WO2018074929A2 - Drugs mimicking calorie restriction and tools and methods for identifying the same - Google Patents

Drugs mimicking calorie restriction and tools and methods for identifying the same Download PDF

Info

Publication number
WO2018074929A2
WO2018074929A2 PCT/NL2017/050689 NL2017050689W WO2018074929A2 WO 2018074929 A2 WO2018074929 A2 WO 2018074929A2 NL 2017050689 W NL2017050689 W NL 2017050689W WO 2018074929 A2 WO2018074929 A2 WO 2018074929A2
Authority
WO
WIPO (PCT)
Prior art keywords
firefly
initiation
translation
renilla
tri
Prior art date
Application number
PCT/NL2017/050689
Other languages
French (fr)
Other versions
WO2018074929A3 (en
Inventor
Cornelis Ferdinand CALKHOVEN
Original Assignee
Rijksuniversiteit Groningen
Academisch Ziekenhuis Groningen
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 Rijksuniversiteit Groningen, Academisch Ziekenhuis Groningen filed Critical Rijksuniversiteit Groningen
Publication of WO2018074929A2 publication Critical patent/WO2018074929A2/en
Publication of WO2018074929A3 publication Critical patent/WO2018074929A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6897Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids involving reporter genes operably linked to promoters
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics

