WO2001025797A2 - Methods of evaluating whether a test agent is an agent affecting a leptin receptor - Google Patents

Methods of evaluating whether a test agent is an agent affecting a leptin receptor Download PDF

Info

Publication number
WO2001025797A2
WO2001025797A2 PCT/JP2000/006814 JP0006814W WO0125797A2 WO 2001025797 A2 WO2001025797 A2 WO 2001025797A2 JP 0006814 W JP0006814 W JP 0006814W WO 0125797 A2 WO0125797 A2 WO 0125797A2
Authority
WO
WIPO (PCT)
Prior art keywords
subunit
mammalian cell
obr
receptor
domain
Prior art date
Application number
PCT/JP2000/006814
Other languages
French (fr)
Other versions
WO2001025797A3 (en
Inventor
Toshio Kitamura
Yuki Isayama
Original Assignee
Sumitomo Chemical Company, Limited
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 Sumitomo Chemical Company, Limited filed Critical Sumitomo Chemical Company, Limited
Priority to AU74515/00A priority Critical patent/AU7451500A/en
Publication of WO2001025797A2 publication Critical patent/WO2001025797A2/en
Publication of WO2001025797A3 publication Critical patent/WO2001025797A3/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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6869Interleukin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • 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
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • G01N2333/715Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons

Definitions

  • the present invention relates to methods of evaluating whether a test agent is an
  • a leptin receptor is a particular homodimeric cytokine receptor that
  • OB leptin
  • OB is produced in fat cells and is released into the
  • OBR may then be activated by binding to OB and by having particular subunits thereof assembled into a
  • OBR When activated, OBR can transduce a particular signal into the
  • cytokine receptors allow a chimeric form thereof to transduce a particular signal by having the chimeric receptor bind to a different cognate ligand.
  • a reporter gene may be utilized as a major target of the signal transduced
  • the use of a reporter gene often leads to superfluous work.
  • the host cell of the chimeric cytokine receptor.
  • the host cell is a mammalian host cell
  • the host cell may tend to loose therefrom the plasmid containing the reporter gene after
  • a reporter gene furthermore, in which a significant amount of host cells in the batch may contain no reporter gene. Furthermore, a reporter gene furthermore, a reporter gene further
  • the present invention provides the following.
  • OBR said method comprising:
  • an OB bindable chimeric receptor comprising an extracellular domain of an OBR subunit, a transmembrane domain of a cytokine receptor subunit and an
  • cytokine receptor subunit gpl30 and IL-2R
  • a receptor subunit selected from a TPOR subunit, EPOR subunit, G-CSFR subunit, GHR subunit and PRLR subunit.
  • a culturing medium comprising OB.
  • a mammalian cell having:
  • cytokine receptor subunit gpl30 and IL-2R ⁇ subunit
  • intracellular domain is an intracellular domain of a non-OBR homodimeric cytokine receptor subunit.
  • a receptor subunit selected from gpl30 and IL-2R ⁇
  • EPOR is an intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR
  • G-CSFR subunit GHR subunit and PRLR subunit.
  • domain is a transmembrane domain of a receptor subunit selected from a homodimeric
  • cytokine receptor subunit gpl30 and IL-2R ⁇ subunit.
  • domain is a transmembrane domain of an OBR subunit.
  • a mammalian cell having:
  • cytokine receptor subunit selected from a non-OBR homodimeric cytokine receptor subunit, gpl30 and
  • transmembrane domains are intracellular and transmembrane domains of a receptor
  • a mammalian cell having:
  • an expression vector comprising a polynucleotide encoding a chimeric receptor subunit
  • cytokine receptor subunit and an intracellular domain of a receptor subunit selected
  • cytokine receptor subunit gpl30 and IL-2R ⁇ subunit
  • domain is an intracellular domain of a receptor subunit selected from a TPOR subunit
  • EPOR subunit G-CSFR subunit, GHR subunit and PRLR subunit.
  • a mammalian cell having: a chimeric receptor anchored to a cell membrane thereof, wherein the chimeric receptor
  • cytokine receptor subunit and an intracellular domain of a receptor subunit selected
  • cytokine receptor subunit gpl30 and IL-2R ⁇ subunit
  • receptor subunit comprises an extracellular domain of an OBR subunit, a transmembrane domain of a cytokine receptor subunit and an intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR subunit, GHR subunit, PRLR
  • a chimeric receptor subunit comprising an extracellular domain of an OBR
  • cytokine receptor subunit a transmembrane domain of a cytokine receptor subunit and an intracellular
  • a screening method comprising:
  • test agent which proliferates the mammalian cell.
  • An OBR activating agent comprising the test agent selected from the screening
  • An anti-obesity pharmaceutical comprising as an active ingredient, the OBR
  • a screening method comprising:
  • test agent which proliferates the mammalian cell.
  • An OBR inhibiting agent comprising the test agent selected from the screening
  • An anti-emaciation pharmaceutical comprising as an active ingredient, the OBR
  • Fig. 1 illustrates expression vector pMX-LTl which encodes an extracellular domain of an OBR subunit and transmembrane and intracellular domains of a TPOR subunit.
  • LTR refers to long terminal repeat
  • Y refers to a virus packaging
  • P refers to a SV40 initial promoter
  • NEO r refers to a neomyacin resistance
  • Ori refers to a replication origin of pMX-LTl and "Amp r " refers to a ampicillin
  • Fig. 2 illustrates expression vector pMX-LT2 which encodes extracellular
  • LTR long terminal repeat
  • Y refers to a virus packaging
  • P refers to a SV40 initial promoter
  • NEO r refers to a neomyacin resistance
  • Ori refers to a replication origin of pMX-LTl and "Amp r " refers to a ampicillin resistance gene.
  • OB leptin
  • OBR leptin receptor
  • IL-2R means interleukin-2 receptor.
  • TPOR means trombopoietin receptor.
  • EPOR means erythropoietin receptor
  • G-CSFR means granulocyte colony-stimulating factor receptor.
  • GHR growth hormone receptor
  • PRLR prolactin receptor
  • the present invention involves methods which entail utilizing a mammalian cell having a proliferation dependency which allows the mammalian cell to proliferate in
  • Such a mammalian cell in the methods contains a polynucleotide which encodes an OB-bindable chimeric receptor subunit.
  • test agent can then be exposed to the mammalian cell in order to evaluate whether the test agent is
  • test agent affect of the test agent can be measured, based on the proliferation of the mammalian
  • the polynucleotide which encodes the chimeric receptor subunit (hereinafter referred to as the chimeric polynucleotide) typically is efficiently expressed in the chimeric polynucleotide
  • the chimeric receptor subunit may then form a chimeric receptor complex anchored to the cell membrane of
  • the chimeric receptor subunit in this regard has an extracellular
  • ECD a transmembrane domain
  • TMD tumor necroscopy derived from a cell
  • ICD intracellular domain
  • TMD and ICD are typically arranged in the chimeric receptor subunit such that the
  • TMD is internally located between the ECD and ICD.
  • the ECD is on the
  • the ICD is on the C-terminal side of the TMD.
  • an appropriate ligand bind to the ECD of the chimeric receptor subunit typically
  • the signal may continue to transduce along a
  • An ECD of an OBR subunit is typically utilized for the ECD of the chimeric
  • the ECD of the chimeric receptor may also have one or more
  • Such an amount of amino acids added, deleted or substituted to the ECD of OBR may be 1 to 3 amino acids.
  • the addition, deletion or substitution of amino acids to the ECD of the OBR subunit is located at the extremity regions of the OBR subunit.
  • the ECD of OBR may additionally have 1 to 30 amino acids.
  • a mutation in the ECD of OBR may additionally have 1 to 30 amino acids.
