty acid oxidation assay, and a respiration assay.
  • RT-PCR real time quantitative PCR
  • cytochrome c ELISA and citrate synthase assay myotubes were treated with Compound 1 at the following concentrations: 1 ⁇ M, 3 ⁇ M, 10 ⁇ M, and 30 ⁇ M.
  • For the fatty acid oxidation assay myotubes were treated with Compound 1 at the following concentrations: 10 ⁇ M, and 30 ⁇ M.
  • RNA was isolated from cell lysates, and cDNA was subsequently synthesized from this RNA.
  • RNA isolation cells were homogenized in TRIzol (Invitrogen, catalog number 15596-026, Carlsbad, CA), arid total RNA was isolated following the manufacturer's instructions. RNA was quantified using the spectrophotometer.
  • Reverse transcription was performed using the BD SprintTM PowerScriptTM kit from BD Biosciences (catalog number 639562). Quantitative real time PCR for the following genes was performed using Assay-on-Demand primer probes from Applied Biosystems (See Post-text Table 8-1 for catalog numbers): B2M, ERR ⁇ , ERR ⁇ , ERR ⁇ , PGC-l ⁇ , PGC-l ⁇ , PPAR ⁇ , PPAR ⁇ , COX-4, cytochrome c, UQCRB, CPT-Ib, LCAD, MCAD, lDH3a, ATP-5b, UCP-2, and UCP-3.
  • Taqman real time quantitative PCR was performed and analyzed following the manufacturer's instructions (Applied Biosystems). Specifically, amplification was performed in triplicate, in a 10 ⁇ l reaction mixture.
  • the reaction mixture included: IX TaqMan® Universal PCR Master Mix (Applied Biosystems, catalog number 4304437), IX of Assay-on-Demand primer probe, and 2 ⁇ l of cDNA sample. Gene expression was calculated by normalizing to total cDNA as measured by B2M endogenous control (Applied Biosystems, catalog number Mm00437762_ml). The samples were initially 50201A
  • cytochrome c enzyme-linked immunosorbent assay was performed using the Rat/Mouse Cytochrome c Immunoassay kit from R&D Systems (catalog number MCTCO, R&D Systems, Minneapolis, MN) following the manufacturer's instructions.
  • citrate synthase assay was performed following the procedure described in above in this application.
  • the cells were washed once with the medium and pelleted before being resuspended in the assay buffer containing 25 mM glucose (Sigma, catalog number G-5400), 1 mM pyruvate (Invitrogen, catalog number 11360-070) and 2% bovine serum albumin (BSA) (MP Biomedicals, catalog number 103703) in D-PBS (Invitrogen, catalog number 14040-133).
  • 50201A bovine serum albumin
  • the cell suspension was diluted to IxIO 6 cells per ml in the assay buffer and kept at 37 0 C until used. Oxygen consumption was measured with a Clark electrode according to the instructions provided by Hansatech (Norfolk, UK). One half of the cell suspension was used for each measurement.
  • the concentrations of oligomycin (MP Biomedicals, catalog number 151786) and FCCP (4-(trifluoromethoxy) carbonyl cyanide phenyl hydrazone) (Sigma, catalog number C2920) were 2 ⁇ g/ml and 2 to 5 ⁇ M, respectively.
  • the experiments were performed in triplicate. The rate of respiration was measured by calculating the slope of of the flux of oxygen consumption using the software provided by the manufacturer.
  • IDH3 ⁇ a component of the Krebs cycle
  • ATP-5b an ATP-synthesizing enzyme
  • the co-activators PGC- l ⁇ and PGC- l ⁇ were also examined for gene expression.
  • the expression of these transcripts showed a similar dose-dependent increase in expression, with PGC-I ⁇ elevated to 1.9-fold expression and PGC-I ⁇ elevated to 1.8-fold (Figure 19).
  • Cytochrome c is a critical element of the electron transport chain, and quantities of the protein serve as a biomarker for mitochondrial number and oxidative phosphorylation activity.
  • citrate synthase activity was determined. This enzyme is often used as an indicator of mitochondrial content or activity in human muscle (Kelley, et al., Diabetes, 51(10): 2944-50 (2002)). Citrate synthase catalyzes the initial step of the Krebs cycle, which supplies substrate for oxidative phosphorylation. Myotubes treated with 30 ⁇ M Compound 1 for 24 hours showed a 28% increase in citrate synthase activity, while 10 ⁇ M and 3 ⁇ M treatment showed an increase of 8% and 9% respectively (Figure 22).

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Pathology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Endocrinology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Reproductive Health (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present invention relates to the novel method for identifying an ERRγ agonist that enhances the mitochondriogenesis. The ERRγ agonist can be a chemical compound or a polypeptide. The present invention further relates to the use of the ERRγ agonist in the treatment of diseases selected from diabetes, type 2 diabetes, insulin resistance, metabolic disease/metabolic syndrome, dyslipidemia, obesity/overweight, neurodegenerative diseases. Furthermore, the compound is useful for improving exercise endurance capacity in a subject.