Definitions

  • the invention generally relates to the fields of medicine and drug development. More particularly, it relates to drugs that are capable of mimicking calorie restriction, and to screening tools and screening methods for identifying the same. Whereas mammals have widely differing lifespans, a common feature of lifespans is that they seems to be adjustable and can be increased by genetic or pharmacological interventions. Dietary calorie restriction (CR) (also called dietary restriction) is the best-documented intervention that delays most age-related physiological changes, and extends life span in all species tested, provided malnutrition is avoided (Weindruch, et al. The Retardation of Aging and Disease by Dietary Restriction (Charles C.
  • CR Dietary calorie restriction
  • mice with mutations in these genes have greater mean lifespans relative to the expected lifespan of control mice.
  • These genes include the Ames dwarf mutation, and the Snell dwarf mutation.
  • these mutations result in small, frail mice which have difficulty feeding. Therefore, it is beheved that the longevity conferred by these mutations is essentially due to calorie restriction.
  • Recent attempts to use gene array analysis, or other genetic screens for genes associated with longevity phenotypes in worms, flies, and rodents have come up with a number of candidate genes. In general, however, they are frequently "stress-response" genes.
  • Sirtuins are highly conserved protein deacetylases and/or ADP- ribosyltransferases that have been shown to extend lifespan in lower model organisms, such as yeast, C. elegans, and drosophila. In mammals, sirtuins have been shown to act as metabolic sensors, responding to environmental signals to coordinate the activity of genes that regulate multiple energy homeostasis pathways. For example, studies have shown that sirtuin activation mimics the effects of caloric restriction, an intervention
  • mTORC l nutrient sensitive mechanistic target of rapamycin complex 1
  • LAP and LAP* Liver-specific Activating Protein
  • LAP* Liver-specific Activating Protein
  • uORF cis- regulatory upstream open reading frame
  • Activated mTORC 1 signahng stimulates the latter re-initiation into LIP but only marginally affects the initiation into LAP.
  • the dependence of LIP expression on the presence of the uORF can be explained by the finding that translation of the uORF prevents translation initiation at the LAP initiation codon. Mechanistically, this is based on the fact that uORF -post-termination ribosomes have to be reloaded with new initiator tRNA (Met-tRNAi Met) in order to perform a second translation re-initiation at the same mRNA molecule.
  • the LAP initiation codon is too close (4 nt) to the uORF for the post-termination ribosomes to be reloaded with new Met-tRNAi M i in time. Therefore, they omit the LAP initiation codon but can be reloaded with Met- tRNAi Met early enough to re-initiate translation at the downstream LIP initiation codon.
  • the sophisticated structure of the C/EBP -mRNA renders its translation responsive to changes in availability or activity of
  • mTORCl stimulates the expression of the metabolic transcription factor CCAAT/Enhancer Binding Protein ⁇
  • C/ ⁇ Liver-specific Inhibitory Protein
  • the present inventors set out to search for novel possibilities to pharmacologically down-regulate LIP in order to induce CR-mimetic effects. To that end, they developed and performed a screening strategy for the identification of compounds that alter C/EBP -uORF dependent translation re-initiation into LIP.
  • the screening strategy involved a novel reporter assay system (see herein below), which was validated with pharmacological and genetic approaches.
  • the screening of a library of FDA approved drugs surprisingly revealed Adefovir Dipivoxil as a single drug that reduces translation re-initiation in different cell lines, reduces endogenous LIP expression and increases ⁇ -oxidation in a mouse hepatoma cell line.
  • the invention provides adefovir, or a prodrug thereof, for use in a method for improving the metabolic health in a subject.
  • the term improving metabolic health' refers to all aspects relating to CR, including increase ⁇ -oxidation, improved insulin sensitivity and glucose tolerance, reduced body weight, reduced fat starage, prevention of steatosis (fatty liver), reduced serum lipids. Also comprised is body weight-related cancer incidence.
  • improving the metabolic health comprises shifting the metabolism towards enhanced beta-oxidation. In another embodiment, improving the metabolic health comprises reducing weight gain or enhancing or supporting weight loss in a subject.
  • the subject is a human subject.
  • the invention is also suitably used to improve the metabolic health in a non-human mammalian subject, like a pet or a husbandry animal.
  • adefovir or an adefovir prodrug for use as a calorie restriction mimetic compound.
  • the adefovir prodrug is adefovir dipivoxil.
  • Adefovir dipivoxil was approved in 2002 for the treatment of chronic hepatitis B.
  • HEPSERATM is the tradename for adefovir dipivoxil, a diester prodrug of adefovir.
  • Adefovir is an acyclic nucleotide analogue of adenosine monophosphate that inhibits the hepatitis B virus (HBV) DNA polymerase by competing with the natural substrate deoxyadenosine triphosphate and by causing DNA chain termination after its incorporation into viral DNA.
  • HBV hepatitis B virus
  • adefovir dipivoxil The chemical name of adefovir dipivoxil is 9-[2- [bis[(pivaloyloxy)methoxy] phosphinyl]methoxy] -ethyl] adenine.
  • Adefovir is phosphorylated to the active metabolite, adefovir diphosphate, by cellular kinases.
  • a further embodiment relates to a weight management composition
  • a weight management composition comprising adefovir or an adefovir prodrug, e.g. adefovir dipivoxil, in combination with one or more further active ingredient(s) supporting or enhancing weight loss which is known in the art or which is yet to be discovered.
  • the composition comprises, in addition to adefovir or an adefovir prodrug, a weight loss effective amount of a noradrenaline stimulating compound such as ephedrine, mahuang (a plant source of ephedrine alkaloids), citrus aurantium (bitter orange), synephrine, norephedrine, psuedophedrine, a methylxanthine, such as caffeine or guarana, and/or a COX-2 inhibitor such as resveratrol, polygonum
  • a noradrenaline stimulating compound such as ephedrine, mahuang (a plant source of ephedrine alkaloids), citrus aurantium (bitter orange), synephrine, norephedrine, psuedophedrine, a methylxanthine, such as caffeine or guarana
  • COX-2 inhibitor such as resveratrol, polygonum
  • the composition comprises a biologically effective amount of an extract of a plant of the genus Eucalyptus as an active ingredient, which extract was found to be useful in a method of inhibiting or preventing obesity (increase in weight) lipid storage disease,
  • adefovir or an adefovir prodrug e.g. adefovir dipivoxil
  • adefovir or an adefovir prodrug is combined with (a) at least a kind of polyphenols selected from the group consisting of hesperidin, a hesperidin derivative, and hesperetin; and (b) at least one xanthine derivative, the mass ratio of (a):(b) in the composition being 1:(0.001 to 5). See US2015/0265645A1.
  • the invention relates to screening tools and screening methods for identifying the further compounds that, like adefovir, are capable of mimicking calorie restriction.
  • the present inventors recognized that the specific translation mechanism that is responsible for LIP translation is potentially draggable, as reduction of LIP through pharmacological inhibition of mTORC 1 exemplifies.
  • the invention provides a dual reporter plasmid system comprising a first nucleic acid sequence encoding a Renilla luciferase transcript and a second nucleic acid sequence encoding a firefly luciferase transcript, wherein said first and second nucleic acid sequence are both under control of the C/EBP -5'UTR- uORF (upstream Open Reading Frame).
  • C/EBP -5'UTR- uORF upstream Open Reading Frame
  • the dual reporter plasmid system of the invention comprises 1) a translation initiation cassette with a nucleic acid sequence encoding a renilla luciferase transcript as surrogate for C/EBPp-LAP expression and 2) a translation re-initiation cassette with a nucleic acid sequence encoding a firefly luciferase transcript as surrogate for ⁇ / ⁇ - ⁇ expression, wherein said first and second nucleic acid sequence are both under control of the C/EBP -5'UTR-uORF. See Figure lA for a schematic drawing.
  • the invention provides a cellular reporter system as a surrogate for measuring the ratio between translation initiation and uORF-dependent re-initiation as it is used as a mechanism for differential translation of the C/EBP -mRNA into the protein isoforms LAP and LIP.
  • the system we present here belongs to the type of phenotypic drug discovery (forward pharmacology) where compounds are identified that cause a desirable change in phenotype, here reduction of C/EBP -uORF- dependent translation re-initiation.
  • the system is not purely phenotypic and target-agnostic (without having prior knowledge of their biological activity or mode of action against a specific molecular target or targets) and is better described as a translation re-initiation and initiation mechanism-informed phenotypic screen (Moffat et al, 2014; Swinney & Anthony, 2011).
  • Phenotypic screens potentially can identify compounds that modify a (disease) phenotype by acting on a yet undiscovered target or targets.
  • a cell comprising a dual reporter plasmid system according to the invention.
  • the plasmid is stably integrated into the genome of the cell.
  • Suitable cells include mammahan cells or cell lines, like HEK293T, NIH-3T3, primary hepatocytes or iPS-derived cell types.
  • a cell comprising the dual reporter system of the invention finds its use in drug-development.
  • a method or an assay for identifying a compound capable of mimicking calorie restriction (CR) and/or having anti-cancer activity comprising the steps of providing a cell culture comprising cells according to the invention, contacting said cell culture with a candidate compound (to allow for luciferase expression), determining expression oiRenilla luciferase and firefly luciferase, calculating the translation re-initiation index (TRI) based on the formula
  • TRI Firefly Re hliiiati011 x ReniHa Reference / Renilla lni1i tion x Firefly Reference and correlating said TRI with the capacity of said compound to mimic calorie restriction, wherein a reduction in TRI is a positive indicator of the compound being capable of mimicking calorie restriction (CR) and or having anti-cancer activity.
  • determining expression of Renilla luciferase and firefly luciferase comprises the quantitation of a stable luminescent signal from two reporter genes in a single sample.
  • kits which provide a convenient "add-and-read" system that generates both firefly and Renilla luciferase luminescence signals from cells that have not been preconditioned or prelysed
  • An additional reporter plasmid may be used for measurement of reference levels of firefly (FireflyRef) and renilla (RenillaRef) luciferase through unrestrained translation (Fig. IB, pcDNA FireflyRef/RenillaRef).
  • This reporter can serve for the counter assay to control for effects not related to reinitiation/initiation efficiency, like the rate of transcription, mRNA stabilization, general effect on translation and/or direct interference with the luciferase activity.
  • the assay or method further comprises cell comprising a second reporter reference plasmid system, comprising a first nucleic acid sequence encoding a renilla luciferase transcript and a second nucleic acid sequence encoding a firefly luciferase transcript without C/EBP -5'UTR-uORF sequences (see Figure 1 B).
  • kits-of-parts comprising a dual reporter plasmid system according to the invention, a luciferase substrate, and optionally a reference reporter plasmid as mentioned herein above.
  • a method and/or kit of the invention is advantageously used in a high throughput drug screening assay, in particular for the identification of compounds capable of improving the metabolic health in a subject.
  • Renilla luciferase (Renilla 1111 ) can only be translated by ribosomes that have scanned over the uORF and directly initiate at the AUG initiation codon of the renilla reading frame. Translation of the firefly luciferase (Firefly Re ' li ) is only possible by ribosomes that have first translated the uORF, resume scanning and re-initiate at the AUG initiation codon of the firefly reading frame. The latter transcript also contains a "dummy" reading frame that captures those ribosomes that have bypassed the uORF to directly initiate at the "dummy" AUG initiation codon. Here, no signal is produced.
  • TSC 1-KO mouse embryonic fibroblasts
  • the bar graphs show the calculated TRI mean values.
  • Immunoblots show the levels of C/ ⁇ , TSC l, phospho- S6K, S6K and the ⁇ -actin loading control.
  • the bar graphs show the calculated TRI mean values.
  • Immunoblots show the levels of C/ ⁇ , 4E-BP 1, 4E-BP2 and the ⁇ -actin loading control.
  • the bar graphs show the calculated TRI mean values.
  • the immunoblots show mTORC 1 inhibition by reduced phosphorylation of 4E-BP1 and S6K in addition to ⁇ -actin as loading control.
  • Immunoblots shows the reduced phosphorylation of eIF2a and the ⁇ -actin loading control.
  • A, B and C Scatter plots for three different plates with measured translation re-initiation/initiation ratio (Firefly Re ini /Renilla Ini ) of 780 FDA- approved drugs.
  • Each plate contained 6 wells treated with DMSO as a solvent and rapamycin as a positive control (bar graph at the right, error bars represent SD).
  • the threshold of three times the standard deviation from the DMSO mean value (3xSD) is indicated. Drugs that decreased the translation re-initiation/initiation ratio more than 3xSD are indicated in red. Drugs indicated by numbers (#1, 2, 3 and 4) were selected for further evaluation.
  • the arrows point to Sirolimus in plate 1 and Everolimus in plate 2.
  • TBI mean values of HEK293T cells transiently transfected with pcDNA3-FireflyRe-ini/RenillaIni and pcDNA3-FireflyRef/RenillaRef reporter plasmids and treated with drug #1, 2, 3 or 4.
  • OCR oxygen consumption rate
  • a fragment of the pcDNA3-LAP-Renilla between the Bglll restriction site and spanning the renilla sequences was amplified by PCR using the following primers, forward: 5'-CGG AAA TGT TGA ATA CTC ATA CTC-'3 and reverse: 5'-GCT CAG ATC TCC TCA GAA GCC ATA GAG C-'3.
  • the amplified fragment was sequenced and cloned in the pcDNA3-LIP- Firefly using the Bglll site to obtain the final pcDNA-Firefly Re ini /Renilla Ini .
  • the human DENR shRNA expression vector was generated by annealing the oligonucleotides shDENR /ora;ard:'5-CCG GCA AGA GTA TGT GGC CTT GCA ACT CGA GTT GCA AGG CCA CAT ACT CTT GTT TTT T-3' and shDENR reverse '5-AAT TAA AAA ACA AGA GTA TGT GGC CTT GCA ACT CGA GTT GCA AGG CCA CAT ACT CTT G-3' and ligating them into the Tet-pLKO-puro vector (Addgene plasmid #21915, described in
  • HEK293T and Hepal-6 cells were maintained in DMEM and MCF7 cells in RPMI (Gibco) supplemented with 10% fetal calf serum (FCS) and 1%
  • pcDNA-Firefly R( '" l i /Renilla lni or pcDNA-Firefly Rei /Renilla Rei were transfected into HEK293T cells using FUGENE (Roche).
  • Cell clones were selected by addition of G418 to the medium (1 mg/ml).
  • the cells were transduced with the inducible lentiviral Tet-pLKO-puro-shDENR followed by selection of the transduced cells with puromycin (1.5 ⁇ g/ml). Then these cells were treated with doxycycline (100 ng/ml)for 36 h.
  • Rapamycin (R-5000, LC Laboratories), PP242 (#P0073, Sigma) and eIF-2a- kinase PKR inhibitor (C16) (#19785, Sigma) were used as chemical inhibitors.
  • Luciferase assay 25000 cells per well were seeded in 96-well plates. After 24 h, cells were cotransfected with the plasmids as indicated using FUGENE HD (Promega), or stably transfectecl cells were used. The next day, luciferase activity was measured in cells treated with 8h of drug treatment or DMSO solvent control by Dual-Glo Luciferase Assay System (#2920, Promega) following the manufacturer's protocol using a GloMax- Multi Detection System (Promega).
  • HEK293T cells were seeded in 96-well plates. After 24h, cells were treated with the different drugs for 8h and cell viability was measured by Cell-Titer Flour cell viability assay system (#G6081, Promega) following the manufacturer's protocol using a GloMax-Multi Detection System (Promega).
  • the cells were washed twice with ice-cold lx PBS and lysed in 50 mM Tris pH 7.5, 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, supplemented with protease and phosphatase inhibitors followed by sonication. Equal amounts of total protein were separated by SDS-PAGE (#456-1094, BIO-RAD), transferred to a PVDF membrane using Trans-Blot Turbo System (#170-4273, BIO-RAD) following the manufacturer's protocol.
  • the following antibodies were used: C/ ⁇ (C19) and SBDS (S-15) from Santa Cruz Biotechnology; phospho-p70S6K (Thr389) (108D2), p70S6K (#9202), Hamartin/TSCl (D43E2) (#6935), phospho-4E-BPl (Thr37/46) (#9459), 4E-BP1 (#9452), 4E-BP2 (#2845), eIF4E (#9742), Phospho-eIF2a (Ser51) (#9721), eIF2a (#9722) and PKR (#3072) from Cell Signaling; DENR (#10656- 1-AP) from ProteintechTM and ⁇ -actin (clone C4) (#691001) from MP Biomedicals. HRP-conjugated secondary antibodies were purchased from Amersham Life Technologies. The bands were visuahzed by
  • HEK293T cells stably expressing Firefly R( '"TM Renilla Ini vector were seeded at a density of 1.5xl0 4 in 30 ⁇ media per well of a 384-well plate (Greiner CELLSTAR® 384 well plates, white) and grown overnight. Next day, the cells were treated for eight hours with 780 FDA-approved drugs library (#BML-2843-0100, Enzo Lifescience) (final concentration 10 ⁇ ), 0.5% v/v DMSO and rapamycin (final concentration 200 nM) using an Echo® 555 Liquid Handler (Labcyte) transferring up to 150 nl to each well. After lysis of the cells, luciferase activity was measured.
  • Oxygen consumption rate and fatty acid oxidation were determined using a Seahorse XF96 Extracellular Flux analyzer (Seahorse Bioscience).
  • 2 l0 Hepa 1-6 cells per well were seeded into a 96- well XF cell culture microplate 24 h prior to the assay and cultured in the presence of 0.5 mM carnitine. Sixteen hours before the assay, cells were treated with the drugs.
  • EXAMPLE 1 Reporter system to emulate (monitor) C/EBPJi-mRNA translation
  • TRI FirefLy R ⁇ ' ini Renilla Ref / Reiiilla lni FireflyTM
  • mTORCl inhibition was monitored by reduced phosphorylation of 4E- BP1 and S6 kinase (S6K). Finally, overexpression of eIF-4E that increases LIP expression accordingly increased TRI values (Fig 2D). Therefore, the reporter system reliably reflects differential translation of the C/ ⁇ - mRNA under changing mTORC 1 signaling.
  • the eIF2a-kinases suppress post-uORF translation re-initiation through inhibitory phosphorylation of eIF2a in response to various stresses (Aitken & Lorsch, 2012; Sonenberg & Hinnebusch, 2009), which results in reduced expression of LIP (Calkhoven et al, 2000).
  • eIF2a-kinase signaling can be measured using the TRI reporter system.
  • DENR density- regulated protein
  • HEK293T cells with a doxycycline-inducible DENR-shRNA expression vector.
  • Induction of DENR- KD (+Dox) resulted in lower TRI comparing to control cells (Fig 2F), confirming the crucial role of DENR in uORF-dependent translation reinitiation.
  • SBDS Shwachman- Bodian-Diamond SyndiOme
  • the TRI reporter system reliably detects changes in the ratio of initiation versus re-initiation by known translationally active drugs or alterations in key translation initiation factors and their regulatory kinases or non-canonical regulators of re-initiation.
  • EXAMPLE 3 Assay development and execution of a high- throughput screening (HTS)
  • HTS HTS we generated HEK293T cells with stably integrated pcDNA-Firefly R - ini /Renilla lni reporter plasmid. Single clones were tested for renilla and firefly expression and the highly expressing cell clones were in addition tested for stability of expression over repeated cell passages. Cell line clones that met these criteria were selected for further experiments. Similarly, a cell line with stably integrated pcDNA-Firefly Rei /Renilla Rei plasmid was generated. To determine an optimal assay window using reference
  • the ENZO library contains three mTORCl inhibitors that are derived from ra amycin (the rapalogs everolimus, sirolimus and temsirolimus) and two of them, sirolimus and everolimus, decreased re-initiation/initiation ratio (Fig 3A, B).
  • the temsirolimus signal was neglected because of aberrantly low luciferase signals (technical failure).
  • Timchenko LT Salisbury E, Wang GL, Nguyen H, Albrecht JH, Hershey JW, Timchenko NA (2006) J Biol Chem 281: 32806-32819 Wander SA, Hennessy BT, Slingerland JM (2011) J Clin Invest 121: 1231- 1241

Abstract

The invention relates to the fields of medicine and drug development, more particularly to drugs that are capable of mimicking calorie restriction (CR), and to screening tools and screening methods for identifying the same. Provided is a dual reporter plasmid system comprising a first nucleic acid sequence encoding a Renilla luciferase transcript and a second nucleic acid sequence encoding a firefly luciferase transcript, wherein said nucleic acid sequences are both under control of the C/EBPβ-5'UTR-uORF. Also provided is adefovir or a prodrug thereof for use in a method for improving the metabolic health in a subject.