  • nucleotide sequence encoding the ECD of the OBR subunit can set forth the desired
  • a TMD of a cytokine receptor subunit is typically utilized for the TMD of the
  • the TMD of the chimeric receptor subunit may also have one or more particular amino acids added, deleted or substituted to the TMD of the
  • cytokine receptor subunit Such an amount of amino acids added, deleted or substituted
  • deletion or substitution to the TMD of the cytokine receptor subunit is located at the
  • sequence encoding the cytokine receptor subunit can set forth the desired addition
  • ICD insulin receptor subunit
  • IL-2R ⁇ subunit IL-2R ⁇ subunit
  • gpl30 is typically utilized for the ICD of the
  • the ICD of the chimeric receptor subunit may have one or more particular amino acids added, deleted or substituted to such an ICD of a receptor
  • cytokine receptor subunit may be 1 to 3 amino acids. Typically, the addition, deletion
  • said ICD of receptor may additionally have 1 to 30 amino acids.
  • mutation in the nucleotide sequence encoding the ICD of the provided receptor subunit can set forth the desired addition, deletion or substitution of an amino acid to the ICD of
  • non-OBR homodimeric cytokine receptor subunit which may be utilized
  • cytokine for the TMD or ICD of the chimeric receptor subunit, is a homodimeric cytokine
  • the TMD or ICD of the chimeric receptor may utilize the TMD or
  • ICD of a receptor selected from a TPOR subunit, EPOR subunit, G-CSF subunit, GHR
  • the TMD of the chimeric receptor is the TMD of the chimeric receptor
  • TMD of a receptor subunit selected from an OBR subunit
  • non-OBR homodimeric cytokine receptor subunit IL-2R j3 subunit and gpl30.
  • the receptor subunit provided for the ICD of the chimeric receptor, such that the
  • chimeric receptor has the TMD and ICD derived from the identical receptor subunit.
  • such cases also include cases in which the chimeric receptor has the TMD
  • TMD of an OBR subunit such that the ECD and the TMD are derived
  • the chimeric polynucleotide can be produced by PCR amplifying from a
  • cytokine receptor subunit and a template encoding the ICD of the selected receptor subunit and by ligating together the amplified products.
  • the PCR amplification utilizes
  • primers possessing 14 to 35 nucleotides which can be produced with a DNA automatic synthesizer.
  • PCR amplifying a template encoding one of the desired domains one of
  • the primers has a nucleotide sequence of the 5' terminal nucleotide sequence encoding the desired domain (hereinafter referred to as the 5' primer). Another one of the primers
  • primers may further contain a nucleotide sequence of a restriction enzyme site at the 5'
  • restriction enzyme site is useful to ligate together the amplified products or to insert
  • the 5' terminus of the primers may be phosphorylated.
  • Such primers utilized in the PCR amplification are typically designed, based on
  • nucleotide sequence in the template encoding the provided domain. For example,
  • the primers can be designed based on the nucleotide sequence encoding
  • OBR subunit LA. Tartaglia et al., 1995, Cell 83: 1263-1271; WO9719952, such as
  • the primers may be designed, based on the nucleotide
  • EPOR subunit S.S.
  • primers having the nucleotide sequence shown in SEQ ID:12 or SEQ ID:13 or the like.
  • the primers can be any suitable primers.
  • the primers may be designed, based on the nucleotide
  • the primers may be designed, based on the nucleotide
  • the primers may be any suitable primers.
  • the PCR amplification can be performed for the ECD or ICD of the chimeric receptor subunit.
  • nucleotide sequence encoding the ECD is upstream
  • encoding the TMD is upstream from the nucleotide sequence encoding ICD.
  • Various forms of the template can be utilized in the PCR amplification. For example, Various forms of the template can be utilized in the PCR amplification.
  • the template may be a plasmid which encodes the desired domain, a cDNA or
  • a chromosomal DNA prepared from mammalian tissues such as the brain, liver and
  • Such templates can be produced according to conventional methods
  • marrow library (Clonetech) and the like, may be utilized to provide the template in the
  • PCR mixture is prepared and is incubated.
  • the PCR mixture may be prepared by
  • mixture may be a Takara Ex Taq (Takara Shuzo), pyrobest DNA polymerase (Takara
  • the incubations in the PCR amplification can be conducted by repeating 25 to
  • the heat denaturation can be, for example, conducted under the conditions of 94°C for
  • the annealing can be, for example, conducted under the
  • the elongation can be, for example, conducted under the conditions of a
  • incubations can be conducted with a thermal cycler (PE Applied Biosystems).
  • subunit can amplified when utilizing the primer having the nucleotide sequence as
  • the polynucleotide encoding the ECD of the OBR subunit can also be amplified
  • the polynucleotide encoding the ICD and TMD of the TPOR subunit can be amplified when utilizing the primer having the
  • subunit can be amplified when utilizing the primer having the nucleotide sequence as
  • the polynucleotide encoding the ICD and TMD of the EPOR subunit can be
  • the polynucleotide encoding the ICD of the G-CSFR subunit can be amplified when
  • the amplified products can then be ligated together to produce the chimeric
  • the amplified products can also be ligated together with a vector in one
  • the vector is useful for introducing the chimeric polynucleotide into a cytokine dependent mammalian host cell.
  • the vector comprises a promoter which is functional
  • the promoter in the mammalian cell and a selective marker gene. It is preferable that the promoter
  • neomyacin resistance gene such as a neomyacin resistance gene, puromyacin resistance gene and the like.
  • Such a selective marker gene can be utilized to confirm that the chimeric polynucleotide
  • retroviruses vector such as pMX (M. Onishi et al., 1996, Experimental Hematology
  • adenovirus vector such as pREP (Invitrogen), an EB vims vector such as pQBI
  • plasmid vector such as pcDNA (Invitrogen)
  • the retrovirus vector for example, can be introduced
  • the retrovirus vector can provide with the mammalian cell, a stable expression
  • the insertion of the chimeric polynucleotide or amplified products into the vector can be conducted according to conventional methods (for example the methods described in J. Sambrook, E.F. Frisch, T. Maniatis, Molecular Cloning 2 nd Edition,
  • a chimeric vector can be any suitable vector.
  • a chimeric vector can be any suitable vector.
  • the ICD of receptor subunit selected from a non-OBR homodimeric cytokine receptor
  • IL-2R j3 subunit IL-2R j3 subunit and g ⁇ l30.
  • An operably downstream site in the vector is
  • Such polynucleotides are generally arranged so that from
  • the polynucleotide encoding the TMD of the cytokine receptor and
  • a chimeric vector can be constructed as follows. The amplified
  • vector is restriction digested with a restriction enzyme at a restriction enzyme site
  • Ligation reaction kit such as Ligation
  • the resulting ligation product can be have its nucleotide
  • nucleotide sequence can be conducted by, for example, using a Thermo Sequenase II Die Terminator kit (Amerscham Pharmacia Biotech) with a DNA sequencer (PE
  • the mammalian cell of the present invention can be produced by introducing
  • the chimeric polynucleotide into a cytokine dependent mammalian host cell, the chimeric polynucleotide.
  • chimeric vector can be introduced into the cytokine dependent mammalian host cell to
  • polynucleotide into the cytokine dependent mammalian host cells there can be
  • polynucleotide into the cytokine dependent mammalian host cell can be confirmed by
  • the selective marker gene is an antibiotic resistance gene
  • Such a transformed cell can be selected by targeting for the antibiotic resistance.