Description

50201A
Function of Estrogen Related Receptor Gamma (£RRγ) in Increasing
Mitochondrial Biogenesis
TECHNICAL FIELD
The present invention relates to methods of identifying ERRγ agonists and the obtained agonists in the treatment of diseases whose conditions are improved by activation of ERRγ.
BACKGROUND
The estrogen-related receptors (ERR) comprise 3 members, ERRα, ERRβ, and ERRγ, that form a subfamily of orphan nuclear receptors for which natural ligands have yet to be identified. The ERRs share significant amino acid homology with the estrogen receptor (ER) within their DNA binding domains (DBD) and ligand binding domains (LBD), but do not respond to estradiol (Giguere, et al., Trends Endocrinol Metab., 13:220-5 (2002); Heard, et al., MoL Endocrinol, 14:382-92(2000).). Whereas ERs are ligand-activated receptors, ERRs can activate gene transcription in a constitutive manner (Hong, et al., J. Bio. Chem., 21 A: 22618-26 (1999); Chen, et al., J. Bio. Chem., 276:28465-70 (2001).) and their activation ability may be determined by the presence of transcriptional coactivators (Kamei, et al., PNAS USA, 100:12378-83(2003); Schreiber, et al., J. Bio. Chem., 278:9013-8 (2003).). Structural studies confirm this finding by demonstrating that the ERRγ LBD can adopt a transcriptionally active conformation and interact with the steroid receptor coactivator 1 (SRC-I) in the absence of any ligand (Greschik, et al., M?/. Cell, 9:303-13 (2002).).
ERRγ is highly expressed in adult tissue such as the brain, skeletal muscle, heart, kidney and retina. It is also expressed in fetal tissue such as the placenta, brain, heart, skeletal muscle, kidney and lung (Bonnelye et al. Mot. Endo., 11: 905-16 (1997).).
SUMMARY OF THE INVENTION
In one aspect, the present invention provides a method for screening/identifying a compound that binds to a polypeptide containing the ligand-binding domain (LBD) of 50201A
ERRγ (a ERRγ LBD-containing polypeptide), and upon binding to ERRy, helps recruiting a PPAR gamma coactivator 1 alpha (PGC- lα) polypeptide into contact with the ERRγ LBD-containing polypeptide. The screening is performed by contacting a compound with a biological sample containing the ERRγ LBD-containing polypeptide and the PGC- lα polypeptide and selecting the compound that binds to the ERRγ LBD- containing polypeptide and recruits the PGC- lα polypeptide. In one embodiment, the ERRγ LBD-containing polypeptide is the ERRγ polypeptide.
In another aspect, the screening assay is a cell-based screening assay. In the cell- based screening assay, a cell expressing an ERRγ LBD-containing polypeptide and a PGC- lα polypeptide, or functional fragments thereof, is contacted with a test compound, and the ability of the test compound to increase the activity of the ERRγ LBD-containing polypeptide is determined. Preferably, the activity is transcriptional activity of the ERRγ LBD-containing polypeptide. Also preferably, the ERRγ LBD-containing polypeptide is ERRγ polypeptide.
BRIEF DESCRIPTION OF FIGURES
Figure 1 shows the diagram of FRET assay configuration.
Figure 2 shows the agonist-induced FRET response.
Figure 3 shows comparison of the ability of ERRα and ERRγ to transactivate a ERE reporter in the presence and absence of PGC-I α.
Figure 4 shows adenoviral overexpression of PGC-I α and ERRγ in HeIa cells.
Figures 5 A and 5B show the expression of mitochondrial marker genes in HeIa cells over expressing PGC-lα and ERRγ.
Figure 6 shows effects of overexpression of PGC-lα and ERRγ on fatty acid oxidation in HeIa cells.
Figure 7 shows effects of overexpression of PGC-I alpha and ERR gamma on expression of PGC-I alpha and ERRγ in ERRα null MEFs. 50201A
Figure 8 shows effect of overexpression of PGC-I alpha and ERR gamma on expression of mitochondrial markers in ERR alpha null MEFs.
Figure 9 shows effect of overexpression of PGC-I alpha and ERR gamma on expression of genes of oxidative stress protection in ERR alpha null MEFs.
Figure 10 shows effects of overexpression of ERR gamma and PGC-I alpha on cytochrome c protein levels in ERR alpha null MEFs.
Figure 11 shows the human ERRγ polypeptide sequence (SEQ ID NO. 1).
Figure 12 shows the LBD domain sequence of human ERRγ sequence (SEQ ID NO. 2).
Figure 13 shows the ERRγ ERE sequence (SEQ ID NO. 3).
Figure 14 shows expression levels of genes of oxidative phosphorylation in mouse myotubes treated with Compound 1.
Figure 15 shows expression levels of genes of fatty acid oxidation in mouse myotubes treated with Compound 1.
Figure 16 shows expression levels of IDH3α and ATP-5b in mouse myotubes treated with Compound 1.
Figure 17 shows expression levels of UCP2 and UCP3 in mouse myotubes treated with Compound 1.
Figure 18 shows expression levels of ERRγ and ERRα in mouse myotubes treated with Compound 1.
Figure 19 shows expression levels of PGC-I α and PGC- lβ in mouse myotubes treated with Compound 1.
Figure 20 shows expression levels of PP ARa, PPARγ, and PPARδ in mouse myotubes treated with Compound 1. 50201A
Figure 21 shows protein expression levels of cytochrome c in mouse myotubes treated with Compound 1.
Figure 22 shows citrate synthase activity in mouse myotubes treated with Compound 1.
Figure 23 shows measurement of cellular respiration in primary mouse myotubes treated with Compound 1.
DETAILED DESCRIPTION
Definitions
As used herein, the term "agonist of ERRγ" refers to a molecule which when bound to the LBD sequence of ERRγ, increases the amount of, or prolongs the duration of, or enhances the activity of ERRγ. Agonists can include polypeptides, nucleic acids, carbohydrates, lipids, or any derivatives thereof, or any other molecules.
As used herein, the term "a sample" refers to a whole organism or a subset of its tissues, cells or component parts (e.g. body fluids, including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen). "A sample" further refers to a homogenate, lysate or extract prepared from a whole organism or a subset of its tissues, cells or component parts, or a fraction or portion thereof, including but not limited to, for example, plasma, serum, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs. Most often, the sample has been removed from an animal, but the term "sample" can also refer to cells or tissue analyzed in vivo, i.e., without removal from animal. Typically, "a sample" will contain cells from the animal, but the term can also refer to non-cellular biological material, such as non-cellular fractions of blood, saliva, or urine, that can be used to measure the cancer-associated polynucleotide or polypeptides levels. "A sample" further refers to a medium, such as a nutrient broth or gel in which an organism has been propagated, which contains cellular components, such as proteins or nucleic acid molecules. 50201 A
As used herein, the term "polypeptide" refers to a polymer in which the monomers are amino acids and are joined together through peptide or disulfide bonds. It also refers to either a full-length naturally-occurring amino acid sequence or a fragment thereof between about 8 and about 500 amino acids in length. Additionally, unnatural amino acids, for example, beta-alanine, phenyl glycine and homoarginine may be included. Commonly-encountered amino acids which are not gene-encoded may also be used in the present invention. All of the amino acids used in the present invention may be either the D- or L-optical isomer. The L-isomers are preferred. A polypeptide sequence also encompasses naturally-occurring allelic variants of said polypeptide.
As used herein, the term "detectable polypeptide" refers to a polypeptide whose presence or absence can be measured directly or indirectly, quantitatively or qualitatively with standard methods well known in the art.
As used herein, the term "mitochondrial marker gene" refers to gene product (RNA or protein) of a nuclear or mitochondrial encoded DNA sequence, that forms a component of the mitochondria, and the measurement of which provides an indicator of mitochondrial function.