Description

Title: Drugs mimicking calorie restriction and tools and methods for identifying the same.
The invention generally relates to the fields of medicine and drug development. More particularly, it relates to drugs that are capable of mimicking calorie restriction, and to screening tools and screening methods for identifying the same. Whereas mammals have widely differing lifespans, a common feature of lifespans is that they seems to be adjustable and can be increased by genetic or pharmacological interventions. Dietary calorie restriction (CR) (also called dietary restriction) is the best-documented intervention that delays most age-related physiological changes, and extends life span in all species tested, provided malnutrition is avoided (Weindruch, et al. The Retardation of Aging and Disease by Dietary Restriction (Charles C.
Thomas, Springfield, 111., 1988)). CR has been shown to potentially increase hfespan by 25-35% in all animals studied to date (mice, rats, several species of monkeys, dogs7 as well as non-metazoan species such as spiders,
Nematodes, and Drosophila). These studies also have shown that CR is a highly, if not the most, effective means now known for reducing cancer incidence and increasing the mean age of onset of age-related diseases and tumors in homeothermic vertebrates (Weindruch et al. (1982) Science 215: 1415). Thus, it seems clear that hfespans can be extended through a relatively simple dietary regimen.
Several genes in mice have been identified as "longevity" genes because mice with mutations in these genes have greater mean lifespans relative to the expected lifespan of control mice. These genes include the Ames dwarf mutation, and the Snell dwarf mutation. However, these mutations result in small, frail mice which have difficulty feeding. Therefore, it is beheved that the longevity conferred by these mutations is essentially due to calorie restriction. Recent attempts to use gene array analysis, or other genetic screens for genes associated with longevity phenotypes in worms, flies, and rodents have come up with a number of candidate genes. In general, however, they are frequently "stress-response" genes.
Sirtuins are highly conserved protein deacetylases and/or ADP- ribosyltransferases that have been shown to extend lifespan in lower model organisms, such as yeast, C. elegans, and drosophila. In mammals, sirtuins have been shown to act as metabolic sensors, responding to environmental signals to coordinate the activity of genes that regulate multiple energy homeostasis pathways. For example, studies have shown that sirtuin activation mimics the effects of caloric restriction, an intervention
demonstrated to significantly extend lifespan, and activates genes that improve glucose homeostasis and the conversion of fat to energy by fatty acid oxidation (Guarente, 2012, Nature Reviews Molecular Cell Biology 13, 207).
Some compounds, such as gingko, ginseng and vitamin C, have been proposed to improve survival, but controlled and statistically significant survival studies reporting the benefit for these compounds are unknown. Vitamin C and a number of drugs reduce the incidence of certain disease conditions, e.g. cardiovascular disease, and so, presumably, would enhance overall longevity. It is also known that an important part of the beneficial effects of CR on health- and life-span is mediated through regulation of protein synthesis that is under control of the nutrient sensitive mechanistic target of rapamycin complex 1 (mTORC l). mTORC l signaling coordinates the regulation of global protein synthesis and autophagy to adapt protein homeostasis to changing nutrient availability and growth factor signaling (Shimobayashi and Hall, 2014). In addition, the expression of a subset of proteins is specifically regulated by mTORC 1 through distinctive
translation of their mRNAs involving cis-regulatory elements that make them responsive to regulators of translation. Previously, we presented that specific translation into the C/ΕΒΡβ protein isoform LIP (Liver-specific Inhibitory Protein) is under control of mTORC 1 trough regulation of the downstream eukaryotic translation initiation factor 4E (eIF-4E) binding proteins (4E-BPs), and that mTORC 1 -inhibition by rapamycin reduces LIP expression (Calkhoven et al., 2000; Zidek et al., 2015).
Translation of the C/EBP -mRNA involves two separate translation mechanisms, initiation and re-initiation: 1) Synthesis of the isoforms LAP and LAP* (Liver-specific Activating Protein) is the result of regular translation initiation where ribosomes scan the mRNA from the 5'-end to the first AUG-codon in a favorable Kozak sequence context to initiate translation (LAP* is often weakly expressed because it has no Kozak sequence); 2) translation into LIP requires the initial translation of a cis- regulatory upstream open reading frame (uORF) in the mRNA leader- sequence, followed by the continuation of mRNA scanning and translation re-initiation from the downstream LIP-AUG coclon (Calkhoven et al., 2000). Activated mTORC 1 signahng stimulates the latter re-initiation into LIP but only marginally affects the initiation into LAP. The dependence of LIP expression on the presence of the uORF can be explained by the finding that translation of the uORF prevents translation initiation at the LAP initiation codon. Mechanistically, this is based on the fact that uORF -post-termination ribosomes have to be reloaded with new initiator tRNA (Met-tRNAi Met) in order to perform a second translation re-initiation at the same mRNA molecule. The LAP initiation codon is too close (4 nt) to the uORF for the post-termination ribosomes to be reloaded with new Met-tRNAiM i in time. Therefore, they omit the LAP initiation codon but can be reloaded with Met- tRNAiMet early enough to re-initiate translation at the downstream LIP initiation codon. The sophisticated structure of the C/EBP -mRNA renders its translation responsive to changes in availability or activity of
translation-regulatory factors.
Thus, as one of its activities, mTORCl stimulates the expression of the metabolic transcription factor CCAAT/Enhancer Binding Protein β
(C/ΕΒΡβ) isoform Liver-specific Inhibitory Protein (LIP). Regulation of LIP expression strictly depends on a translation re-initiation event that requires a conserved cis-regulatory upstream open reading frame (uORF) in the C EBP -mRNA. We have shown recently that experimental suppression of LIP in mice, reflecting reduced mTORCl -signaling at the C/ΕΒΡβ level, results in CR-type of metabolic improvements. (Zidek et al (2015) EMBO Rep. 16, 1022-1036). However, means or methods to reduce C/EBP6-LIP levels with the aim to mimic CR are at present not available.
Recognizing the therapeutic potential of compounds capable of modulating levels of LIP, the present inventors set out to search for novel possibilities to pharmacologically down-regulate LIP in order to induce CR-mimetic effects. To that end, they developed and performed a screening strategy for the identification of compounds that alter C/EBP -uORF dependent translation re-initiation into LIP. The screening strategy involved a novel reporter assay system (see herein below), which was validated with pharmacological and genetic approaches. The screening of a library of FDA approved drugs surprisingly revealed Adefovir Dipivoxil as a single drug that reduces translation re-initiation in different cell lines, reduces endogenous LIP expression and increases β-oxidation in a mouse hepatoma cell line. Herewith, the invention provides adefovir, or a prodrug thereof, for use in a method for improving the metabolic health in a subject. As used herein, the term improving metabolic health' refers to all aspects relating to CR, including increase β-oxidation, improved insulin sensitivity and glucose tolerance, reduced body weight, reduced fat starage, prevention of steatosis (fatty liver), reduced serum lipids. Also comprised is body weight-related cancer incidence.
In one embodiment, improving the metabolic health comprises shifting the metabolism towards enhanced beta-oxidation. In another embodiment, improving the metabolic health comprises reducing weight gain or enhancing or supporting weight loss in a subject.
Preferably , the subject is a human subject. Of particular interest are elderly subjects or subjects suffering from, or at risk of developing, a condition selected from the group consisting of metabolic syndrome, obesity (foiexample, visceral fat accumulation, subcutaneous fat accumulation), type 2 diabetes, pre-diabetes, hypertension, disorders of lipid metabolism (for example, fatty liver, hyperlipidemia, dyslipidemia), insulin resistance, endothelial dysfunction, pro-inflammatory state, and pro-coagulative state.
However, the invention is also suitably used to improve the metabolic health in a non-human mammalian subject, like a pet or a husbandry animal.
Also provided herein is adefovir or an adefovir prodrug for use as a calorie restriction mimetic compound.
In a preferred embodiment of the invention, the adefovir prodrug is adefovir dipivoxil. Adefovir dipivoxil was approved in 2002 for the treatment of chronic hepatitis B. HEPSERA™ is the tradename for adefovir dipivoxil, a diester prodrug of adefovir. Adefovir is an acyclic nucleotide analogue of adenosine monophosphate that inhibits the hepatitis B virus (HBV) DNA polymerase by competing with the natural substrate deoxyadenosine triphosphate and by causing DNA chain termination after its incorporation into viral DNA. The chemical name of adefovir dipivoxil is 9-[2- [bis[(pivaloyloxy)methoxy] phosphinyl]methoxy] -ethyl] adenine. Adefovir is phosphorylated to the active metabolite, adefovir diphosphate, by cellular kinases.
A further embodiment relates to a weight management composition comprising adefovir or an adefovir prodrug, e.g. adefovir dipivoxil, in combination with one or more further active ingredient(s) supporting or enhancing weight loss which is known in the art or which is yet to be discovered.
In one embodiment, the composition comprises, in addition to adefovir or an adefovir prodrug, a weight loss effective amount of a noradrenaline stimulating compound such as ephedrine, mahuang (a plant source of ephedrine alkaloids), citrus aurantium (bitter orange), synephrine, norephedrine, psuedophedrine, a methylxanthine, such as caffeine or guarana, and/or a COX-2 inhibitor such as resveratrol, polygonum
cuspidatum, Scutellaria baicalensis, white willow bark, turmeric, curcumin, rosemary, green tea, ocimum sanctum (holy basil), or ginger. See US
6,475,530.
In another embodiment, the composition comprises a biologically effective amount of an extract of a plant of the genus Eucalyptus as an active ingredient, which extract was found to be useful in a method of inhibiting or preventing obesity (increase in weight) lipid storage disease,
hyperlipidemia, arteriosclerosis and thrombosis, and a method of inhibiting or reducing an amount of triglyceride in blood and an amount of cholesterol in blood (see US2007/0054000A1). In a further embodiment of the invention, adefovir or an adefovir prodrug, e.g. adefovir dipivoxil, is combined with (a) at least a kind of polyphenols selected from the group consisting of hesperidin, a hesperidin derivative, and hesperetin; and (b) at least one xanthine derivative, the mass ratio of (a):(b) in the composition being 1:(0.001 to 5). See US2015/0265645A1.