  • the successful introduction of the chimeric polynucleotide can be confirmed by selecting a transformed cell which can proliferate in a culturing medium
  • polynucleotide is inserted, is a mammalian cell which dependently needs to be exposed
  • cytokine dependent mammalian host cells include a mammalian hematopoietic
  • mammalian epithelial cell examples include
  • hematopoietic cell include pluripotent mammalian hematopietic stem cells, cells which
  • Examples of cells which differentiate from pluripotent mammalian hematopietic cells include mammalian lymphocytes,
  • mammalian lymphocytes examples include mammalian lymphocyte stem cells,
  • mammalian lymphocyte stem cells include mammalian T cells, mammalian B cells,
  • lymphocytes there can be utilized as such lymphocytes, BaF-3 cells (RIKEN RCB0805 R. Palacios et al., 1985, Cell 41: 727-734), TALL-101 cells (B. Lange et al., 1987, Blood, 74:192-
  • FDC-Pl cells ATCC No. CRL-12103
  • TF-1 cells ATCC No.
  • mammalian cell of the present invention can be cloned by culturing
  • the concentration of OB in the culturing medium can be from 5pM
  • the level of stimulation from the OB or level of the proliferation dependency on OB can be measured by having the concentration of the OB in the
  • Such a level of stimulation from OB is a level based on the concentration of OB which provides a proliferating speed of 50% of the maximum proliferating speed thereof, in the culturing medium containing OB and
  • the mammalian cell can be utilized to evaluate whether a test
  • agent is an agent affecting OBR.
  • Such an evaluation may be provided by culturing the
  • the mammalian cell of the present invention is typically cultured under normal
  • Such a basic culturing medium can be a culturing medium in which fetal bovine
  • cytokine which is added to the basic culturing medium is a cytokine needed to
  • Such a particular cytokine may be interleukin-3 when
  • BaF-3 is utilized as the cytokine dependent mammalian host cell.
  • conditions to culture the mammalian cell is typically provided by an incubator in which
  • the temperature is set to provide a temperature of 37°C with 5% (v/v) CO 2 gas.
  • test agent is typically added
  • a basic culturing medium utilized for the mammalian cells.
  • a basic culturing medium utilized for the mammalian cells.
  • a basic culturing medium utilized for the mammalian cells.
  • a basic culturing medium utilized for the mammalian cells.
  • a basic culturing medium utilized for the mammalian cells.
  • a basic culturing medium utilized for the mammalian cells.
  • a basic culturing medium utilized for the mammalian cells.
  • culturing medium containing the test agent may be fractioned into a 96 well plate.
  • the cultured mammalian cells can then be fractioned into each of the wells and be further
  • test agent is usually added to the basic culturing medium so that the concentration of the test agent therein is InM to
  • ImM and preferably 50nM to 50 ⁇ M.
  • capabilities of said mammalian cell but may generally be 5x10 cells/ml to 5x10
  • the cultured mammalian cells can be washed with the basic
  • test agent the OB or the particular cytokine utilized to culture said mammalian cells.
  • the proliferation of the mammalian cells can be measured by utilizing
  • exposing the mammalian cell with the test agent is compared to a control culture.
  • cell of the present invention is unexposed to a test agent, a second control culture in
  • the first control culture can be cultured in the basic
  • the second and third controls can be cultured by using the basic culturing medium to which the particular cytokine needed to proliferate the cytokine dependent mammalian host cell is added.
  • the methods of the present invention can be utilized to screen a test agent as a
  • the OB-like activating abilities of the test agent can be
  • cytokines activating the chimeric receptor there is selected as the OB-
  • test agent which can provide a significantly higher proliferation
  • test agent usually provides no significant difference of proliferation between the second control
  • Such a OB-like activating agent is a substance other than OB, which has a physiological function substantially identical to OB, such as
  • test agent having the OB-like activating agent which can be screened from
  • the present invention can be, for example, N-(2-methoxyethyl) O-(5-methoxy-2-
  • the methods of the present invention can be utilized to screen a
  • test agent as an OB inhibiting agent.
  • the OB inhibiting abilities of the test agent can be
  • the concentration of OB in the utilized culturing medium is the concentration of OB in the utilized culturing medium
  • test agent selected as the OB inhibiting agent, a test agent which can provide a significantly lower
  • control in screening for the OB inhibiting agent is usually cultured with a corresponding
  • test agent usually provides no significant difference of proliferation between the second control culture and the third
  • a pharmaceutical composition comprising as an active ingredient, the OB-like
  • activating agent which is selected from the screening methods above, can be used as an
  • the OB inhibiting agent which is selected from the screening methods above, can be used as an anti-emaciation pharmaceutical
  • compositions comprising as the active
  • a mammal such as a human, can be applied orally or non-orally with an
  • compositions can be used as a typical formulation such as
  • compositions can be used as a typical liquid formulation such as a solution, emulsion,
  • formulations can be combined with an acceptable carrier, vehicle, adhesive,
  • composition may be O.lmg to 500mg. Further, such an effective amount may be provided.
  • Example 1 Production of the mammalian cells of the present invention containing a
  • polynucleotide encoding the ECD of an OBR subunit and the ICD of a TPOR subunit
  • polynucleotides encodes the ECD of an OBR subunit as well as the TMD and ICD
  • the second of the polynucleotides encodes the ECD and TMD of
  • a human brain cDNA library (CLONTECH) and a TF-1 cell cDNA library are provided.
  • first set of the primers is designed, based on the nucleotide sequence of a OBR subunit
  • Primer 1 has the nucleotide sequence shown in SEQ ID:1
  • Primer 2 has the nucleotide
  • Primer 3 has the nucleotide sequence shown in SEQ ID:3.
  • a second set of primers is designed, based on the nucleotide sequence of a TPOR subunit (GenBank Accession No. M90102). As such primers in the second set, Primers
  • Primer 4 has the nucleotide sequence shown in SEQ ID: 4
  • Primer 5 has the nucleotide sequence shown in SEQ ID: 4
  • nucleotide sequence shown in SEQ ID:5 and Primer 6 has the nucleotide sequence
  • restriction enzyme sites additionally contain a particular restriction enzyme site, so that restriction enzyme sites
  • a PCR amplification is conducted with 50 ⁇ l of a PCR mixture containing lOng
  • the PCR amplification has an incubation at 72°C for 10 minutes.
  • DNA 1 which moves in the agarose gel as a size
  • Primer 1 lOOpmol of Primer 3
  • 1.25U of the Pyrobest DNA polymerase 4. ⁇ l (2.5mM)
  • DNA 2 moves in the agarose gel as a size corresponding to about
  • a cDNA library is prepared from TF-1 cells (ATCC No. CRL-2003 T.
  • DNA 3 amplified DNA (hereinafter referred to as DNA 3), which moves in the agarose gel as a
  • DNA 4 the amplified DNA (hereinafter referred to as DNA 4), which moves in the agarose gel as a
  • Each of the amplified DNAs 1, 2, 3 and 4 are phenol-chloroform extracted and
  • nucleotide sequence shown in SEQ ID:8 are synthesized with a DNA automatic
  • Primers 7 and 8 are synthesized to additionally contain a particular restriction enzyme site.
  • a PCR amplification is then conducted with 50 l of a PCR solution containing
  • DNA 5 moves in the agarose gel as a size corresponding to about 1.5kbp.
  • DNA 5 is
  • restriction digest products of DNA 5 and pMX are subjected to agarose gel
  • the desired DNA fragments are obtained with Gene Clean Kit (BIO 101
  • the E. coli JM 109 transformant is cultured
  • DNA 3 and DNA 4 (prepared in (1.1)) are subjected, respectively, to a restriction digest
  • DNA 1, DNA 2, DNA 3, DNA 4 and pMX-neo are then prepared from the agarose gel with Gene Clean Kit (BIO 101 Inc.). Ten micro-liters (lO ⁇ l) of water is added, respectively, to each of the prepared
  • DNA fragments to produce aqueous solutions of the prepared DNAs 1, 2, 3, and 4.