As used herein, the term "a significant increase in the level" or "a significant increase in the expression level" refers to either an increase of the level or the expression level from the control level by an amount greater than the standard error of the assay employed. The term also refers to an increase by at least about 5%, about 10%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, or about 90%, about 100%, about 150%, or about 200%, or greater.
As used herein, the term "a," "an," "the" and similar terms used in the context of the present invention (especially in the context of the claims) are to be construed to cover both the singular and plural unless otherwise indicated herein or clearly contradicted by the context. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. All methods described herein can 50201A
be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g. "such as") provided herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention.
The present invention is based on the surprising discovery that ERRγ agonists result in enhance mitochondriogenesis.
In one aspect, the present invention provides a method for screening/identifying a compound that binds to a polypeptide containing the ligand-binding domain (LBD) of ERRγ (a ERRγ LBD-containing polypeptide), and upon binding to ERRγ, helps recruiting a PPAR gamma coactivator 1 alpha (PGC- lα) polypeptide into contact with the ERRγ LBD-containing polypeptide. The screening is performed by contacting a compound with a biological sample containing the ERRγ LBD-containing polypeptide and the PGC-I α polypeptide and selecting the compound that binds to the ERRγ LBD- containing polypeptide and recruits the PGC- lα polypeptide. In one embodiment, the ERRγ LBD-containing polypeptide is the ERRγ polypeptide.
To assay the binding described herein, a number of methods known in the art can be employed. The binding of the compound can be tested by detecting a direct binding to the ERRγ LBD-containing polypeptide and the close contact of the ERRγ-containing polypeptide with the PGC-I α polypeptide, or by detecting the signal that indicates the direct binding to ERRγ LBD-containing polypeptide or the close contact of ERRγ LBD- containing polypeptide with the PGC- lα polypeptide.
For purposes of detecting a direct binding, competition assays provide a suitable format for identifying test compounds that specifically bind to the ERRγ LBD-containing polypeptide. In such formats, test compounds are screened in competition with a compound already known to bind to the ERRγ LBD-containing polypeptide. If the test compounds inhibit binding of the compound known to bind the ERRγ LBD-containing polypeptide, then the test compounds also bind to the ERRγ LBD-containing polypeptide. 50201A
Numerous types of competitive binding assays are known, for example: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (ElA), sandwich competition assay (see Stahli et al., Methods in En∑ymology 9:242-253 (1983)); solid phase direct biotin-avidin EIA (see Kirkland et al., J. Immunol 137:3614-3619 (1986)); solid phase direct labeled assay, solid phase direct labeled sandwich assay (see Harlow and Lane, "Antibodies, A Laboratory Manual '," Cold Spring Harbor Press (1988)); solid phase direct label RIA using 125I label (see Morel et al., MoI. Immunol 25(1) :7-15 (1988)); solid phase direct biotin-avidin EIA (Cheung et al., Virology 176:546-552 (1990)); and direct labeled RIA (Moldenhauer et al., Scand. J. Immunol. 32:77-82 (1990)). Typically, such an assay involves the use of purified polypeptide bound to a solid surface or cells bearing either of these, an unlabelled test compound and a labeled reference compound. Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test compound. Usually the test compound is present in excess. Test compounds identified by competition assay include compounds binding to the same epitope as the reference compound and compounds binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference compound for steric hindrance to occur. Usually, when a competing agent is present in excess, it will inhibit specific binding of a reference compound to a common target polypeptide by at least 50 or 75%.
For purposes of detecting the signal that indicates the direct binding of the compound to the ERRγ LBD-containing polypeptide and consequently the close contact of the ERRγ LBD-containing polypeptide with the PGC-lα polypeptide, fluorescence resonance energy transfer (FRET) can be employed. FRET is a quantum mechanical phenomenon that occurs between a fluorescence donor (D) and a fluorescence acceptor (A) in close proximity to each other (usually <100 A of separation) if the emission spectrum of D overlaps with the excitation spectrum of A. The molecules to be tested, e.g. the ERRγ LBD-containing polypeptide and the PGC-lα polypeptide, are labeled with a complementary pair of donor and acceptor fluorophores. While bound closely together by the ERRγ-LBD/PGC-lα interaction in the presence of the test compound, the fluorescence emitted upon excitation of the donor fluorophore will have a different wavelength than that emitted in response to that excitation wavelength when the text 50201 A
compound is not present, providing for quantitation of bound versus unbound molecules by measurement of emission intensity at each wavelength. See also Zhou et al., "Nuclear Receptors have distinct affinities for coactivators: characterization by fluorescence resonance energy transfer," MoL Endocrin., 12: 1594-1604 (1998).
In another aspect, the screening assay is a ceil-based screening assay. In the cell- based screening assay, a cell expressing an ERRγ LBD-containing polypeptide and a PGC- lα polypeptide, or functional fragments thereof, is contacted with a test compound, and the ability of the test compound to increase the activity of the ERRγ LBD-containing polypeptide is determined. Preferably, the activity is transcriptional activity of the ERRγ LBD-containing polypeptide. Also preferably, the ERRγ LBD-containing polypeptide is ERRγ polypeptide.
Determining the ability of the test compound to increase the activity of the ERRγ LBD-containing polypeptide can be accomplished by assessing induction of a reporter gene expressed in the cell and comprising a sequence that is recognized by the ERRγ DNA-binding domain (DBD) wherein the reporter gene encodes a detectable polypeptide. The level of the detectable polypeptide is significantly increased in the presence of the test compound relative to that in the absence of the test compound. The detectable polypeptide can be any kind of polypeptide that is detected through standard methods. Preferably, the detectable polypeptide is selected from chloramphenicol acetyl transferase, green fluorescent protein, or yellow fluorescent protein, or luciferase, β- galactosidase, secreted alkaline phosphatase, etc. The cell can be a mammalian cell, an insect cell, a bacterial cell, or a yeast cell, etc..
Alternatively, determining the ability of the test compound to increase the activity of the ERRγ LBD-containing polypeptide can be accomplished by assessing the increase in the expression levels of the mitochondrial marker genes in cells/tissues. The increased expression levels of mitochondrial marker genes in cells/tissues indicate increased mitochondrial functions, which positively correlate with insulin sensitivity and improvement of anti-diabetic properties of the cells/tissues. See Goodpaster BH et al, 50201A
Diabetes, 52: 2191-2197, 2003; Menshikova et al, American J Physiol Endocrinol Metab 288: E818 - E825, 2005.