In yet another aspect, the invention relates to screening tools and screening methods for identifying the further compounds that, like adefovir, are capable of mimicking calorie restriction.
Conditions that reduce LIP levels, like CR or other means of mTORC 1 inhibition, are currently being explored for its feasibihty to improve metabolic health and to treat cancer. Long term CR, however, is not manageable for most people and current treatment with mTORC 1 inhibitors are of limited success and come with serious side effects (Stallone et al., 2009; Wander et al., 2011; Willemsen et al., 2016; Zaytseva et al., 2012). Transcription factor function is considered to be largely undruggable and, in case of LIP, it would almost inevitably mean that LAP is targeted too, since LAP includes all sequences that make up LIP.
As described herein above, the present inventors recognized that the specific translation mechanism that is responsible for LIP translation is potentially draggable, as reduction of LIP through pharmacological inhibition of mTORC 1 exemplifies.
Therefore, they set out to develop a screening strategy for the identification of compounds that alter C/EBPp-uORF dependent translation re-initiation into LIP and thereby ultimately identify compounds that have CR-mimetic and or anti-cancer activities. Provided herein is a cellular system containing C/EBP -uORF controlled luciferase-reporters that measures simultaneously translation initiation and re-initiation and is suited for high-throughput screening strategies. More specifically, the invention provides a dual reporter plasmid system comprising a first nucleic acid sequence encoding a Renilla luciferase transcript and a second nucleic acid sequence encoding a firefly luciferase transcript, wherein said first and second nucleic acid sequence are both under control of the C/EBP -5'UTR- uORF (upstream Open Reading Frame). Whereas reporter systems based on C EBPa-uORF regulation have been used in the past (Wiesenthal et al, 2006a; Wiesenthal et al, 2006b), these systems use a different uORF.
Moreover, they are based on immunotags and require Western blot analyis or ELISA-type assays, which are not suitable for HTS applications.
The dual reporter plasmid system of the invention comprises 1) a translation initiation cassette with a nucleic acid sequence encoding a renilla luciferase transcript as surrogate for C/EBPp-LAP expression and 2) a translation re-initiation cassette with a nucleic acid sequence encoding a firefly luciferase transcript as surrogate for ϋ/ΕΒΡβ-ΚΙΡ expression, wherein said first and second nucleic acid sequence are both under control of the C/EBP -5'UTR-uORF. See Figure lA for a schematic drawing.
Herewith, the invention provides a cellular reporter system as a surrogate for measuring the ratio between translation initiation and uORF-dependent re-initiation as it is used as a mechanism for differential translation of the C/EBP -mRNA into the protein isoforms LAP and LIP.
The system we present here belongs to the type of phenotypic drug discovery (forward pharmacology) where compounds are identified that cause a desirable change in phenotype, here reduction of C/EBP -uORF- dependent translation re-initiation. The system, however, is not purely phenotypic and target-agnostic (without having prior knowledge of their biological activity or mode of action against a specific molecular target or targets) and is better described as a translation re-initiation and initiation mechanism-informed phenotypic screen (Moffat et al, 2014; Swinney & Anthony, 2011). Phenotypic screens potentially can identify compounds that modify a (disease) phenotype by acting on a yet undiscovered target or targets. The successive identification of the target(s) using for example techniques as chemical proteomics (Rix & Superti-Furga, 2009) could give novel insights into translation control mechanisms and involved upstream signaling pathways and might thereby also broaden our knowledge of potential players in metabolic diseases, aging and cancer.
Also provided is a cell comprising a dual reporter plasmid system according to the invention. Preferably, the plasmid is stably integrated into the genome of the cell. Suitable cells include mammahan cells or cell lines, like HEK293T, NIH-3T3, primary hepatocytes or iPS-derived cell types.
In one embodiment, a cell comprising the dual reporter system of the invention finds its use in drug-development. For example, provided herein is a method or an assay for identifying a compound capable of mimicking calorie restriction (CR) and/or having anti-cancer activity, comprising the steps of providing a cell culture comprising cells according to the invention, contacting said cell culture with a candidate compound (to allow for luciferase expression), determining expression oiRenilla luciferase and firefly luciferase, calculating the translation re-initiation index (TRI) based on the formula
TRI = Firefly Re hliiiati011 x ReniHaReference / Renillalni1i tion x FireflyReference and correlating said TRI with the capacity of said compound to mimic calorie restriction, wherein a reduction in TRI is a positive indicator of the compound being capable of mimicking calorie restriction (CR) and or having anti-cancer activity. In one embodiment, determining expression of Renilla luciferase and firefly luciferase comprises the quantitation of a stable luminescent signal from two reporter genes in a single sample. For example, commercial kits are available which provide a convenient "add-and-read" system that generates both firefly and Renilla luciferase luminescence signals from cells that have not been preconditioned or prelysed An additional reporter plasmid may be used for measurement of reference levels of firefly (FireflyRef) and renilla (RenillaRef) luciferase through unrestrained translation (Fig. IB, pcDNA FireflyRef/RenillaRef). This reporter can serve for the counter assay to control for effects not related to reinitiation/initiation efficiency, like the rate of transcription, mRNA stabilization, general effect on translation and/or direct interference with the luciferase activity. Accordingly, in one embodiment the assay or method further comprises cell comprising a second reporter reference plasmid system, comprising a first nucleic acid sequence encoding a renilla luciferase transcript and a second nucleic acid sequence encoding a firefly luciferase transcript without C/EBP -5'UTR-uORF sequences (see Figure 1 B).
Also provided herein is a kit-of-parts comprising a dual reporter plasmid system according to the invention, a luciferase substrate, and optionally a reference reporter plasmid as mentioned herein above.
A method and/or kit of the invention is advantageously used in a high throughput drug screening assay, in particular for the identification of compounds capable of improving the metabolic health in a subject.
LEGEND TO THE FIGURES
Figure 1. Translation Re-initiation Index (TRI) determination.
A) Representation of pcDNA3-FireflyRe illi/Renillalni plasmid containing the initiation and re-initiation cassette as indicated. Renilla luciferase (Renilla1111) can only be translated by ribosomes that have scanned over the uORF and directly initiate at the AUG initiation codon of the renilla reading frame. Translation of the firefly luciferase (FireflyRe' li) is only possible by ribosomes that have first translated the uORF, resume scanning and re-initiate at the AUG initiation codon of the firefly reading frame. The latter transcript also contains a "dummy" reading frame that captures those ribosomes that have bypassed the uORF to directly initiate at the "dummy" AUG initiation codon. Here, no signal is produced.
B) Representation of pcDN A3 -Firefly R('r/RenillaR r plasmid for measurements of reference levels of firefly luciferase and renilla luciferase . The formula used for calculation of the translation re-initiation index (TRI) is shown framed.
Figure 2. Reporter system validation.
A) Mouse embryonic fibroblasts (MEFs) that are deficient in the mTORC 1 -inhibitor protein TSC 1 (TSC 1-KO) or wt MEFs transiently transfected with pcDNAS-Firefly^-^^/Renilla111' and pcDNA- FireflyRei7RenillaRei reporter plasmids. The bar graphs show the calculated TRI mean values. Immunoblots show the levels of C/ΕΒΡβ, TSC l, phospho- S6K, S6K and the β-actin loading control.
B) 4E-BP 1/2 double knockout (DKO) MEFs and wt MEFs transiently transfected with reporter constructs as described in (a). The bar graphs show the calculated TRI mean values. Immunoblots show the levels of C/ΕΒΡβ, 4E-BP 1, 4E-BP2 and the β-actin loading control. C) HEK293T cells stably expressing pcDNA3-FireflyRf -inVRenillaIni or pcDNA-FireflyRef ReniIlaRef reporters treated with 200 nM rapamycin or 10 nM PP242 for 8 h. The bar graphs show the calculated TRI mean values. The immunoblots show mTORC 1 inhibition by reduced phosphorylation of 4E-BP1 and S6K in addition to β-actin as loading control.
D) HEK293T cells described in (c) transfected with elF expression vectors. The bar graphs show the calculated TRI mean values. Immunoblots show levels of eIF4, eIF2a, PKR and the a-actin loading control.
E) HEK293T cells described in (c) treated with the PKR inhibitor (C16) for 8 h. The bar graphs show the calculated TRI mean values.
Immunoblots shows the reduced phosphorylation of eIF2a and the β-actin loading control.
F) DENR-KD (+Dox) and DENR-Control (-Dox) HEK293T cells transiently transfected with reporter constructs as described in (a). The bar graphs show the calculated TRI mean values. Immunoblots show levels of DENR and the β-actin loading control.
G) C33A cells with stable SBDS knockdown (shSBDS) or control cells (shCTRL) transiently transfected with reporter constructs as described in (a). The bar graphs show the calculated TRI mean values. Immunoblots show levels of SBDS and the β-actin loading control. Statistical differences were analyzed by Student's t-tests. Error bars represent ±SD (n = 5), *P < 0.05, **P < 0. 01, ***P < 0. 001.
Figure 3. High throughput screening of a drug library.
A, B and C) Scatter plots for three different plates with measured translation re-initiation/initiation ratio (FireflyRe ini/RenillaIni) of 780 FDA- approved drugs. Each plate contained 6 wells treated with DMSO as a solvent and rapamycin as a positive control (bar graph at the right, error bars represent SD). The threshold of three times the standard deviation from the DMSO mean value (3xSD) is indicated. Drugs that decreased the translation re-initiation/initiation ratio more than 3xSD are indicated in red. Drugs indicated by numbers (#1, 2, 3 and 4) were selected for further evaluation. The arrows point to Sirolimus in plate 1 and Everolimus in plate 2.
D) Names and structural formulas of the drug #1-4 (retrieved from the ChemSpider database, www.clieinspider.coin). Statistical differences were analyzed by Student's t-tests. Error bars represent ±SD (n = 6), **P < 0.01, ***p < 0. 001. Figure 4. Validation of the drugs that decrease translation reinitiation/initiation ratio.
A) TBI mean values of HEK293T cells transiently transfected with pcDNA3-FireflyRe-ini/RenillaIni and pcDNA3-FireflyRef/RenillaRef reporter plasmids and treated with drug #1, 2, 3 or 4.
B and C)TRI mean values of Hepal-6 or MCF7 cells transiently transfected with the constructs described in (A) and treated with drugs #2, 3 or 4.
D) Treatment with drug #4 (adefovir dipivoxil; 10 μΜ for 8 h) decreases C/EBPp-LIP/LAP ratio in HEK293T cells as analyzed by immunoblotting. β-actin was used as a loading control. Bar chart at the right shows quantification of the relative changes in LIP/LAP -isoform ratio by adefovir dipivoxil and rapamycin compared to DMSO solvent.