  • aqueous solution of the prepared DNA 3 fragment are allowed to ligate together in 50 ⁇ l
  • the expression vectors pMX-LTl and pMX-LT2 are analyzed with a DNA
  • pMX-LTl contains a polynucleotide encoding the ECD of an OBR subunit as well as the TMD and ICD of a TPOR subunit.
  • PMX-LT2 is confirmed to contain a polynucleotide encoding amino acid 1 to 863 (from
  • polynucleotide encoding the ECD and TMD of an OBR subunit as well as ICD of a
  • the expression vectors prepared in (1.3), pMX-LTl and pMX-LT2, are transfected into virus packaging BOSC23 cells by using lipofectamine (GibcoBRL).
  • pMX-LTl a mixture is produced by combining a solution containing 3 ⁇ l of
  • D-MEM fetal bovine serum
  • FBS fetal bovine serum
  • the BOSC23 cells are then cultured for 5 hours in
  • the BOSC23 cells are cultured for 24
  • the expression vector pMX-LT2 is subjected under similar conditions
  • BaF-3 cells are cultured in a basic culturing medium additionally containing
  • Polyprene (hexadimethrine bromide, Sigma) is added to the concentration of lO ⁇ g/ml
  • mouse IL-3 is added to the concentration of lng/ml to the first and second viral
  • BaF-3 cells are then further cultured for 1 hour to produce first infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred to as cl cells
  • the cl cells and c2 cells are cultured, respectively, in a basic culturing medium
  • the cl cells and c2 cells are suspended to 5 cells/ml with a basic culturing
  • Example 2 Production of the mammalian cells of the present invention containing a
  • polynucleotide encoding the ECD of an OBR subunit as well as the TMD and ICD of an
  • a human brain cDNA library (CLONTECH) and a TF-1 cell cDNA library is provided.
  • Two sets of primers are designed to amplify the desired nucleotide sequence in the
  • a first set of the primers is designed, based on the
  • nucleotide sequence of an OBR subunit GenBank Accession No. U43168.
  • Primers 1 and 9 are synthesized with a DNA automatic synthesizer (PE).
  • Primer 1 has the nucleotide sequence shown in SEQ ID:1
  • Primer 9 has the nucleotide sequence shown in SEQ ID:9.
  • a second set of primers is shown in SEQ ID:9.
  • Primer 10 has the nucleotide sequence shown in SEQ ID: 10 and Primer 11 has the nucleotide sequence
  • restriction enzyme sites additionally contain a particular restriction enzyme site, so that restriction enzyme sites
  • a PCR amplification is conducted similarly to the conditions of (1.1) with 50 ⁇ l
  • Primer 1 lOOpmol of Primer 9, 1.25U of Pyrobest DNA polymerase (Takara Shuzo),
  • DNA 6 amplified DNA
  • DNA 7 which moves in the agarose gel to a size corresponding to about 780bp.
  • DNA7 are phenol-chloroform extracted and are collected by ethanol precipitation.
  • pMX-neo are then prepared from the agarose gel with Gene Clean Kit (BIO 101 Inc.).
  • aqueous solution of the prepared DNA 7 fragment are allowed to ligate together in 50 ⁇ l
  • transform a competent E. coli JM109 strain The transformants are cultured in 100ml of
  • the expression vector pMX-LE is then prepared from colonies of the transformant with QIAGEN Plasmid Maxi Prep.
  • the expression vector pMX-LE is analyzed with a DNA automatic sequencer
  • pMX-LE is confirmed to contain a polynucleotide
  • pMX-LE contains a polynucleotide encoding the ECD of an OBR
  • a viral medium is produced similarly to the procedures of (1.4) with pMX-LE.
  • TF-1 cells are cultured in a basic culturing medium additionally containing
  • human GM-CSF (hereinafter referred to as human GM-CSF) with 5% C0 gas and at 37°C .
  • TF-1 cells are centrifuged at lOOOrpm for 3 minutes to collect 1 x 10 5 cells thereof.
  • Polyprene (hexadimethrine bromide, Sigma) is added to the concentration of lO ⁇ g/ml and human GM-CSF is added to the concentration of lng/ml to the viral medium.
  • TF-1 cells are suspended in 1ml of the resulting viral medium and are then transferred
  • c3 cells infected TF-1 cells
  • the c3 cells are cultured in a basic culturing medium additionally containing
  • the c3 cells are: lng/ml of human GM-CSF and lmg/ml of GENETICIN (GibcoBRL).
  • the c3 cells are:
  • OB are transferred to a 96 well plate at lOO ⁇ l/well and are cultured for 5 days at 37°C
  • the cl cells and c2 cells are collected by centrifuging said cl
  • the cl cells and the c2 cells are suspended in a
  • the basic culturing medium and a basic culturing medium additionally
  • the suspensions of the c2 cells are also added, respectively, at
  • the cl cells and the c2 cells are cultured for 48 hours with 5%
  • the gain is set to 50
  • the excited wavelength is set to 595nm
  • the detection wavelength is set to 595nm.
  • the experiment is repeated 40 or 80 times.
  • the cl-8 cell is 13,000 and the for clone 7 of
  • the c2-7 cell is 11,000.
  • the cl-8 cells in a 100mm dish are cultured in lO ⁇ l of a basic
  • the cl-8 cells are suspended in basic culturing medium.
  • Culturing mediums are prepared by adding OB to basic culturing medium to
  • cl-8 cells is added at 90 ⁇ l/well (5x10 cells/well) to the basic culturing medium (final
  • Alamar blue is added to each of the wells. After further culturing the cl-8 cells for 24
  • Table 4 The values in Table 4 illustrate the resulting value from correspondingly
  • Example 4 Screening of a OB-like activating agent
  • cl-8 cells are cultured with 5% C0 2 gas at 37°C in lO ⁇ l of a
  • the cl-8 cells are suspended in a basic culturing medium.
  • Culturing mediums are prepared by adding a test agent to a basic culturing
  • test agent there is utilized the test agent
  • test agent specified amounts are added, respectively, to the wells of a 96 well
  • the cl-8 cells are then cultured for 48 hours at 37 C
  • Example 5 Screening of an OB inhibiting agent In a 100mm dish, cl-8 cells are cultured with 5% C0 gas at 37°C in lO ⁇ l of a
  • cl-8 cells are suspended in a basic culturing medium additionally containing 50pM of
  • Culturing mediums are prepared by adding to basic culturing mediums, OB to
  • test agent is added, respectively, to the wells of a 96 well plate at lO ⁇ l/well.
  • the suspension of the cl-8 cells is also added at 90 ⁇ l/well (5xl0 3 cells/well) to the
  • the cl-8 cells are then cultured for 48 hours at 37°C
  • An OB inhibiting agent can be then screened by selecting a test agent which
  • the test agent provides a level of proliferation
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.
  • An oligonucleotide primer designed for PCR amplification An oligonucleotide primer designed for PCR amplification.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Child & Adolescent Psychology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention provides methods of evaluating whether a test agent is an agent affecting the activity of OBR. In said methods, a mammalian cell is cultured and exposed to the test agent. The mammalian cell has a polynucleotide encoding a chimeric receptor subunit comprising an extracellular domain of an OBR subunit, a transmembrane domain of a cytokine receptor subunit and an intracellular domain of a receptor subunit selected from a non-OBR homodimeric cytokine receptor subunit, gp130 and IL-2R β subunit; and has a proliferation dependency on OB. By measuring the proliferation of the mammalian cell, it can be evaluated whether the test agent is an agent which can affect the activity of OBR.

Description

DESCRIPTION
METHODS OF EVALUATING WHETHER A TEST AGENT
IS AN AGENT AFFECTING A LEPTIN RECEPTOR
BACKGROUND OF THE INVENTION
Field of the Invention
The present invention relates to methods of evaluating whether a test agent is an
agent affecting the activity of a leptin receptor.