Another aspect of the present invention pertains to the compound obtained from the above screening assays. The compound which is ERRγ agonist, can be a chemical compound or a polypeptide. The compound is useful for the prevention, delay of progression or the treatment of diabetes preferably type 2 diabetes, insulin resistance, metabolic disease/metabolic syndrome, dyslipidemia, obesity/weight loss, Neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease or Huntington's disease. Furthermore, the compound is useful for improving exercise endurance capacity in a subject.
EXAMPLES
The examples below are non-limiting and are merely representative of various aspects and features of the present invention.
Example 1
FRET Assay to Detect Binding
The FRET assay is designed to detect agonist-induced activation of ERRγ in the presence of the coactivator peptide PGC- lα. The following components were added in a final volume of 50 μL: (His)6-hERRγ LBD or GST-hERRy LBD, Europium-labeled anti- (His)β antibody or Europium-labeled anti-GST antibody and Cy5 -labeled PGC-I α peptide (Cy5-RPCSELLKYLTT (SEQ ID NO. 4) with C-terminal acid, custom made and labeled at AnaSpec). Mix 1 contained antibody, hERRγ LBD in buffer in a volume of 19 μL and was added to 30 μL of Mix 2 which contained Cy5-peptide in buffer (1. 6xHis- ERRγ-LBD, 3.6mg/ml, MW 27KDa; 2. Cy5-PGC-I peptide from AnaSpec; 3. Other buffer components from Sigma; 4. EU-antiHisβ-Ab from Perkin Elmer; and 5. Test compounds.). The assays were carried out in black 384-well plates and incubated at room temperature for 3hrs before FRET signals were measured using a Wallac Victor 2 (Perkin Elmer) plate reader. The ratio of emission signals (665 nm/615 ran) was used to determine the FRET assay response. In some cases, the FRET signal to background ratio 50201A
was used and this is simply defined as the FRET ratio in the presence of protein or FRET ratio in the absence of protein. The test compounds were dissolved in DMSO at 10 mM and used as indicated.
Table 1 Reagent List
Reagent Vendor Catalog #
Europium-labeled anti-(His)6 antibody Perkin Elmer ADOI lO Europium-labeled anti-GST antibody Perkin Elmer AD0254 Cy5-labeled PGC- lα peptide AnaSpec Custom Order
(Cy5-RPCSELLKYLTT (SEQ ID NO. 4); C-terminal acid of PGC-I α)
Dithiothreitol Sigma D-0632
Tris Sigma T-2194
384-well plates Corning 3654
The ability of agonist compounds to induce an increased FRET response between the Europium-labeled anti-(His)6 antibody/(His)6-hERRγ LBD complex and Cy5- RPCSELLKYLTT (SEQ ID NO. 4) was assessed by using two compounds described in the literature as ERRγ agonists, GSK4716 (Glaxo-Smith-Kline; Compound 1) and GSK9089 (Glaxo-Smith-Kline; Compound 2). See Zuercher WJ5 Gaillard S, Miller- Orband LA, et al. (2005), "Identification and structure-activity relationship of phenolic acyl hydrazones as selective agonists for the estrogen-related orphan nuclear receptors ERRβ and ERRγ," J Med Chem; 48:3107-9.).
Figure imgf000011_0001
Compound 1 Compound 2
The compounds were solubilized in 10 mM DMSO and the concentration range of the titration was from 100 μM down to 2 nM. The final DMSO concentration was 2%. A DMSO control (no compound) at 2% was also prepared. All samples were made in duplicate. As shown in Figure 2, Compound 1 was able to induce up to a 60% increase in 50201A
the FRET signal at the highest concentration tested. The FRET response is dose- dependent and an EC50 of 2.1 μM was derived from the data. Similarly, Compound 2 also induced a dose-dependent FRET response having a maximal response of 70% at saturating concentrations of compound with an EC50 of 0.54 μM. Both EC50's were similar to the reported values of 1.3 μM and 0.13 μM for the Compound 1 and Compound 2 equivalents, respectively.
Example 2
Cell-Based Assay
Materials and Methods
Mouse embryonic fibroblasts
Mouse embryonic fibroblast cells (MEFs) were isolated from ERRα null mice. Briefly, timed heterozygote matings were set up. On day 13 and a half post coiturn, pregnant females was euthanized and the embryos isolated into PBS. Each embryo was individually dissected to remove liver and eye tissue. These tissues were saved for genotyping. The embryos were then individually homogenized in PBS using 3 ml syringes and 18 and a half gauge needles. These crude suspensions were the.n plated in 10 cm cell culture dishes containing DMEM (Gibco, catalog number 11965-092), 10% FBS (Gibco catalog number 10082-147) and 1% Penicillin / Streptomycin (Gibco catalog number 15140-122). After two days in culture, cells were passaged. At the second passage, similar genotypes were pooled, and cryopreserved in FBS containing 10% DMSO (Sigma, catalog number D2650).
Transfection
HeLa cells were transfected as follows. A pre-made transfection reaction mixture was prepared according to manufacturer's instructions. For each well of cells to be transfected, 40 ng of ERE-luciferase reporter (firefly luminescence measurement, Promega), 3 ng of phRL-SV40 (for Renilla luminescence measurement, Promega), 120 ng of total receptor cDNA plasmid (mouse PGC- lα or human ERRγ or control plasmid, 50201 A
and 0.325 μl of Fugene 6 transfection reagent (Roche catalog number NC9167392) were added to 10 μl of Opti-MEM (Gibco catalog number 31985-062). When a single receptor plasmid was transfected, the total amount was made up with control plasmid to 120 ng. This mixture was added into each well of a 96 well assay plate (Fisher catalog number 07200566). Cells (8000/well) were subsequently added to each well. The cells were incubated at 37°C for 72 hours.
Luciferase assay
Luciferase activity was measured with the Dual-Glo luciferase assay system (Promega catalog number PRE2940) according to manufacturer's instructions. Briefly, 72 hours after transfection, one volume of Dual-Glo luciferase reagent equal to the culture medium volume (90ul per cell for 96 well plate ) was added to each well. After 10 minutes of gently mixing, the firefly luminescence was measured with En Vision (Envision ™ 2100 multilabel reader, Perkin Elmer, Wellesley, MA) at 0.1 sec/well. Subsequently, one volume of Dual-Glo Stop Sc GIo reagent was added to each well. Renilla luminescence was measured at the same speed after 10 minutes mixing. The ratio of firefly luminescence over renilla luminescence (for transfection efficiency control) was calculated.
Adenoviral transduction
HeLa cells or MEFs were trypsinized and counted using a hemocytometer and plated in a 6 well plate at 0.5 x 106 cells per well. Cells were then transduced with adenovirus expressing 1) GFP, 2) mouse PGC- lα, 3) human ERRγ or 4) human ERRγ with mouse PGC- lα. The ERRγ and PGC-I α adenoviruses expressed GFP as a secondary transcript through an internal ribosomal entry site in the adenoviral vector.
The transduction medium consisted of DMEM (Gibco, catalog number 11965- 092) and 2% FBS (Gibco, catalog number 10082-147). For each adenovirus, HeLa cells were transduced 4000 viral particles/cell, while MEFs were infected at a viral titer of 30,000 viral particles/cell. When using two adenoviruses in combination, the adenoviral 50201 A
titer was doubled, resulting in 8000 viral particles/cell in the case of HeLa cells and 60,000 viral particles/cell in the case of MEFs. The media was replenished 24 hrs after infection. The percent of GFP-positive cells in each well was determined visually with a fluorescence microscope and served as an index for transduction efficiency.
JRNA preparation and real time quantitative PCR analysis of gene expression
Total RNA was extracted from HeLa or MEF cells using Trizol (Invitrogen, catalog number 15596-026) according to the instructions provided by the supplier. The RNA concentration and purity were determined using an Agilent 2100 Bioanalyzer according to the manufacturer's manual. A 2 μg aliquot of total RNA was used for cDNA synthesis performed with Superscript III RNaseH Reverse Transcriptase (Invitrogen, catalog number 18080-044) according to the manufacturer's manual. The synthesized cDNA was diluted 1 :4 in water to a final volume of 200 μl and stored at -200C until used. Quantitative real time PCR was performed using Taqman (Applied Biosystems) as well known in the art. (Jemiolo and Trappe, BBRC, 320: 1043-1050, 2004). Relative mRNA expression levels were calculated comparing the level of expression of target genes to that of B2M of the infected cells to the cells infected with adenoviral GFP. Samples were assayed in quadruplicate and expressed as the mean ± SEM of the fold change relative to the control (set at 100%).