E and F)Same experiment as described in (D) with Hepal-6 or MCF7 cells, respectively. C/ΕΒΡβ -LIP/LAP isoform ratios were quantified by chemiluminescence and digital imaging. Statistical differences were analyzed by Student's t-tests. Error bars represent ±SD (n = 3), *P < 0.05, **P < 0.01, ***P < 0. 001. Figure 5. Adefovir dipivoxil augments fatty acid oxidation.
Increased oxygen consumption rate (OCR) upon usage of palmitate as exogenous energy source (fatty acid oxidation) in Hep a 1-6 cells treated with adefovir dipivoxil (#4). Panel A: OCR without palmitate-BSA. Panel B: OCR with palmitate-BSA, the latter being abolished by ectopic expression of C/EBPp-LIP (panel B, right bar graph). Statistical differences were analyzed by Student's t-tests. Error bars represent ±SD (n = 5), **P < 0.01.
EXPERIMENTAL SECTION Materials and Methods
DNA constructs
For generation of pcDNA-FireflyR('"kli/ReniHaIni, we first cloned the rat C EBPp-5'UTR until the LAP initiation codon in frame with the renilla sequence (from pGL4) in pcDNA3, and separately cloned C/EBPp-sequences spanning the 5'UTR and sequences until the LIP initiation codon with a +1 frame shift (7 nt upstream of the AUG) with the firefly sequences (from pGL3) in pcDNA3. A fragment of the pcDNA3-LAP-Renilla between the Bglll restriction site and spanning the renilla sequences was amplified by PCR using the following primers, forward: 5'-CGG AAA TGT TGA ATA CTC ATA CTC-'3 and reverse: 5'-GCT CAG ATC TCC TCA GAA GCC ATA GAG C-'3. The amplified fragment was sequenced and cloned in the pcDNA3-LIP- Firefly using the Bglll site to obtain the final pcDNA-FireflyRe ini/RenillaIni. For the reference construct pcDNA-FireflyKef/ReniHaRef, a similar cloning strategy was employed using solely renilla and firefly coding regions (from pGL4 and pGL3 respectively). eIF4E, eIF2a-S51D, eIF2a-S5lA and PKRA6 pcDNA3-based expression vectors are described in (Calkhoven et al, 2000). The human DENR shRNA expression vector was generated by annealing the oligonucleotides shDENR /ora;ard:'5-CCG GCA AGA GTA TGT GGC CTT GCA ACT CGA GTT GCA AGG CCA CAT ACT CTT GTT TTT T-3' and shDENR reverse '5-AAT TAA AAA ACA AGA GTA TGT GGC CTT GCA ACT CGA GTT GCA AGG CCA CAT ACT CTT G-3' and ligating them into the Tet-pLKO-puro vector (Addgene plasmid #21915, described in
(Wiederschain et al, 2009)).
Cell culture
HEK293T and Hepal-6 cells were maintained in DMEM and MCF7 cells in RPMI (Gibco) supplemented with 10% fetal calf serum (FCS) and 1%
Penicillin/Streptomycin at 37°C with 5% C02. TSCl-KO and 4E-BP1/2-DKO MEFs were previously described(Le Bacquer et al, 2007; Zhang et al, 2003). SBDS-KD C33A cells were used before (In et al, 2016). Hepal-6 cells ectopically expressing C/EBP -LIP were described at(Zidek et al, 2015). Cell number and viability was determined using the TC20™ automated cell counter (Bio-Rad) following the manufacturers instruction. For generation of cells stably expressing the reporter constructs, pcDNA-FireflyR('" li/Renillalni or pcDNA-FireflyRei/RenillaRei were transfected into HEK293T cells using FUGENE (Roche). Cell clones were selected by addition of G418 to the medium (1 mg/ml). For knockdown of DENR in HEK293T cells, the cells were transduced with the inducible lentiviral Tet-pLKO-puro-shDENR followed by selection of the transduced cells with puromycin (1.5 μg/ml). Then these cells were treated with doxycycline (100 ng/ml)for 36 h.
Rapamycin (R-5000, LC Laboratories), PP242 (#P0073, Sigma) and eIF-2a- kinase PKR inhibitor (C16) (#19785, Sigma) were used as chemical inhibitors.
Luciferase assay
For the Luciferase assay, 25000 cells per well were seeded in 96-well plates. After 24 h, cells were cotransfected with the plasmids as indicated using FUGENE HD (Promega), or stably transfectecl cells were used. The next day, luciferase activity was measured in cells treated with 8h of drug treatment or DMSO solvent control by Dual-Glo Luciferase Assay System (#2920, Promega) following the manufacturer's protocol using a GloMax- Multi Detection System (Promega).
Viability Assay
For the viability assay, 40000 HEK293T cells were seeded in 96-well plates. After 24h, cells were treated with the different drugs for 8h and cell viability was measured by Cell-Titer Flour cell viability assay system (#G6081, Promega) following the manufacturer's protocol using a GloMax-Multi Detection System (Promega).
Immimoblotting
For protein extraction, the cells were washed twice with ice-cold lx PBS and lysed in 50 mM Tris pH 7.5, 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, supplemented with protease and phosphatase inhibitors followed by sonication. Equal amounts of total protein were separated by SDS-PAGE (#456-1094, BIO-RAD), transferred to a PVDF membrane using Trans-Blot Turbo System (#170-4273, BIO-RAD) following the manufacturer's protocol. The following antibodies were used: C/ΕΒΡβ (C19) and SBDS (S-15) from Santa Cruz Biotechnology; phospho-p70S6K (Thr389) (108D2), p70S6K (#9202), Hamartin/TSCl (D43E2) (#6935), phospho-4E-BPl (Thr37/46) (#9459), 4E-BP1 (#9452), 4E-BP2 (#2845), eIF4E (#9742), Phospho-eIF2a (Ser51) (#9721), eIF2a (#9722) and PKR (#3072) from Cell Signaling; DENR (#10656- 1-AP) from Proteintech™ and β-actin (clone C4) (#691001) from MP Biomedicals. HRP-conjugated secondary antibodies were purchased from Amersham Life Technologies. The bands were visuahzed by
chemiluminescence (ECL, Amersham Life Technologies) using ImageQuant LAS 4000 mini imaging machine (GE Healthcare Bioscience AB) and the supplied software was used for the quantification of the bands.
Library screening
HEK293T cells stably expressing FireflyR('"™ RenillaIni vector were seeded at a density of 1.5xl04 in 30 μΐ media per well of a 384-well plate (Greiner CELLSTAR® 384 well plates, white) and grown overnight. Next day, the cells were treated for eight hours with 780 FDA-approved drugs library (#BML-2843-0100, Enzo Lifescience) (final concentration 10 μΜ), 0.5% v/v DMSO and rapamycin (final concentration 200 nM) using an Echo® 555 Liquid Handler (Labcyte) transferring up to 150 nl to each well. After lysis of the cells, luciferase activity was measured.
Measurements of cellular oxygen consumption rate and fatty acid oxidation
Oxygen consumption rate and fatty acid oxidation were determined using a Seahorse XF96 Extracellular Flux analyzer (Seahorse Bioscience). For fatty acid oxidation assay, 2 l0 Hepa 1-6 cells per well were seeded into a 96- well XF cell culture microplate 24 h prior to the assay and cultured in the presence of 0.5 mM carnitine. Sixteen hours before the assay, cells were treated with the drugs. One hour before the assay, the cells were washed twice with FAO assay buffer (Seahorse Bioscience), and 15 min before the assay, the fatty acid oxidation substrate palmitate-BSA or a BSA control (Seahorse Bioscience) was added and the oxygen consumption rate (OCR) with or without palmitate-BSA was measured.
EXAMPLE 1: Reporter system to emulate (monitor) C/EBPJi-mRNA translation
In order to identify small molecule compounds that modulate
C/EBP -uORF dependent and differential translation of the C/EBPp-mRNA into the protein isoforms LAP and LIP we designed a reporter system as a surrogate for this process. A single dual reporter plasmid was constructed that expresses a renilla luciferase transcript for the simulation of
translation initiation into LAP and a firefly luciferase transcript for the simulation of translation re-initiation into LIP, both under control of the C/EBPB-5'UTR (Fig 1A, pcDNA-FireflyRi -""/Renilla1"1). Comparable to translation into LAP from the genuine C/EBPp-niRNA, regular translation initiation after leaky scanning over the uORF results in expression of renilla luciferase from the initiation cassette (Renilla'11') (Fig 1A, Initiation
Cassette). This cassette does not contain the downstream LIP-AUG and therefore uORF translation and eventual subsequent downstream reinitiation events are not detected. The efficiency of uORF-dependent reinitiation, simulating translation into LIP, is measured as expression levels of firefly luciferase from the re-initiation cassette (Firefly R illi) (Fig 1A, Re- initiation Cassette). Ribosomes omitting translation of the uORF in the reinitiation cassette initiate translation at the proximal ORF consisting of C ΕΒΡβ sequences that would mimic translation at the LAP initiation site (Fig 1A, grey box), however, its product is not detected with the luciferase assay. In this way, only re-initiation is measured and direct initiation is left unnoticed. Alterations in the ratio between re-initiation and initiation are depicted as FireflyRe im/ReniUaIm values for the compound library screens.
An additional reporter plasmid was constructed for measurement of reference levels of firefly (FireflyRei) and renilla (RenillaRf l luciferase through unrestrained translation (Fig IB, pcDNA-FireflyRfif/RenillaRef)- This reporter is used for the counter assay to control for effects not related to reinitiation/initiation efficiency like the rate of transcription, mRNA
stabilization, general effect on translation or direct interference with the luciferase activity. Using measurements obtained with the pcDNA3- FireflyRil ini/RenillaIni and the pcDN A3 -Firefly ^VReniHa^ plasmids a translation re-initiation index (TRI) can be calculated by using the formula TRI = FirefLyR<' ini RenillaRef / Reiiillalni Firefly™ (Fig IB, boxed formula). Hence, an TRI > 1 indicates enhanced re-initiation and an TRI < 1 indicates reduced re-initiation, respectively. TRI provides a more stringent
normalized measurement for the validation of compounds identified by FireflyRe"ini/Renillalni reporter system.
EXAMPLE 2: Validation of the reporter system
The differential translation of the C/EBPp-mRNA into LAP and LIP protein isoforms is under control of the mTORCl/4E-BP/eIF4E pathway (Calkhoven et al, 2000; Zidek et al, 2015). To examine the performance of the reporter in response to mTORCl hyperactivation we separately transfectecl the pcDNA-FireflyRf' inVReniUaIm and pcDNA-FireflyRei7ReniUaR<!f reporter plasmids in mouse embryonic fibroblasts (MEFs) that are deficient in the mTORCl -inhibitory protein TSCl (TSCl-KO) (Kwiatkowski et al, 2002) or in wt MEFs and calculated the TRI. Compared to the wt MEFs the TRI was increased in the TSCl-KO MEFs, revealing enhanced translation re-initiation, which also resulted in enhanced expression of LIP (Fig 2A). A significant part of the mTORCl -dependent LIP regulation is mediated through the inhibitory phosphorylation of 4E-binding proteins (4E-BPs) by mTORCl, resulting in release of the eukaryotic translation initiation factor 4E (eIF-4E) (Zidek et al, 2015). In accordance, we found higher TRI values and increased LIP expression in 4E-BPl/2-double knockout (4E-BP1/2-DKO) MEFs comparing to wt MEFs (Fig 2B). mTORCl signaling can be
pharmacologically inhibited by the allosteric inhibitor rapamycin or the catalytic inhibitor PP424. To examine the performance of the reporter under pharmacological conditions we generated HEK293T based cell hnes with stable integrated pcDNA-Fire%Re inVRenilla[ni or pcDNA-FireflyRil|7RenillaR('f reporters. Treatment of the reporter cell lines with 200 nM rapamycin or 10 iiM PP242 decreased LIP expression and lowered TRI values, revealing suppression of re-initiation under inhibited mTORCl conditions (Fig 2C). The mTORCl inhibition was monitored by reduced phosphorylation of 4E- BP1 and S6 kinase (S6K). Finally, overexpression of eIF-4E that increases LIP expression accordingly increased TRI values (Fig 2D). Therefore, the reporter system reliably reflects differential translation of the C/ΕΒΡβ- mRNA under changing mTORC 1 signaling.