Description of Related Art
A leptin receptor (OBR) is a particular homodimeric cytokine receptor that
participates in regulating mammalian body weight and hematopoiesis by binding to its
cognate ligand, leptin (OB). OB is a type of cytokine, described in Y. Zhang, 1994,
Nature 372:425-432. Typically, OB is produced in fat cells and is released into the
blood stream, in order to reach a target cell containing OBR. OBR may then be activated by binding to OB and by having particular subunits thereof assembled into a
homodimer complex. When activated, OBR can transduce a particular signal into the
cell along a particular and intricate signal transduction pathway.
It is known that chimeric receptor complexes have proven quite productive for
analyzing the mechanism of cytokine receptor activation (Tartaglia et al.,
WO97/19952; Morella et al., 1995, J. Biol. Chem. 270:8298-8310; Baumann et al.,
1994, Mol. Cell. Biol. 14:138-146 ; Vigon et al, 1993, Oncogene 8:2607-2615). Certain characteristics of cytokine receptors allow a chimeric form thereof to transduce a particular signal by having the chimeric receptor bind to a different cognate ligand. In such cases, a reporter gene may be utilized as a major target of the signal transduced
from the chimeric cytokine receptor.
The use of a reporter gene often leads to superfluous work. The reporter gene
usually requires that a cell which can properly host the reporter gene be chosen as the
host cell of the chimeric cytokine receptor. When the host cell is a mammalian host cell,
the host cell may tend to loose therefrom the plasmid containing the reporter gene after
an extended amount of time. Such properties of such host cells can often necessitate
reintroducing the reporter gene into the host cell. Such properties may also result in
inadvertently culturing a batch of the host cells, in which a significant amount of host cells in the batch may contain no reporter gene. Furthermore, a reporter gene further
distances a system utilizing a chimeric cytokine receptor away from the native system
of the cytokine receptor. A system which is less alike the native system introduces
uncertainties which may decrease the reliability of the results achieved therefrom.
SUMMARY OF THE INVENTION
The present invention provides the following.
1. A method of evaluating whether a test agent is an agent affecting the activity of
OBR, said method comprising:
culturing a mammalian cell having:
an OB bindable chimeric receptor comprising an extracellular domain of an OBR subunit, a transmembrane domain of a cytokine receptor subunit and an
intracellular domain of a receptor subunit selected from a non-OBR homodimeric
cytokine receptor subunit, gpl30 and IL-2R |3 subunit; and
a proliferation dependency on OB; exposing the test agent to the mammalian cell; and
measuring the proliferation of the mammalian cell.
2. The method according to the above 1, wherein the intracellular domain is an
intracellular domain of a non-OBR homodimeric cytokine receptor subunit.
3. The method according to the above 1, wherein the intracellular domain is an
intracellular domain of a receptor subunit selected from gpl30 and IL-2R β subunit.
4. The method according to the above 1, wherein the intracellular domain is an
intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR subunit, G-CSFR subunit, GHR subunit and PRLR subunit.
5. The method according to the above 1, wherein the mammalian cell is cultured
with a culturing medium containing substantially no cytokine proteins activating the
chimeric receptor.
6. The method according to the above 1, wherein the mammalian cell is cultured
with a culturing medium comprising OB.
7. A mammalian cell having:
a polynuceotide encoding a chimeric receptor subunit comprising an extracellular
domain of an OBR subunit, a transmembrane domain of a cytokine receptor subunit and
an intracellular domain of a receptor subunit selected from a non-OBR homodimeric
cytokine receptor subunit, gpl30 and IL-2R β subunit; and
a proliferation dependency on OB.
8. The mammalian cell according to the above 7, wherein the mammalian cell is
derived from a hematopoietic mammalian cell.
9. The mammalian cell according to the above 7, wherein the mammalian cell is derived from a mammalian lymphocyte. 10. The mammalian cell according to the above 7, wherein the mammalian cell is
derived from a cell selected from BaF-3, TALL-101, OCl-Ly9, OCl-Lyl3.1, FDC-Pl,
TF-1, UT-7 and F-36-P.
11. The mammalian cell according to the above 10, wherein the mammalian cell is
derived from BaF-3.
12. The mammalian cell according to the above 7, wherein the intracellular domain is an intracellular domain of a non-OBR homodimeric cytokine receptor subunit.
13. The mammalian cell according to the above 7, wherein the intracellular domain
is an intracellular domain of a receptor subunit selected from gpl30 and IL-2R β
subunit.
14. The mammalian cell according to the above 7, wherein the intracellular domain
is an intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR
subunit, G-CSFR subunit, GHR subunit and PRLR subunit.
15. The mammalian cell according to the above 7, wherein said transmembrane
domain is a transmembrane domain of a receptor subunit selected from a homodimeric
cytokine receptor subunit, gpl30 and IL-2R β subunit.
16. The mammalian cell according to the above 15, wherein the transmembrane
domain is a transmembrane domain of an OBR subunit.
17. A mammalian cell having:
a polynucleotide encoding a chimeric receptor subunit comprising an extracellular
domain of an OBR subunit and intracellular and transmembrane domains of a receptor
subunit selected from a non-OBR homodimeric cytokine receptor subunit, gpl30 and
IL-2R β subunit; and
a proliferation dependency on OB. 18. The mammalian cell according to the above 17, wherein the intracellular and
transmembrane domains are intracellular and transmembrane domains of a receptor
subunit selected from a TPOR subunit, EPOR subunit, G-CSFR subunit, GHR subunit
and PRLR subunit.
19. A mammalian cell having:
an expression vector comprising a polynucleotide encoding a chimeric receptor subunit
comprising an extracellular domain of an OBR subunit, a transmembrane domain of a
cytokine receptor subunit and an intracellular domain of a receptor subunit selected
from a non-OBR homodimeric cytokine receptor subunit, gpl30 and IL-2R β subunit;
and
a proliferation dependency on OB.
20. The mammalian cell according to the above 19, wherein the intracellular
domain is an intracellular domain of a receptor subunit selected from a TPOR subunit,
EPOR subunit, G-CSFR subunit, GHR subunit and PRLR subunit.
21. A mammalian cell having: a chimeric receptor anchored to a cell membrane thereof, wherein the chimeric receptor
comprises an extracellular domain of an OBR subunit, a transmembrane domain of a
cytokine receptor subunit and an intracellular domain of a receptor subunit selected
from a non-OBR homodimeric cytokine receptor subunit, gpl30 and IL-2R β subunit;
and
a proliferation dependency on OB.
22. A polynucleotide encoding a chimeric receptor subunit, wherein the chimeric
receptor subunit comprises an extracellular domain of an OBR subunit, a transmembrane domain of a cytokine receptor subunit and an intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR subunit, GHR subunit, PRLR
subunit, gpl30 and IL-2R β subunit.
23. A chimeric receptor subunit comprising an extracellular domain of an OBR
subunit, a transmembrane domain of a cytokine receptor subunit and an intracellular
domain of a receptor subunit selected from a TPOR subunit, EPOR subunit, GHR
subunit, PRLR subunit, gpl30 and IL-2R ]3 subunit.
24. A screening method comprising:
conducting the method the above 1 on a plurality of test agents and
selecting a test agent which proliferates the mammalian cell.
25. An OBR activating agent comprising the test agent selected from the screening
method of the above 24.
26. An anti-obesity pharmaceutical comprising as an active ingredient, the OBR
activating agent of the above 25.
27. A screening method comprising:
conducting the method of the above 1 on a plurality of test agents and
selecting a test agent which proliferates the mammalian cell.
28. An OBR inhibiting agent comprising the test agent selected from the screening
method of the above 27.