Protein extraction
Infected cells were lysed using Cell Extraction Buffer (Biosource, catalog number FNNOOl 1) supplemented with Complete Mini protease inhibitor cocktail (Roche, catalog number 1836153) for 30 minutes on ice. The cell lysate was centrifuged at 14,000 rpm for 20 minutes. The supernatant was transferred to a fresh tube and stored at -200C until use. The protein was quantified using the Bio-RAD DC protein Assay kit (Biorad, Hercules, CA, Catalog number 500-0116) according to the manufacturer's instructions. 50201A
Cytochrome CELISA
The Cytochrome C protein level was measured by ELISA using an immunoassay kit from R&D Systems (catalog number DCTCO) according to the manufacturer's instructions. Samples were assayed in triplicate and expressed as the mean ± SEM.
Results
We investigated whether ERRγ could increase transactivation of an ERE- luciferase reporter in HeLa cells (Figure 3). In comparison to ERRγ, ERRγ is a stronger inducer of transcription on the ERE in this cell line, with an approximate 50-fold induction with ERRγ as compared to 6 fold induction with ERRα. On addition of exogenous PGC-lα, this induction further increased (174 fold in the presence of PGC-lα compared to 50 fold without). PGC-lα induced ERRγ more strongly than ERRα (174 fold as compared to 34 fold).
We wanted to examine the effects of overexpression of ERRγ on mitochondrial function in HeLa cells. We infected HeLa cells with adenoviral vectors expressing either PGC-lα alone, ERRγ alone or ERRγ and PGC-lα together. As expected, PGC-lα was levels were induced in cells that were infected with PGC-lα virus, and ERRγ levels were increased in cells infected with ERRγ virus (Figure 4). In the presence of both ERRγ and PGC-lα, PGC-lα was more highly expressed than in cells transduced with PGC-lα alone. This is possibly due to an enhancement in the feed forward mechanism exerted by the presence of both exogenous ERRγ and PGC-lα on the endogenous mouse PGC-lα gene. The levels of adenoviral ERRγ were significantly lower in the cells transduced with both ERRγ and PGC-lα.
We next examined the levels of mitochondrial gene expression in HeLa cells infected with either PGC-I α alone, ERRγ alone or ERRγγ and PGC-lα together. Interestingly, while the levels of cytochrome C, ERRα, COX-4 and IDH3 were induced in cells that were infected with PGC-lα virus, they were further enhanced in cells containing both ERRγ and PGC-lα. ERRγ overexpression alone did not change levels of 5020 IA
these genes in HeLa cells (Figures 5 A and 5B). This could be due to low levels of PGC- lα expression in HeLa cells.
Rates of fatty acid oxidation were also increased in the presence of either PGC-I α alone or both ERRγ and PGC- lα, implying that the changes in gene expression can also translate into an improvement in mitochondrial function (Figure 6).
Thus, we have shown that ERRγ increases mitochondrial gene expression as well function in HeLa cells. However, there is a concurrent increase in of ERRα levels in these cell. It is possible that the effects of ERRγ are secondary to this effect. Therefore, we performed a series of similar experiments in ERRα null MEFs. Interestingly, we found that ERRγ in combination with PGC-I α could increase mitochondrial gene expression (COX-4 and IDH3) in ERRα null MEFs (Figure 8). To verify that these cells overexpressed PGC-I α and ERRγ, we measured the expression of these transcripts as well (Figure T). Both of these transcripts were several fold overexpressed in the appropriate cells. Further, we found that the expression of genes of the oxidative stress protection pathway (SOD2, PRX3, PRX5, TXN2 and TXNRD2) were also induced in the presence of ERRγ and PGC-Iq together (Figure 9).
Overexpression of ERRγ as well as ERRγ in combination with PGC-lα increased cytochrome c protein levels in ERRα null MEFs (Figure 10).
Example 3
Characterization of Compound 1 on mitochondrial function in primary mouse mvotubes
Primary mouse myoblasts from FVB mice were isolated and maintained as described in.. Megeney, L.A., B. Kablar, K. Garrett, J.E. Anderson, and M.A. Rudnicki. 1996. MyoD is required for myogenic stem cell function in adult skeletal muscle. Genes Dev. 10: 1173-1183 For experimentation, mouse myoblasts were grown to 80% confluence in F-10/Ham's media containing 20% fetal bovine serum, 1% Penicillin/Streptomycin and2.5 ng/mL bFGF (human recombinant). The cells were then plated to 700,000 cells per well in 6-well plates, and allowed to differentiate into myo tubes for 36 to 48 hours in DMEM containing 5% equine serum and 50201A
Penicillin/Streptomycin. The cells were treated with the ERRγ/β agonist Compound 1 for 24 hours. Assays performed included analysis of gene expression by real time quantitative PCR (RT-PCR), cytochrome c ELISA, citrate synthase assay, fatty acid oxidation assay, and a respiration assay. For the RT-PCR, cytochrome c ELISA and citrate synthase assay, myotubes were treated with Compound 1 at the following concentrations: 1 μM, 3 μM, 10 μM, and 30 μM. For the fatty acid oxidation assay, myotubes were treated with Compound 1 at the following concentrations: 10 μM, and 30 μM. For the respiration assay, myotubes were treated with Compound 1 at a concentration of 30 μM. Each dose was tested in trplicate, and a set of wells treated with an equivalent volume of DMSO used was included in every experimental readout for control purposes.
For the purpose of assessing gene expression by RT-PCR, RNA was isolated from cell lysates, and cDNA was subsequently synthesized from this RNA. For RNA isolation, cells were homogenized in TRIzol (Invitrogen, catalog number 15596-026, Carlsbad, CA), arid total RNA was isolated following the manufacturer's instructions. RNA was quantified using the spectrophotometer.
Reverse transcription was performed using the BD Sprint™ PowerScript™ kit from BD Biosciences (catalog number 639562). Quantitative real time PCR for the following genes was performed using Assay-on-Demand primer probes from Applied Biosystems (See Post-text Table 8-1 for catalog numbers): B2M, ERRγ, ERRβ, ERRα, PGC-lα, PGC-lβ, PPARα, PPARγ, PPARδ, COX-4, cytochrome c, UQCRB, CPT-Ib, LCAD, MCAD, lDH3a, ATP-5b, UCP-2, and UCP-3.
Taqman real time quantitative PCR was performed and analyzed following the manufacturer's instructions (Applied Biosystems). Specifically, amplification was performed in triplicate, in a 10 μl reaction mixture. The reaction mixture included: IX TaqMan® Universal PCR Master Mix (Applied Biosystems, catalog number 4304437), IX of Assay-on-Demand primer probe, and 2 μl of cDNA sample. Gene expression was calculated by normalizing to total cDNA as measured by B2M endogenous control (Applied Biosystems, catalog number Mm00437762_ml). The samples were initially 50201A
incubated for 2 min at 500C for optimum uracyl-N-glycosylase activity. The PCR program started with a 950C denaturing for 10 min, followed by 40 cycles of 95°C/15 sec and 60°C/l min in a 384-well thermal cycler (Perkin-Elmer Applied Biosystems). Each amplification run contained "No Template" controls (buffer and primers only). Amplification data were collected by the 7700 Sequence Detector and analyzed using the Sequence Detection System software developed by Perkin-Elmer (Applied Biosystems). The fractional cycle number reflecting a positive PCR result is called the cycle threshold (Ct).