The eIF2a-kinases suppress post-uORF translation re-initiation through inhibitory phosphorylation of eIF2a in response to various stresses (Aitken & Lorsch, 2012; Sonenberg & Hinnebusch, 2009), which results in reduced expression of LIP (Calkhoven et al, 2000). Augmenting eIF2 activity by treatment with the eIF2a-kinase inhibitor C16, which reduces in eIF2a- phosphorylation, resulted in elevated translation re-initiation measured as higher TRI values and in an increase in LIP expression (Fig 2E). Similarly, activation of eIF2a by overexpression of the dominant negative mutant of the eIF2a-kinase PKR (PKRAG) or the non-phosphorylatable eIF2 S5lA mutant but not the phosphorylation mimicking eIF2aS51D mutant resulted higher TRI values (Fig 2D). Thus, the alterations in eIF2a-kinase signaling can be measured using the TRI reporter system.
In addition to the canonical translation initiation pathways we examined the effects of two proteins that were shown to determine the efficiency of translation re-initiation by different mechanisms. The density- regulated protein (DENR) has been identified as a key regulator of eukaryotic re-initiation downstream of uORFs, and knockdown of DENR impairs the post-uORF re-initiation of translation (Schleich et al, 2014). To examine the effect of DENR expression we generated HEK293T cells with a doxycycline-inducible DENR-shRNA expression vector. Induction of DENR- KD (+Dox) resulted in lower TRI comparing to control cells (Fig 2F), confirming the crucial role of DENR in uORF-dependent translation reinitiation. Recently, we have shown that deficiency in the Shwachman- Bodian-Diamond SyndiOme (SBDS) protein impairs translation re-initiation into the Ο/ΕΒΡβ-LIP isoform (In et al, 2016). In accordance, we measured lower TRI values in cells with shSBDS mediated knockdown of SBDS (Fig 2G).
Taken together, the TRI reporter system reliably detects changes in the ratio of initiation versus re-initiation by known translationally active drugs or alterations in key translation initiation factors and their regulatory kinases or non-canonical regulators of re-initiation.
EXAMPLE 3: Assay development and execution of a high- throughput screening (HTS)
To establish a reporter cell line suitable for high-throughput screening
(HTS) we generated HEK293T cells with stably integrated pcDNA-FireflyR - ini/Renillalni reporter plasmid. Single clones were tested for renilla and firefly expression and the highly expressing cell clones were in addition tested for stability of expression over repeated cell passages. Cell line clones that met these criteria were selected for further experiments. Similarly, a cell line with stably integrated pcDNA-FireflyRei/RenillaRei plasmid was generated. To determine an optimal assay window using reference
compounds we performed kinetic experiments with the mTORCl inhibitors rapamycin and PP242 over 24 hours. Both drugs showed the strongest decrease in TRI value at 8 hours, which was chosen as a suitable time point to perform the HTS (Fig EV2A).
We screened a library consisting of 780 FDA-approved drugs (ENZO #BML-2841) using the FireflyR<! ""/RenillaIm HEK293T reporter cell line. 1.5xl0 cells per well (50 μΐ volume) were seeded in a 384-well format and the FireflyRe'im/RenillaIni system was exposed to 10 μΜ of a single drug per well for 8h (three assay plates in total). Each assay plate contained 6 wells with 0.5% v/v DMSO added as reference (Fig 3A-C; bar graph at the right). Rapamycin was included as positive control on each plate (n=6) and induced a significant decrease in re-initiation/initiation ratio (FireflyRe-ini/¾enillalni) for each plate (plate 1, 0.82 ± 0.04; plate 2, 0.85 ± 0.025; plate 3, 0.9 ± 0.028) (Fig 3A-C; bar graph at the right). 45 drugs were found to decrease the translation re-initiation/initiation ratio (FireflyRe inVReniIlaTni) below the threshold of three times the standard deviation from the mean value of DMSO treated cells (hit rate 5.7%) (Fig 3A-C). The ENZO library contains three mTORCl inhibitors that are derived from ra amycin (the rapalogs everolimus, sirolimus and temsirolimus) and two of them, sirolimus and everolimus, decreased re-initiation/initiation ratio (Fig 3A, B). The temsirolimus signal was neglected because of aberrantly low luciferase signals (technical failure).
EXAMPLE 4: Validation of the drugs that decrease translation reinitiation/initiation ratio
Four drugs with the strongest effect on decreasing re-initiation/initiation ratio were chosen for further validation using counter assays (Fig 3A-C, numbered values #1-4)
clrug #l Raloxifene
drug #2 Febuxostat
drug #3 Thioguanine-6
drug #4 Adefovir Dipivoxil
See also Fig 3D for drug names and structural formulas. Treatment of the pcDNA-FireflyR f/Renilla}M or pcDNA3-FireflyRe ini/RemllaIni containing HEK293T cell line using 10 μΜ of each drug for 8 h in a 96-well format revealed for drug #3 and #4 enhancement of the renilla reference signal compared to DMSO control (Fig EV2B, C). The calculated TRIs (see formula Fig IB) revealed that similar to the result of the HTS screen drug #4 showed the strongest effect compared to drug #2 and #3, however, we could not confirm an effect of drug #1 on the TRI (Fig 4A). Only drug #4 significantly lowered TRI values in two additional different cell lines, the mouse hepatoma cell line Hepal-6 and the human breast cancer cell hne MCF-7 (Fig 4B, C). Drugs #2 did significantly lower the TRI in Hepal-6 but not in MCF7 cells, and drug #3 did not alter the TRI in Hepal-6 and even increased the TRI in MCF7 cells (Fig 4B, C). These two drugs were excluded from further examination. To examine dose-dependency of treatment the TRI was measured for serially diluted concentrations between 1 nM and 100 μΜ for drugs #4 compared to drug #1 (Fig EV2D). Drug #4 showed a dose- response with a half maximal effective concentration (EC 50) of 1 μΜ, while drug #1 showed a much higher EC50 of 70.4 μΜ. Cytotoxicity over 8 hours treatment was examined using fluorescent cell viability assay (CellTiter- Fluor, Promega) and revealed for both drugs #1 and #4 only serious cytotoxicity at 100 μΜ concentrations (Fig EV2E). In addition, treatment with drug #4 lowered endogenous LIP/LAP ratio comparable to treatment with rapamycin in HEK293T, Hepa 1-6 and MCF-7 cells (Fig 4D-F). Taken together, the counter assays revealed that drug #4 (Adefovir Dipivoxil) is best in affect re-initiation measured as reduced TRI and lower LIP levels in cells that originate from three different tissues.
We have recently shown that suppression of LIP expression results in a metabolic shift towards more fatty acid oxidation in cell culture
experiments and using a mouse model that is deficient in LIP expression (C/EBPpAu0RF mice) (Ziclek et al, 2015). To examine an eventual similar effect of drug #4 (Adefovir Dipivoxil) that also lowers LIP levels we measured fatty acid oxidation (FAO) of exogenously added palmitate-BSA in Hepal-6 cells using the Seahorse XF extracellular flux analyzer.
Surprisingly, treatment with Adefovir Dipivoxil resulted in an enhanced FAO-related oxygen consumption rate (OCR) that could be reverted by ectopic expression of LIP (Fig 5).
Taken together, our experiments show that the TRI reporter system can be successfully applied in HTS campaigns to identify compounds that alter the ratio between translation initiation and re-initiation depending on the C/EBPp-uORF-5'UTR, which potentially display CR-mimetic properties. REFERENCES
Aitken CE, Lorsch JR (2012) Nat Struct Mol Biol 19: 568-576 Albert V, Hall MN (2015) EMBO Rep 16: 881-882
Ashburn TT, Thor KB (2004) Nat Rev Drug Discov 3: 673-683 Barbosa C, Peixeiro I, Romao L (2013) PLoS Genet 9: el003529
Begay V, Smink JJ, Loddenkemper C, Zimmermann K, Rudolph C, Scheller M, Steinemann D, Leser U, Schlegelberger B, Stein H et al (2015) J Mol Med (Berl) 93: 39-49 Calkhoven CF, Muller C, Leutz A (2000) Genes Dev 14: 1920-1932
Descombes P, Schibler U (1991) Cell 67: 569-579
Esteves CL, Kelly V, Begay V, Man TY, Morton NM, Leutz A, Seckl JR, Chapman KE (2012) PLoS One 7: e37953
Fontana L, Partridge L, Longo VD (2010) Science 328: 321-326
Guarente L (2013) Genes Dev 27: 2072-2085
In K, Zaini MA, Muller C, Warren AJ, von Lindern M, Calkhoven CF (2016) Nucleic Acids Res 44: 4134-4146
Johnson SC, Rabinovitch PS, Kaeberlein M (2013) Nature 493: 338-345 Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S (2004) Curr Biol 14: 885-890
Karagiannides I, Tchkonia T, Dobson DE, Steppan CM, Cummins P, Chan G, Salvatori K, Hadzopoulou-Cladaras M, Kirkland JL (2001) Am J Physiol Regul Integr Comp Physiol 280: R1772-1780
Kennedy BK, Lamming DW (2016) Cell Metab 23: 990-1003 Kwiatkowski DJ, Zhang H, Bandura JL, Heiberger KM, Glogauer M, el- Hashemite N, Onda H (2002) Hum Mol Genet 11: 525-534
Le Bacquer O, Petroulakis E, Paglialunga S, Poulin F, Richard D, Cianflone K, Sonenberg N (2007) J Clin Invest 117: 387-396
Moffat JG, Rudolph J, Bailey D (2014) Nat Rev Drug Discov 13: 588-602
Rix U, Superti-Furga G (2009) Nat Chem Biol 5: 616-624 Schleich S, Strassburger K, Janiesch PC, Koledachkina T, Miller KK, Haneke K, Cheng YS, Kuchler K, Stoecklin G, Duncan KE et al (2014) Nature 512: 208-212
Selman C, Tullet JM, Wieser D, Irvine E, Lingard SJ, Choudhury AI, Claret M, Al-Qassab H, Carmignac D, Ramadani F et al (2009) Science 326: 140- 144
Shimobayashi M, HaU MN (2014) Nat Rev Mol Cell Biol 15: 155-162 Solon-Biet SM, McMahon AC, Ballard JW, Ruohonen K, Wu LE, Cogger VC, Warren A, Huang X, Pichaud N, Melvin RG et al (2014) Cell Metab 19: 418- 430 Sonenberg N, Hinnebusch AG (2009 CeU 136: 731-745
Stallone G, Infante B, Grandaliano G, Gesualdo L (2009) Transplantation 87: S23-26 Swinney DC, Anthony J (2011) Nat Rev Drug Discov 10: 507-519
Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J, Gao Y, Reichhng LJ, Sim T, Sabatini DM, Gray NS (2009) J Biol Chem 284: 8023-8032 Thoreen CC, Sabatini DM (2009) Autophagy 5: 725-726
Timchenko LT, Salisbury E, Wang GL, Nguyen H, Albrecht JH, Hershey JW, Timchenko NA (2006) J Biol Chem 281: 32806-32819 Wander SA, Hennessy BT, Slingerland JM (2011) J Clin Invest 121: 1231- 1241
Wethmar K, Begay V, Smink JJ, Zaragoza K, Wiesenthal V, Dorken B, Calkhoven CF, Leutz A (2010) Genes Dev 24: 15-20
Wiederschain D, Wee S, Chen L, Loo A, Yang G, Huang A, Chen Y,
Caponigro G, Yao YM, Lengauer C et al (2009) Cell Cycle 8: 498-504
Wiesenthal V, Leutz A, Calkhoven CF (2006a) Nat Protoc 1: 1531-1537 Wiesenthal V, Leutz A, Calkhoven CF (2006b) Nucleic Acids Res 34: e23
Willemsen AE, Grutters JC, Gerritsen WR, van Erp NP, van Herpen CM, Tol J (2016) Int J Cancer 138: 2312-2321
Wu JJ, Liu J, Chen EB, Wang JJ, Cao L, Narayan N, Fergusson MM, Rovira, II, AUen M, Springer DA et al (2013) Cell Rep 4: 913-920
Zahnow CA, Younes P, Laucirica R, Rosen JM (1997) J Natl Cancer Inst 89: 1887-1891
Zaytseva YY, Valentino JD, Gulhati P, Evers BM (2012) Cancer Lett 319: 1- 7 Zhang H, Cicchetti G, Onda H, Koon HB, Asrican K, Bajraszewski N,
Vazquez F, Carpenter CL, Kwiatkowski DJ (2003) J Chn Invest 112: 1223- 1233
Zidek LM, Ackermann T, Hartleben G, Eichwald S, Kortman G, Kiehntopf M, Leutz A, Sonenberg N, Wang ZQ, von Maltzahn J et al (2015) EMBO Rep 16: 1022-1036
Zoncu R, Efeyan A, Sabatini DM (2011) Nat Rev Mol CeU Biol 12: 21-35