29. An anti-emaciation pharmaceutical comprising as an active ingredient, the OBR
inhibiting agent of the above 28.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 illustrates expression vector pMX-LTl which encodes an extracellular domain of an OBR subunit and transmembrane and intracellular domains of a TPOR subunit. In Fig. 1, "LTR" refers to long terminal repeat, "Y" refers to a virus packaging
signal, "P" refers to a SV40 initial promoter, "NEOr" refers to a neomyacin resistance
gene, "Ori" refers to a replication origin of pMX-LTl and "Ampr" refers to a ampicillin
resistance gene.
Fig. 2 illustrates expression vector pMX-LT2 which encodes extracellular and
transmembrane domains of an OBR subunit and an intracellular domains of a TPOR
subunit. In Fig. 2, "LTR" refers to long terminal repeat, "Y" refers to a virus packaging
signal, "P" refers to a SV40 initial promoter, "NEOr" refers to a neomyacin resistance
gene, "Ori" refers to a replication origin of pMX-LTl and "Ampr" refers to a ampicillin resistance gene.
DETAILED DESCRIPTION OF THE INVENTION
As used herein, the following abbreviations will abbreviate the terms indicated below.
"OB" means leptin.
"OBR" means leptin receptor.
"IL-2R" means interleukin-2 receptor.
"TPOR" means trombopoietin receptor.
"EPOR" means erythropoietin receptor.
"G-CSFR" means granulocyte colony-stimulating factor receptor.
"GHR" means growth hormone receptor.
"PRLR" means prolactin receptor.
The present invention involves methods which entail utilizing a mammalian cell having a proliferation dependency which allows the mammalian cell to proliferate in
response to a stimulation from OB. Such a mammalian cell in the methods contains a polynucleotide which encodes an OB-bindable chimeric receptor subunit. A test agent
can then be exposed to the mammalian cell in order to evaluate whether the test agent is
an agent affecting OBR. When the mammalian cell is exposed to the test agent, the
affect of the test agent can be measured, based on the proliferation of the mammalian
cell.
The polynucleotide which encodes the chimeric receptor subunit (hereinafter referred to as the chimeric polynucleotide) typically is efficiently expressed in the
mammalian cell to provide for the chimeric receptor subunit. The chimeric receptor subunit may then form a chimeric receptor complex anchored to the cell membrane of
the mammalian cell. The chimeric receptor subunit in this regard has an extracellular
domain (hereinafter referred to as ECD), a transmembrane domain (hereinafter referred
to as TMD) and an intracellular domain (hereinafter referred to as ICD). The ECD,
TMD and ICD are typically arranged in the chimeric receptor subunit such that the
TMD is internally located between the ECD and ICD. Typically, the ECD is on the
N-terminal side of the TMD and the ICD is on the C-terminal side of the TMD.
The signal triggered by forming the chimeric receptor complex and by having
an appropriate ligand bind to the ECD of the chimeric receptor subunit typically
transduces through the ICD of the chimeric receptor subunit. After the signal is
transduced to the ICD of the subunit, the signal may continue to transduce along a
native signal transduction pathway of the mammalian cell.
An ECD of an OBR subunit is typically utilized for the ECD of the chimeric
receptor subunit. The ECD of the chimeric receptor may also have one or more
particular amino acids added, deleted or substituted to the ECD of the OBR subunit.
Such an amount of amino acids added, deleted or substituted to the ECD of OBR may be 1 to 3 amino acids. Typically, the addition, deletion or substitution of amino acids to the ECD of the OBR subunit is located at the extremity regions of the OBR subunit. In
having additional amino acids at the N-terminus of the ECD of the chimeric receptor,
the ECD of OBR may additionally have 1 to 30 amino acids. A mutation in the
nucleotide sequence encoding the ECD of the OBR subunit can set forth the desired
addition, deletion or substitution of an amino acid to the ECD of the OBR subunit.
A TMD of a cytokine receptor subunit is typically utilized for the TMD of the
chimeric receptor subunit. The TMD of the chimeric receptor subunit may also have one or more particular amino acids added, deleted or substituted to the TMD of the
cytokine receptor subunit. Such an amount of amino acids added, deleted or substituted
to the TMD of the cytokine receptor may be 1 to 3 amino acids. Typically, the addition,
deletion or substitution to the TMD of the cytokine receptor subunit is located at the
extremity regions of the chimeric receptor subunit. A mutation in the nucleotide
sequence encoding the cytokine receptor subunit can set forth the desired addition,
deletion or substitution of an amino acid to the TMD of the cytokine receptor subunit.
An ICD of a receptor subunit selected from a non-OBR homodimeric cytokine
receptor subunit, IL-2R β subunit and gpl30, is typically utilized for the ICD of the
chimeric receptor subunit. The ICD of the chimeric receptor subunit may have one or more particular amino acids added, deleted or substituted to such an ICD of a receptor
subunit. Such an amount of amino acids added, deleted or substituted to the ECD of the
cytokine receptor subunit may be 1 to 3 amino acids. Typically, the addition, deletion
or substitution to the ICD of the provided receptor subunit is located at the extremity
regions thereof. In having additional amino acids at the C-terminus of the ICD of the
chimeric receptor, said ICD of receptor may additionally have 1 to 30 amino acids. A
mutation in the nucleotide sequence encoding the ICD of the provided receptor subunit can set forth the desired addition, deletion or substitution of an amino acid to the ICD of
said provided receptor subunit.
The non-OBR homodimeric cytokine receptor subunit, which may be utilized
for the TMD or ICD of the chimeric receptor subunit, is a homodimeric cytokine
receptor excluding OBR. For example, when utilizing the TMD or ICD of the non-
OBR homodimeric cytokine receptor correspondingly as the TMD or ICD of the chimeric receptor, the TMD or ICD of the chimeric receptor may utilize the TMD or
ICD of a receptor selected from a TPOR subunit, EPOR subunit, G-CSF subunit, GHR
subunit and PRLR subunit.
There are cases in which the chimeric receptor subunit can have the TMD
thereof derived from the TMD of the receptor subunit utilized for the ECD or ICD of
the chimeric receptor subunit. In such cases, the TMD of the chimeric receptor
generally utilizes therein a TMD of a receptor subunit selected from an OBR subunit,
non-OBR homodimeric cytokine receptor subunit, IL-2R j3 subunit and gpl30. For
example, there includes cases in which the TMD of the chimeric receptor is the TMD of
the receptor subunit provided for the ICD of the chimeric receptor, such that the
chimeric receptor has the TMD and ICD derived from the identical receptor subunit.
Further, such cases also include cases in which the chimeric receptor has the TMD
thereof be the TMD of an OBR subunit, such that the ECD and the TMD are derived
from the OBR subunit.
The chimeric polynucleotide can be produced by PCR amplifying from a
template encoding the ECD of an OBR subunit, a template encoding the TMD of a
cytokine receptor subunit and a template encoding the ICD of the selected receptor subunit and by ligating together the amplified products. The PCR amplification utilizes
primers possessing 14 to 35 nucleotides, which can be produced with a DNA automatic synthesizer. In PCR amplifying a template encoding one of the desired domains, one of
the primers has a nucleotide sequence of the 5' terminal nucleotide sequence encoding the desired domain (hereinafter referred to as the 5' primer). Another one of the primers
in such cases has a nucleotide sequence complementary to the 3' terminal nucleotide
sequence encoding the desired domain (hereinafter referred to as the 3' primer). Such
primers may further contain a nucleotide sequence of a restriction enzyme site at the 5'
terminus thereof, so that the restriction enzyme site is added to the amplified product. The restriction enzyme site is useful to ligate together the amplified products or to insert
the constructed chimeric polynucleotide into an expression vector, as described below.