Average gene expression values were calculated for each group relative to B2M, using 2'&ACt (as described by Applied Biosystems, Foster City, CA) with the expression in the vehicle treated cells (0 μM) normalized to a value of 1. The data are expressed as mean ± SEM (n = 3 replicate wells). Statistical significance was measured using Student' s t test.
The cytochrome c enzyme-linked immunosorbent assay (ELISA) was performed using the Rat/Mouse Cytochrome c Immunoassay kit from R&D Systems (catalog number MCTCO, R&D Systems, Minneapolis, MN) following the manufacturer's instructions.
The citrate synthase assay was performed following the procedure described in above in this application.
Respiration was measured in myotubes incubated with Compound 1 for 24 hrs according to the published method with modifications (St-Pierre, et al., JBC, 278(29): 26597-603 (2003)). Cells were washed once with phosphate-buffered saline (PBS) and treated for 5 min at 37°C with 2 ml of trypsin (Mediatech, catalog number 25-052-CI). Without removing trypsin, 10 ml of DMEM plus 10% FBS was added into each well. The cells were transferred to a 15 ml tube and centrifuged for 5 min at 1000 rpm. The cells were washed once with the medium and pelleted before being resuspended in the assay buffer containing 25 mM glucose (Sigma, catalog number G-5400), 1 mM pyruvate (Invitrogen, catalog number 11360-070) and 2% bovine serum albumin (BSA) (MP Biomedicals, catalog number 103703) in D-PBS (Invitrogen, catalog number 14040-133). 50201A
The cell suspension was diluted to IxIO6 cells per ml in the assay buffer and kept at 370C until used. Oxygen consumption was measured with a Clark electrode according to the instructions provided by Hansatech (Norfolk, UK). One half of the cell suspension was used for each measurement. The concentrations of oligomycin (MP Biomedicals, catalog number 151786) and FCCP (4-(trifluoromethoxy) carbonyl cyanide phenyl hydrazone) (Sigma, catalog number C2920) were 2 μg/ml and 2 to 5 μM, respectively. The experiments were performed in triplicate. The rate of respiration was measured by calculating the slope of of the flux of oxygen consumption using the software provided by the manufacturer.
Data manipulations and graph generation were performed using Microsoft Excel and GraphPad Prism 4 software. Data are presented as mean ± SEM. In all experiments, each dose was tested in triplicate. The only exception to this were the respiration measurements where the experiment was repeated three times. Statistical analysis was preformed using a two-tailed Student's t test.
Results
We examined the expression of various markers of mitochondrial gene expression in differentiated mouse myotubes treated with Compound 1 at the following concentrations: 30 μM, 10 μM, 3 μM and 1 μM. Stock solutions were prepared such that a constant volume of drug solution was added to each well of cells. All effects were compared to that vehicle (DMSO) alone. Mitochondrial pathways/genes examined included oxidative phosphorylation, fatty acid oxidation, Krebs cycle, ATP synthase, and uncoupling proteins. Additionally, expression of transcriptional regulators functionally related to ERRγ were examined. For almost all of the genes examined, we found a dose- dependent increase in gene expression following treatment with Compound 1. All gene expression results are from 24-hour Compound 1 treatment.
Genes of the oxidative phosphorylation demonstrated an elevated expression with increasing concentrations of Compound 1. COX-4 expression was increased by 2-fold, cytochrome c by 3.7-fold, and UQCRB by 2-fold when treated with Compound 1 at a concentration of 30 μM (Figure 14). 50201A
Similarly, three genes examined that are associated with fatty acid oxidation, were shown to be upregulated in a dose-dependent manner following 24-hour treatment with Compound 1. CPT-Ib expression was increased by over 3-fold, LCAD by 2.8-fold, and MCAD by 1.6-fold following 30 μM treatment of AJQ710 (Figure 15).
Two other mitochondrial genes examined, IDH3α, a component of the Krebs cycle, and ATP-5b, an ATP-synthesizing enzyme, demonstrated a dose-dependent induction in expression with increasing doses of Compound 1. IDH3α expression was increased by over 2-fold, and ATP-5b was increased by 1.6-fold after 24 hour treatment with 30 μM of Compound 1 (Figure 16).
We measured mRNA expression for the uncoupling proteins UCP-2 and UCP-3. UCP-2 showed marginal upregulation in expression with all doses of Compound 1, with its highest expression, 1.5-fold, following 10 μM treatment. UCP-3 showed a dose- dependent increase in expression, with an increase of over 3-fold following 30 μM treatment of Compound 1 (Figure 17).
We measured the expression of the ERR family in response to treatment of mytoubes with agonist. Compound 1 purportedly activates both ERRγ and ERRβ, but does not activate ERRα and the ERs (Zuercher, et al., J. Med. Chem., 48(9): 3107-9 (2005)). The expression of ERRα was increased to a greater extent than ERRβ or ERRγ following treatment with Compound 1. ERRγ was elevated to 1.4-fold expression, ERRβ showed a 1.5-fold increase in expression, and ERRα showed a 3.7-fold increase in expression with 30 μM treatment of Compound 1 (Figure 18).
The co-activators PGC- lα and PGC- lβ were also examined for gene expression. The expression of these transcripts showed a similar dose-dependent increase in expression, with PGC-I α elevated to 1.9-fold expression and PGC-I β elevated to 1.8-fold (Figure 19).
We further examined gene expression for the PPARs, a family of nuclear receptors that play an important role in lipid metabolism. PP ARa and PPARδ showed no change in expression following Compound 1 treatment. PPARγ showed a dose- dependent increase in expression with a maximal 2.2-fold expression following 30 μM Compound 1 treatment (Figure 20).
To assess mitochondrial activity at the protein level, a cytochrome c ELISA was performed. Cytochrome c is a critical element of the electron transport chain, and quantities of the protein serve as a biomarker for mitochondrial number and oxidative phosphorylation activity. We observed a dose-dependent increase of cytochrome c with compound treatment, with an increase of 88% when myotubes were treated with 30 μM Compound 1 for 24 hours (Figure 21).
To measure mitochondrial activity, citrate synthase activity was determined. This enzyme is often used as an indicator of mitochondrial content or activity in human muscle (Kelley, et al., Diabetes, 51(10): 2944-50 (2002)). Citrate synthase catalyzes the initial step of the Krebs cycle, which supplies substrate for oxidative phosphorylation. Myotubes treated with 30 μM Compound 1 for 24 hours showed a 28% increase in citrate synthase activity, while 10 μM and 3 μM treatment showed an increase of 8% and 9% respectively (Figure 22).
We further investigated whether induction of mitochondrial gene expression has an impact on oxidative phosphorylation. Cellular respiration was measured in mouse primary myotubes treated with Compound 1 for 24 hr. Consistent with the induction of expression of the respiratory chain components, basal respiration was increased by 37% in Compound 1 -treated muscle cells, compared with the vehicle treated controls (Figure 23). Oligomycin-ϊnsensitive respiration, the proton leak, was increased by 36%, although this increase was not statistically significant. In the presence of FCCP, respiration rate was increased by 32% by Compound 1, as compared to the vehicle-treated control cells (Figure 23). These results indicate that an ERRγ agonist can increase mitochondria] oxidative capacity through coupled respiration.
OTHER EMBODIMENTS
Other embodiments will be evident to those of skill in the art. It should be understood that the foregoing detailed description is provided for clarity only and is
- 20 - 5020 IA
merely exemplary. The spirit and scope of the present invention are not limited to the above examples, but are encompassed by the following claims.