Claims

Claims
1. A dual reporter plasmid system comprising a first nucleic acid sequence encoding a Renilla luciferase transcript and a second nucleic acid sequence encoding a firefly luciferase transcript, wherein said first and second nucleic acid sequence are both under control of the C/EBPp-5'UTR- uORF (upstream Open Reading Frame).
2. A cell comprising a dual reporter plasmid system according to claim 1.
3. Cell according to claim 2, wherein said plasmid is stably integrated into the genome of said cell.
4. A method for identifying a compound capable of mimicking calorie restriction comprising steps of
- providing a cell culture comprising cells according to claim 2 or 3;
- contacting said cell culture with a candidate compound to allow for luciferase expression;
determining expression of Renilla luciferase and firefly luciferase;
- calculating the translation re-initiation index (TRI) based on the
formula.. TRI = FireflyRe mitia, ,on x RenillaReference / RenillaTniiia(io x
Firefly Iteferenc ; and
- correlating said TRI with the capacity of said compound to mimic calorie restriction, wherein a reduction in TRI is a positive indicator.
5. Method according to claim 4, further comprising the use of a reference cell comprising a reference dual reporter plasmid system
comprising a first nucleic acid sequence encoding a Renilla luciferase transcript and a second nucleic acid sequence encoding a firefly luciferase transcript, that are not under the control of C/EBPp-5'UTR-uORF
sequences.
6. Kit-of-parts comprising a dual reporter plasmid system according to claim 1, a luciferase substrate, and optionally a reference reporter plasmid.
7. A high throughput screening assay comprising the use of a method according to claim 4 or 5 and/or a kit-of-parts according to claim 6.
8. Adefovir dipivoxil for use in a method for improving the metabolic health in a subject.
9. Adefovir dipivoxil for use according to claim 8, wherein improving the metabolic health comprises shifting the metabolism towards enhanced beta-oxidation.
10. Adefovir dipivoxil for use according to claim 8, wherein improving the metabolic health comprises enhancing or supporting weight loss in said subject.
11. Adefovir dipivoxil for use according to any one of claims 8-10, wherein the subject is a human subject.
12. Adefovir dipivoxil for use according to any one of claims 8-11, wherein the subject is suffering from, or at risk of developing, a condition selected from the group consisting of metabolic syndrome, obesity, type 2 diabetes, pre-diabetes, hypertension, disorders of lipid metabolism, insuhn resistance, endothelial dysfunction, pro-inflammatory state, and pro- coagulative state.
13. Adefovir dipivoxil for use as a calorie restriction mimetic.
14. A weight management composition comprising Adefovir dipivoxilin combination with a further active ingredient supporting enhancing weight loss.
PCT/NL2017/050689 2016-10-20 2017-10-20 Drugs mimicking calorie restriction and tools and methods for identifying the same WO2018074929A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16194897 2016-10-20
EP16194897.1 2016-10-20

Publications (2)

Publication Number Publication Date
WO2018074929A2 true WO2018074929A2 (en) 2018-04-26
WO2018074929A3 WO2018074929A3 (en) 2018-06-14

Family

ID=57209211

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2017/050689 WO2018074929A2 (en) 2016-10-20 2017-10-20 Drugs mimicking calorie restriction and tools and methods for identifying the same

Country Status (1)

Country Link
WO (1) WO2018074929A2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6475530B1 (en) 2000-05-31 2002-11-05 Eric H. Kuhrts Methods and compositions for producing weight loss
US20070054000A1 (en) 2000-01-18 2007-03-08 Nagaoka Perfumery Co., Ltd Anti-obestic composition
US20150265645A1 (en) 2010-05-24 2015-09-24 House Wellness Foods Corporation Composition For Prevention, Amelioration Or Treatment Of Metabolic Syndrome

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2708780A1 (en) * 2007-12-11 2009-06-18 Research Development Foundation Small molecules for neuronal differentiation of embryonic stem cells

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070054000A1 (en) 2000-01-18 2007-03-08 Nagaoka Perfumery Co., Ltd Anti-obestic composition
US6475530B1 (en) 2000-05-31 2002-11-05 Eric H. Kuhrts Methods and compositions for producing weight loss
US20150265645A1 (en) 2010-05-24 2015-09-24 House Wellness Foods Corporation Composition For Prevention, Amelioration Or Treatment Of Metabolic Syndrome