The 5' terminus of the primers may be phosphorylated.
Such primers utilized in the PCR amplification are typically designed, based on
the nucleotide sequence in the template, encoding the provided domain. For example,
when the template encodes the ECD or TMD of an OBR subunit in the PCR
amplification, the primers can be designed based on the nucleotide sequence encoding
an OBR subunit (LA. Tartaglia et al., 1995, Cell 83: 1263-1271; WO9719952), such as
a primer having a nucleotide sequence shown in SEQ ID:1, SEQ ID:2, SEQ ID:3 or SEQ ID:9 or the like. When the template encodes the TMD or ICD of a TPOR subunit
in the PCR amplification, the primers may be designed, based on the nucleotide
sequence encoding a TPOR subunit (I. Vigon et al., 1994, Genomics 20(1): 5-12 and
R.C. Skoda et al., EMBO 12(7): 2645-2653), such as a primer having a nucleotide
sequence shown in SEQ ID:4, SEQ ID:5 or SEQ ID:6 or the like. When the template
encodes the TMD or ICD of an EPOR subunit, in the PCR amplification, the primers
may be designed, based on the nucleotide sequence encoding an EPOR subunit (S.S.
Jones et al., 1990, Blood 76: 31-35; A.D. D'Andrea et al., 1989, Cell 57 (2):277-285 and S. Nagao et al., 1993, J. Biol. Chem. 268 15:11208-11216), such as a primer having a nucleotide sequence shown in SEQ ID: 10 or SEQ ID: 11 or the like. When the
template encodes the ICD of a G-CSFR subunit in the PCR amplification, the primers
can be designed, based on the nucleotide sequence encoding a G-CSFR subunit (R.
Fukunaga et al., 1990, Proc. Natl. Acad. Sci. U.S.A. 87: 8702-8706), such as the
primers having the nucleotide sequence shown in SEQ ID:12 or SEQ ID:13 or the like.
When the template encodes the TMD or ICD of a GHR subunit, the primers can be
designed, based on the nucleotide sequence encoding a GHR subunit (D.W. Leung et
al., 1987, Nature 330: 537-543; GenBank Accession No. NM_010284and LS.
Mathews et al., 1989, J. Biol. Chem. 264: 9905-9910). When the template encodes the TMD or ICD of a PRLR subunit, the primers may be designed, based on the nucleotide
sequence encoding a PRLR subunit (J.M. Boutin et al., 1989, Mol. Endocrinol. 3:
1455-1461; D.L. Clarke et al.,1993, Endocrinology 133:224-232 and R. Zhang et al.,
1990, Biochem. Biophys. Res. Commun. 168(2):415-422). When the template encodes
the TMD or ICD of gpl30, the primers may be designed, based on the nucleotide
sequence encoding gpl30 (M. Hibi et al., 1990, Cell 63: 149-1157; M. Saito, 1992, J.
Immunol. 148(12):4066-4071 and Y. Wang et al., 1992, Genomics, 14:666-672).
When the template encodes the TMD or ICD of an IL-2 β subunit, the primers may be
designed, based on the nucleotide sequence encoding an IL-2 β subunit (M.
Hatakeyama et al, 1989, Science 244: 551-556; A. Shimizu et al., 1985, Nucleic Acid
Res. 13:1505-1516 and T.H. Page et al., 1991, Eur. J. Immunol. 21: 2133-2138).
When producing the chimeric polynucleotide encoding the chimeric receptor
subunit in which the TMD thereof is derived from the identical receptor subunit utilized
for the ECD or ICD of the chimeric receptor subunit, the PCR amplification can
continuously amplify a template encoding such domains in connection to provide an amplified product in which the nucleotide sequence of the ECD is connected with the
nucleotide sequence of the TMD or in which the nucleotide sequence of TMD is
connected with the nucleotide sequence of the ICD. Further, the template in such cases
are typically arranged such that the nucleotide sequence encoding the ECD is upstream
from the nucleotide sequence encoding the TMD or that the nucleotide sequence
encoding the TMD is upstream from the nucleotide sequence encoding ICD.
Various forms of the template can be utilized in the PCR amplification. For
example, the template may be a plasmid which encodes the desired domain, a cDNA or
a chromosomal DNA prepared from mammalian tissues such as the brain, liver and
lungs and the like. Such templates can be produced according to conventional methods
(such as the methods described in J. Sambrook, E.F. Frisch, T. Maniatis, Molecular
Cloning 2nd Edition, Cold Springs Harbor Laboratory press). Further, a commercially
available library, such as a human brain cDNA library (Stratagene), human bone
marrow library (Clonetech) and the like, may be utilized to provide the template in the
PCR amplification .
To PCR amplify the templates encoding the desired domains, an appropriate
PCR mixture is prepared and is incubated. The PCR mixture may be prepared by
mixing in a reaction buffer, the 5' primer, 3' primer, the template, a heat-resistant DNA
polymerase and a dNTP mixture. The 5' primer and 3' primer in the PCR mixture
correspond to the template in the PCR mixture, in that said 5' primer and 3' primer can
anneal to the template. The heat-resistant DNA polymerase in the PCR amplification
mixture may be a Takara Ex Taq (Takara Shuzo), pyrobest DNA polymerase (Takara
Shuzo) or the like.
The incubations in the PCR amplification can be conducted by repeating 25 to
40 times an incubation cycle including heat denaturation, annealing and elongation. The heat denaturation can be, for example, conducted under the conditions of 94°C for
10 seconds to 1 minute. The annealing can be, for example, conducted under the
conditions of a temperature of from 35°C to the meting point of the primers (Tm value)
for 1 minute. The elongation can be, for example, conducted under the conditions of a
72°C for 1 to 5 minutes. After repeating the incubation cycle in the PCR amplification,
an incubation for a elongation reaction of 72°C for 10 minutes may be added. Such
incubations can be conducted with a thermal cycler (PE Applied Biosystems).
In the above PCR amplification, a polynucleotide encoding the ECD of the OBR
subunit can amplified when utilizing the primer having the nucleotide sequence as
shown in SEQ ID: 1 and the primer having the nucleotide sequence as shown in SEQ
ID: 2. The polynucleotide encoding the ECD of the OBR subunit can also be amplified
when utilizing the primer having the nucleotide sequence as shown in SEQ ID: 1 and
the primer having the nucleotide sequence as shown in SEQ ID: 9. The polynucleotide
encoding the ECD and TMD of the OBR subunit can be amplified when utilizing the
primer having the nucleotide sequence as shown in SEQ ID: 1 and the primer having the
nucleotide sequence as shown in SEQ ID: 3. The polynucleotide encoding the ICD and TMD of the TPOR subunit can be amplified when utilizing the primer having the
nucleotide sequence as shown in SEQ ID: 4 and the primer having the nucleotide
sequence as shown in SEQ ID: 6. The polynucleotide encoding the ICD of the TPOR
subunit can be amplified when utilizing the primer having the nucleotide sequence as
shown in SEQ ID: 5 and the primer having the nucleotide sequence as shown in SEQ
ID: 6. The polynucleotide encoding the ICD and TMD of the EPOR subunit can be
amplified when utilizing the primer having the nucleotide sequence as shown in SEQ
ID: 10 and the primer having the nucleotide sequence as shown in SEQ ID: 11. The polynucleotide encoding the ICD of the G-CSFR subunit can be amplified when
utilizing the primer having the nucleotide sequence as shown in SEQ ID: 12 and the
primer having the nucleotide sequence as shown in SEQ ID: 13.
The amplified products can then be ligated together to produce the chimeric
polynucleotide. The amplified products can also be ligated together with a vector in one
pot to construct a chimeric vector.