Claims

50201A
The Claims
1 A method of identifying an ERRγ-binding compound comprising:
a) contacting a compound with a sample comprising (i) a polypeptide comprising
ERRγ LBD sequence and (ii) a PGC- let polypeptide;
b) selecting the compound that binds to said polypeptide and increases said polypeptide's interaction with PGC-I α. polypeptide.
2 A method of identifying an ERRγ agonist comprising:
a) contacting a candidate agonist with a cell expressing a polypeptide comprising an
ERRγ LBD sequence and an ERRγ DBD sequence and a PGC-Ia9 wherein the cell comprises a reporter gene that is recognized by the ERRγ DBD sequence, and wherein the reporter gene encodes a detectable polypeptide;
b) measuring the level of the detectable polypeptide relative to that in the absence of the candidate agonist; and
c) selecting the candidate agonist that induces significant increase in the level of the detectable polypeptide.
3 A method of identifying an ERRγ agonist comprising:
a) contacting a candidate agonist with a cell expressing (i) a polypeptide comprising an ERRγ polypeptide sequence and a PGC- lα polypeptide, wherein the cell comprises a reporter gene that is recognized by the ERRγ DBD sequence, and wherein the reporter gene encodes a detectable polypeptide;
b) measuring the level of the detectable polypeptide relative to that in the absence of the candidate agonist; and
c) selecting the candidate agonist that induces significant increase in the level of the detectable polypeptide. 50201A
4 The method of claim 2, wherein the agonist is a chemical compound, or a polypeptide.
5 The method of claim 3, wherein the agonist is a chemical compound, or a polypeptide.
6 The method of claim 2, wherein the detectable polypeptide comprises luciferase, β- galactosidase, or secreted alkaline phosphatase.
7 The method of claim 3, wherein the detectable polypeptide comprises luciferase β- galactosidase, or secreted alkaline phosphatase.
8 A method of identifying an ERRγ agonist comprising:
a) contacting a candidate agonist with a cell expressing a polypeptide comprising an
ERRγ LBD sequence and an ERRγ DBD sequence and a PGC- lα polypeptide;
b) measuring the expression level of a mitochondrial marker gene in the cell relative to that in the absence of the candidate agonist; and
c) selecting the candidate agonist that induces significant increase in the expression level of the mitochondrial marker gene.
9 The method of claim S, wherein the mitochondrial marker gene is one selected from the group consisting of PGC-I α, PGC- lβ, cytochrome c, citrate synthase, isocitrate dehydrogenase, NADH oxidase, UCP-3, ERRα, ERRγ, SOD2, cytochrome oxidase IV, ATP synthase, carnitine palmitoyl transferase -Ib.
10 The method of claim 8, wherein the agonist is a chemical compound, or a polypeptide.
11 A method of identifying an ERRγ agonist comprising:
a) contacting a candidate agonist with a cell expressing a polypeptide comprising an ERRγ polypeptide sequence and a PGC-I α polypeptide; 50201A
b) measuring the expression level of a mitochondrial marker gene in the cell relative to that in the absence of the candidate agonist; and
c) selecting the candidate agonist that induces significant increase in the expression level of the mitochondrial marker gene.
12 The method of claim 11, wherein the mitochondrial marker gene is one selected from the group consisting of PGC-I α, PGC- lβ, cytochrome c, citrate synthase, isocitrate dehydrogenase, NADH oxidase, UCP-3, ERRa5 ERRγ, SOD2, cytochrome oxidase IV, ATP synthase, carnitine palmitoyl transferase -Ib
13 The method of claim 11, wherein the agonist is a chemical compound, or a polypeptide.
PCT/US2007/010592 2006-05-03 2007-05-02 FUNCTION OF ESTROGEN RELATED RECEPTOR GAMMA (ERRγ) IN INCREASING MITOCHONDRIAL BIOGENESIS WO2007130431A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US79722906P 2006-05-03 2006-05-03
US60/797,229 2006-05-03

Publications (2)

Publication Number Publication Date
WO2007130431A2 true WO2007130431A2 (en) 2007-11-15
WO2007130431A3 WO2007130431A3 (en) 2008-03-20

Family

ID=38617476

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/010592 WO2007130431A2 (en) 2006-05-03 2007-05-02 FUNCTION OF ESTROGEN RELATED RECEPTOR GAMMA (ERRγ) IN INCREASING MITOCHONDRIAL BIOGENESIS

Country Status (1)