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
AITKEN CE; LORSCH JR, NAT STRUCT MOL BIOL, vol. 19, 2012, pages 568 - 576
ALBERT V; HALL MN, EMBO REP, vol. 16, 2015, pages 881 - 882
ASHBURN TT; THOR KB, NAT REV DRUG DISCOV, vol. 3, 2004, pages 673 - 683
BARBOSA C; PEIXEIRO I; ROMAO L, PLOS GENET, vol. 9, 2013, pages e1003529
BEGAY V; SMINK JJ; LODDENKEMPER C; ZIMMERMANN K; RUDOLPH C; SCHELLER M; STEINEMANN D; LESER U; SCHLEGELBERGER B; STEIN H ET AL., J MOL MED (BERL, vol. 93, 2015, pages 39 - 49
CALKHOVEN CF; MULLER C; LEUTZ A, GENES DEV, vol. 14, 2000, pages 1920 - 1932
DESCOMBES P; SCHIBLER U, CELL, vol. 67, 1991, pages 569 - 579
ESTEVES CL; KELLY V; BEGAY V; MAN TY; MORTON NM; LEUTZ A; SECKL JR; CHAPMAN KE, PLOS ONE, vol. 7, 2012, pages e37953
FONTANA L; PARTRIDGE L; LONGO VD, SCIENCE, vol. 328, 2010, pages 321 - 326
GUARENTE L, GENES DEV, vol. 27, 2013, pages 2072 - 2085
GUARENTE, NATURE REVIEWS MOLECULAR CELL BIOLOGY, vol. 13, 2012, pages 207
IN K; ZAINI MA; MULLER C; WARREN AJ; LINDERN M; CALKHOVEN CF, NUCLEIC ACIDS RES, vol. 44, 2016, pages 4134 - 4146
JOHNSON SC; RABINOVITCH PS; KAEBERLEIN M, NATURE, vol. 493, 2013, pages 338 - 345
KAPAHI P; ZID BM; HARPER T; KOSLOVER D; SAPIN V; BENZER S, CURR BIOL, vol. 14, 2004, pages 885 - 890
KARAGIANNIDES I; TCHKONIA T; DOBSON DE; STEPPAN CM; CUMMINS P; CHAN G; SALVATORI K; HADZOPOULOU-CLADARAS M; KIRKLAND JL, AM J PHYSIOL REGUL INTEGR COMP PHYSIOL, vol. 280, 2001, pages R1772 - 1780
KENNEDY BK; LAMMING DW, CELL METAB, vol. 23, 2016, pages 990 - 1003
KWIATKOWSKI DJ; ZHANG H; BANDURA JL; HEIBERGER KM; GLOGAUER M; EL-HASHEMITE N; ONDA H, HUM MOL GENET, vol. 11, 2002, pages 525 - 534
LE BACQUER O; PETROULAKIS E; PAGLIALUNGA S; POULIN F; RICHARD D; CIANFLONE K; SONENBERG N, J CLIN INVEST, vol. 117, 2007, pages 387 - 396
MOFFAT JG; RUDOLPH J; BAILEY D, NAT REV DRUG DISCOV, vol. 13, 2014, pages 588 - 602
RIX U; SUPERTI-FURGA G, NAT CHEM BIOL, vol. 5, 2009, pages 616 - 624
SCHLEICH S; STRASSBURGER K; JANIESCH PC; KOLEDACHKINA T; MILLER KK; HANEKE K; CHENG YS; KUCHLER K; STOECKLIN G; DUNCAN KE ET AL., NATURE, vol. 512, 2014, pages 208 - 212
SELMAN C; TULLET JM; WIESER D; IRVINE E; LINGARD SJ; CHOUDHURY AI; CLARET M; AL-QASSAB H; CARMIGNAC D; RAMADANI F ET AL., SCIENCE, vol. 326, 2009, pages 140 - 144
SHIMOBAYASHI M; HALL MN, NAT REV MOL CELL BIOL, vol. 15, 2014, pages 155 - 162
SOLON-BIET SM; MCMAHON AC; BALLARD JW; RUOHONEN K; WU LE; COGGER VC; WARREN A; HUANG X; PICHAUD N; MELVIN RG ET AL., CELL METAB, vol. 19, 2014, pages 418 - 430
SONENBERG N; HINNEBUSCH AG, CELL, vol. 136, 2009, pages 731 - 745
STALLONE G; INFANTE B; GRANDALIANO G; GESUALDO L, TRANSPLANTATION, vol. 87, 2009, pages 23 - 26
SWINNEY DC; ANTHONY J, NAT REV DRUG DISCOV, vol. 10, 2011, pages 507 - 519
THOREEN CC; KANG SA; CHANG JW; LIU Q; ZHANG J; GAO Y; REICHLING LJ; SIM T; SABATINI DM; GRAY NS, J BIOL CHEM, vol. 284, 2009, pages 8023 - 8032
THOREEN CC; SABATINI DM, AUTOPHAGY, vol. 5, 2009, pages 725 - 726
TIMCHENKO LT; SALISBURY E; WANG GL; NGUYEN H; ALBRECHT JH; HERSHEY JW; TIMCHENKO NA, J BIOL CHEM, vol. 281, 2006, pages 32806 - 32819
WANDER SA; HENNESSY BT; SLINGERLAND JM, J CLIN INVEST, vol. 121, 2011, pages 1231 - 1241
WEINDRUCH ET AL., SCIENCE, vol. 215, 1982, pages 1415
WEINDRUCH; CHARLES C. THOMAS ET AL.: "The Retardation of Aging and Disease by Dietary Restriction", 1988, SPRINGFIELD
WETHMAR K; BEGAY V; SMINK JJ; ZARAGOZA K; WIESENTHAL V; DORKEN B; CALKHOVEN CF; LEUTZ A, GENES DEV, vol. 24, 2010, pages 15 - 20
WIEDERSCHAIN D; WEE S; CHEN L; LOO A; YANG G; HUANG A; CHEN Y; CAPONIGRO G; YAO YM; LENGAUER C ET AL., CELL CYCLE, vol. 8, 2009, pages 498 - 504
WIESENTHAL V; LEUTZ A; CALKHOVEN CF, NAT PROTOC, vol. 1, 2006, pages 1531 - 1537
WIESENTHAL V; LEUTZ A; CALKHOVEN CF, NUCLEIC ACIDS RES, vol. 34, 2006, pages e23
WILLEMSEN AE; GRUTTERS JC; GERRITSEN WR; VAN ERP NP; VAN HERPEN CM; TOL J, INT J CANCER, vol. 138, 2016, pages 2312 - 2321
WU JJ; LIU J; CHEN EB; WANG JJ; CAO L; NARAYAN N; FERGUSSON MM; ROVIRA, II; ALLEN M; SPRINGER DA ET AL., CELL REP, vol. 4, 2013, pages 913 - 920
ZAHNOW CA; YOUNES P; LAUCIRICA R; ROSEN JM, J NATL CANCER INST, vol. 89, 1997, pages 1887 - 1891
ZAYTSEVA YY; VALENTINO JD; GULHATI P; EVERS BM, CANCER LETT, vol. 319, 2012, pages 1 - 7
ZHANG H; CICCHETTI G; ONDA H; KOON HB; ASRICAN K; BAJRASZEWSKI N; VAZQUEZ F; CARPENTER CL; KWIATKOWSKI DJ, J CLIN INVEST, vol. 112, 2003, pages 1223 - 1233
ZIDEK ET AL., EMBO REP., vol. 16, 2015, pages 1022 - 1036
ZIDEK LM; ACKERMANN T; HARTLEBEN G; EICHWALD S; KORTMAN G; KIEHNTOPF M; LEUTZ A; SONENBERG N; WANG ZQ; MALTZAHN J ET AL., EMBO REP, vol. 16, 2015, pages 1022 - 1036
ZONCU R; EFEYAN A; SABATINI DM, NAT REV MOL CELL BIOL, vol. 12, 2011, pages 21 - 35

Also Published As

Publication number Publication date
WO2018074929A3 (en) 2018-06-14

Similar Documents

Publication Publication Date Title
Keane et al. mTOR-dependent translation amplifies microglia priming in aging mice
Shimobayashi et al. Multiple amino acid sensing inputs to mTORC1
Fiedler et al. MAP4K4 inhibition promotes survival of human stem cell-derived cardiomyocytes and reduces infarct size in vivo
Chen et al. Response of rat muscle to acute resistance exercise defined by transcriptional and translational profiling
Zhang et al. S6K1 regulates GSK3 under conditions of mTOR-dependent feedback inhibition of Akt
Burbank et al. Early alterations of bile canaliculi dynamics and the rho kinase/myosin light chain kinase pathway are characteristics of drug-induced intrahepatic cholestasis
Tilstra et al. NF-κB inhibition delays DNA damage–induced senescence and aging in mice
Ma et al. Mammalian target of rapamycin regulates murine and human cell differentiation through STAT3/p63/Jagged/Notch cascade
Shioi et al. Rapamycin attenuates load-induced cardiac hypertrophy in mice
Buss et al. Cyclin-dependent kinase 6 phosphorylates NF-κB P65 at serine 536 and contributes to the regulation of inflammatory gene expression
Ghosh et al. Nontoxic chemical interdiction of the epithelial-to-mesenchymal transition by targeting cap-dependent translation
Malaguarnera et al. Metformin inhibits androgen-induced IGF-IR up-regulation in prostate cancer cells by disrupting membrane-initiated androgen signaling
Xu et al. The alteration of protein prenylation induces cardiomyocyte hypertrophy through Rheb–mTORC1 signalling and leads to chronic heart failure
Cho et al. Treatment of sepsis pathogenesis with high mobility group box protein 1-regulating anti-inflammatory agents
Cruz et al. Protein kinase A activation inhibits DUX4 gene expression in myotubes from patients with facioscapulohumeral muscular dystrophy
JP2011518126A (en) Methods for treating and screening IKKi inhibitors, and related IKKi diagnostic methods
Goodman et al. Dynamic changes to the skeletal muscle proteome and ubiquitinome induced by the E3 ligase, ASB2β
Hum et al. Mechanical loading in osteocytes induces formation of a Src/Pyk2/MBD2 complex that suppresses anabolic gene expression
Zaini et al. A screening strategy for the discovery of drugs that reduce C/EBPβ-LIP translation with potential calorie restriction mimetic properties
Kudelko et al. Label-free quantitative proteomics reveals survival mechanisms developed by hypertrophic chondrocytes under ER stress
Wang et al. The cytoplasmic tail of FPC antagonizes the full-length protein in the regulation of mTOR pathway
Ogura et al. Prenylated quinolinecarboxylic acid derivative suppresses immune response through inhibition of PAK2
Sato et al. Blockade of hedgehog signaling attenuates biliary cystogenesis in the polycystic kidney (PCK) rat
WO2018074929A2 (en) Drugs mimicking calorie restriction and tools and methods for identifying the same
US20150018383A1 (en) Biomarkers of renal disorders

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: 17792211

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17792211

Country of ref document: EP

Kind code of ref document: A2