Country Link
WO (1) WO2007130431A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2338879A1 (en) * 2009-12-15 2011-06-29 Arteria New phenylhydrazone derivatives and their use as pharmaceuticals
US8962546B2 (en) 2011-03-01 2015-02-24 Salk Institute For Biological Studies Modulation of estrogen receptor-related receptor gamma (ERRγ) and uses therefor
CN112662705A (en) * 2020-12-21 2021-04-16 山东省成体细胞产业技术研究院有限公司 Method for improving insulin secretion of islet cells through gene modification

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003064468A2 (en) * 2002-01-31 2003-08-07 Centre National De La Recherche Scientifique (Cnrs) The ligand binding pocket peptide fragment of estrogen-related receptor 3 (err3), its crystal structure, and uses thereof
EP1548434A1 (en) * 2002-08-08 2005-06-29 Japan Science and Technology Agency Method of screening drug
US20060270836A1 (en) * 2005-05-27 2006-11-30 Abad Marta C EERgamma peptide fragment complexes and uses thereof in drug discovery

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003064468A2 (en) * 2002-01-31 2003-08-07 Centre National De La Recherche Scientifique (Cnrs) The ligand binding pocket peptide fragment of estrogen-related receptor 3 (err3), its crystal structure, and uses thereof
EP1548434A1 (en) * 2002-08-08 2005-06-29 Japan Science and Technology Agency Method of screening drug
US20060270836A1 (en) * 2005-05-27 2006-11-30 Abad Marta C EERgamma peptide fragment complexes and uses thereof in drug discovery

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
COWARD P ET AL: "4-Hydroxytamoxifen binds to and deactivates the estrogen-related receptor gamma" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC, US, vol. 98, no. 15, 17 July 2001 (2001-07-17), pages 8880-8884, XP002971442 ISSN: 0027-8424 *
GOWDA ET AL: "Development of a coactivator displacement assay for the orphan receptor estrogen-related receptor-gamma using time-resolved fluorescence resonance energy transfer" ANALYTICAL BIOCHEMISTRY, ACADEMIC PRESS, SAN DIEGO, CA, US, vol. 357, no. 1, 1 October 2006 (2006-10-01), pages 105-115, XP005632068 ISSN: 0003-2697 *
LIU JIANWEI ET AL: "A homogeneous in vitro functional assay for estrogen receptors: coactivator recruitment." MOLECULAR ENDOCRINOLOGY (BALTIMORE, MD.) MAR 2003, vol. 17, no. 3, March 2003 (2003-03), pages 346-355, XP002466112 ISSN: 0888-8809 *
PARK YUN-YONG ET AL: "An autoregulatory loop controlling orphan nuclear receptor DAX-1 gene expression by orphan nuclear receptor ERRgamma." NUCLEIC ACIDS RESEARCH 2005, vol. 33, no. 21, 2005, pages 6756-6768, XP002466113 ISSN: 1362-4962 *
ZUERCHER WILLIAM J ET AL: "Identification and structure-activity relationship of phenolic acyl hydrazones as selective agonists for the estrogen-related orphan nuclear receptors ERRbeta and ERRgamma." JOURNAL OF MEDICINAL CHEMISTRY 5 MAY 2005, vol. 48, no. 9, 5 May 2005 (2005-05-05), pages 3107-3109, XP002466114 ISSN: 0022-2623 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2338879A1 (en) * 2009-12-15 2011-06-29 Arteria New phenylhydrazone derivatives and their use as pharmaceuticals
US8962546B2 (en) 2011-03-01 2015-02-24 Salk Institute For Biological Studies Modulation of estrogen receptor-related receptor gamma (ERRγ) and uses therefor
CN112662705A (en) * 2020-12-21 2021-04-16 山东省成体细胞产业技术研究院有限公司 Method for improving insulin secretion of islet cells through gene modification

Also Published As

Publication number Publication date
WO2007130431A3 (en) 2008-03-20

Similar Documents

Publication Publication Date Title
Hu et al. Targeting mitochondrial dynamics by regulating Mfn2 for therapeutic intervention in diabetic cardiomyopathy
Morselli et al. Hypothalamic PGC-1α protects against high-fat diet exposure by regulating ERα
Maynard et al. Human HIF‐3Α4 is a Dominant‐Negative regulator of HIF‐1 and is Down‐Regulated in renal cell carcinoma
Zeng et al. ATF6 modulates SREBP2‐mediated lipogenesis
Ramadoss et al. STAT3 targets the regulatory regions of gluconeogenic genes in vivo
Li et al. Genome-wide coactivation analysis of PGC-1α identifies BAF60a as a regulator of hepatic lipid metabolism
Citron et al. Protein crosslinking, tissue transglutaminase, alternative splicing and neurodegeneration
Chen et al. Phosphorylation of the liver X receptors
Canettieri et al. Dual role of the coactivator TORC2 in modulating hepatic glucose output and insulin signaling
Chu et al. Mild hypoxia‐induced cardiomyocyte hypertrophy via up‐regulation of HIF‐1α‐mediated TRPC signalling
Mikkonen et al. Sumo-1 regulates body weight and adipogenesis via PPARγ in male and female mice
Lee et al. SMILE upregulated by metformin inhibits the function of androgen receptor in prostate cancer cells
Zhou et al. ERRβ: a potent inhibitor of Nrf2 transcriptional activity
Yang et al. High-glucose induces cardiac myocytes apoptosis through Foxo1/GRK2 signaling pathway
Fujimura et al. FK614, a novel peroxisome proliferator-activated receptor γ modulator, induces differential transactivation through a unique ligand-specific interaction with transcriptional coactivators
US20070293579A1 (en) Use of carp inhibitors for the treatment of heart diseases
WO2007130431A2 (en) FUNCTION OF ESTROGEN RELATED RECEPTOR GAMMA (ERRγ) IN INCREASING MITOCHONDRIAL BIOGENESIS
Guan et al. Bioactive peptide apelin rescues acute kidney injury by protecting the function of renal tubular mitochondria
Ray et al. Regulation of signal transducer and activator of transcription 3 enhanceosome formation by apurinic/apyrimidinic endonuclease 1 in hepatic acute phase response
Li et al. HIGD1A links SIRT1 activity to adipose browning by inhibiting the ROS/DNA damage pathway
Oort et al. γ-Synuclein Is an Adipocyte-Neuron Gene Coordinately Expressed with Leptin and Increased in Human Obesity, 3
US9144591B2 (en) Transcription modulator compositions
Li et al. In search of alternatively spliced alpha‐Klotho Kl1 protein in mouse brain
WO2006102653A2 (en) Methods and compositions for treating insulin resistance and obesity-induced diseases
EP3315967A1 (en) Tm4sf19 as marker for diagnosing obesity and method using same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07776591

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07776591

Country of ref document: EP

Kind code of ref document: A2