The vector is useful for introducing the chimeric polynucleotide into a cytokine dependent mammalian host cell. The vector comprises a promoter which is functional
in the mammalian cell and a selective marker gene. It is preferable that the promoter
can also functional in conventional host organisms such as Escherichia coli and yeast.
As an example of the selective marker gene, there is mentioned an antibiotic resistance
gene such as a neomyacin resistance gene, puromyacin resistance gene and the like.
Such a selective marker gene can be utilized to confirm that the chimeric polynucleotide
has been introduced into the mammalian cell. Examples of such vectors include a
retroviruses vector such as pMX (M. Onishi et al., 1996, Experimental Hematology
24:324-329), pBabe-neo (J. P. Morgenstern et al. Nucleic Acid Res. 18: 3587-3596) and pBabe-puro (J. P. Morgenstern et al. Nucleic Acid Res. 18: 3587-3596), an
adenovirus vector such as pREP (Invitrogen), an EB vims vector such as pQBI
(QUANTUM BIOTECHNOLOGIES), a plasmid vector such as pcDNA (Invitrogen)
and pRc (Invitrogen) and the like. The retrovirus vector, for example, can be introduced
into the mammalian cell to provide a high efficacy rate with cell infection methods.
Further, the retrovirus vector can provide with the mammalian cell, a stable expression
of the chimeric polynucleotide therein.
The insertion of the chimeric polynucleotide or amplified products into the vector can be conducted according to conventional methods (for example the methods described in J. Sambrook, E.F. Frisch, T. Maniatis, Molecular Cloning 2nd Edition,
Cold Springs Harbor Laboratory press). In this regard, a chimeric vector can be
constructed by inserting operably downstream from the promoter the polynucleotides
encoding the ECD of the OBR subunit, the TMD of the cytokine receptor subunit and
the ICD of receptor subunit selected from a non-OBR homodimeric cytokine receptor
subunit, IL-2R j3 subunit and gρl30. An operably downstream site in the vector is
usually located therein so that the promoter and the chimeric polynucleotide are in a
relationship permitting the promoter to induce the expression of the chimeric polynucleotide therein. Such polynucleotides are generally arranged so that from
upstream to downstream, respectively, there is said polynucleotide encoding the ECD
of the OBR subunit, the polynucleotide encoding the TMD of the cytokine receptor and
then the polynucleotide encoding the ICD of selected subunit.
For example, a chimeric vector can be constructed as follows. The amplified
products which contain the restriction enzyme sites added by the primers, as described
above, are restriction digested with the appropriate restriction enzyme. Further, the
vector is restriction digested with a restriction enzyme at a restriction enzyme site
operably downstream from the promoter therein. Such restriction digested fragments
can be then subjected to agarose gel electrophoresis to obtain the desired
polynucleotides with Gene Clean Kit (BIO 101 Inc.). Such obtained polynucleotides
are ligated together with a commercially available ligation reaction kit, such as Ligation
Kit Ver.l (Takara Shuzo). The resulting ligation product can be have its nucleotide
sequence analyzed to confirm that the polynucleotide encoding the desired chimeric
receptor is in an expressible form in the constructed chimeric vector. Analysis of the
nucleotide sequence can be conducted by, for example, using a Thermo Sequenase II Die Terminator kit (Amerscham Pharmacia Biotech) with a DNA sequencer (PE
Applied Biosystems).
The mammalian cell of the present invention can be produced by introducing
into a cytokine dependent mammalian host cell, the chimeric polynucleotide. The
chimeric vector can be introduced into the cytokine dependent mammalian host cell to
provide for the chimeric polynucleotide. As methods for introducing the chimeric
polynucleotide into the cytokine dependent mammalian host cells, there can be
mentioned a retrovirus infection method (M. Onishi et al., 1996, Experimental
Hematology 24:324-329), electroporation method (A. ting et al. EMBO J. 15:6189-
6196) potassium phosphate method (S. Grimm et al. Proc. Natl. Acad. Sci. USA
93:10923-10927) and the like. A successful introduction of the chimeric
polynucleotide into the cytokine dependent mammalian host cell can be confirmed by
selecting a transformed cell which comprises the characteristics provided by the
selective marker gene. When the selective marker gene is an antibiotic resistance gene,
such a transformed cell can be selected by targeting for the antibiotic resistance.
Alternatively, the successful introduction of the chimeric polynucleotide can be confirmed by selecting a transformed cell which can proliferate in a culturing medium
containing OB and substantially no cytokines.
Such a cytokine dependent mammalian host cell, to which the chimeric
polynucleotide is inserted, is a mammalian cell which dependently needs to be exposed
to a cytokine to proliferate, with the proviso that the cytokine is not OB. Examples of
such cytokine dependent mammalian host cells include a mammalian hematopoietic
cell, mammalian epithelial cell and the like. Examples of the mammalian
hematopoietic cell include pluripotent mammalian hematopietic stem cells, cells which
differentiate therefrom and the like. Examples of cells which differentiate from pluripotent mammalian hematopietic cells include mammalian lymphocytes,
mammalian bone marrow cells, blood cells differentiating therefrom and the like.
Examples of such mammalian lymphocytes include mammalian lymphocyte stem cells,
cells differentiating therefrom and the like. Examples of such cells differentiating from
mammalian lymphocyte stem cells include mammalian T cells, mammalian B cells,
mammalian NK cells, blood cells differentiating therefrom and the like. In this regard,
there can be utilized as such lymphocytes, BaF-3 cells (RIKEN RCB0805 R. Palacios et al., 1985, Cell 41: 727-734), TALL-101 cells (B. Lange et al., 1987, Blood, 74:192-
199), OCl-Ly9 cells (N. Tweeddale et al., 1989, Blood, 74: 572-578), OCl-Lyl3.1
cells (N. Tweeddale, 1989, Blood 74: 572-578) and the like. As the bone marrow cells,
there may be utilized FDC-Pl cells (ATCC No. CRL-12103), TF-1 cells (ATCC No.
CRL-2003, T. Kitamura et al., 1989, J. Cell. Physiol. 140:323-334), Ut-7 cells (N.
Komatsu et al., 1991, Cancer Res. 51:341-348), F36-P cells (RIKEN RCB0775 S.
Chiba et al., 1991, Blood 78: 2261-2268) and the like. As the epithelial cells, there is
mentioned HK-2 (ATCC No. CRL-2190).
Further, the mammalian cell of the present invention can be cloned by culturing
the mammalian cell containing the chimeric polynucleotide in a culturing medium
containing OB and substantially no cytokines and then by utilizing conventional cloning methods such as the methylcellulose method, penicillin cup method and the
like. When cloning, the concentration of OB in the culturing medium can be from 5pM
to InM. In such cases, there is cloned, a mammalian cell which receives an efficient
level of stimulation from the OB and which has an efficient level of proliferation
dependency on OB. The level of stimulation from the OB or level of the proliferation dependency on OB can be measured by having the concentration of the OB in the
culturing medium at various levels. Such a level of stimulation from OB is a level based on the concentration of OB which provides a proliferating speed of 50% of the maximum proliferating speed thereof, in the culturing medium containing OB and
substantially no cytokines. An inverse relationship is present between the level of
stimulation from OB and the concentration of OB in said culturing medium containing
OB and substantially no cytokines, such that the level of stimulation is higher as the
concentration of OB is lower.
In this regard, the mammalian cell can be utilized to evaluate whether a test
agent is an agent affecting OBR. Such an evaluation may be provided by culturing the
mammalian cell and measuring the proliferation of the mammalian cell when exposed
to the test agent.
The mammalian cell of the present invention is typically cultured under normal
conditions in a basic culturing medium to which OB or a particular cytokine has been
added. Such a basic culturing medium can be a culturing medium in which fetal bovine
serum is added to RPMI1640 (GibcoBRL) to amount to 10% (v/v). The particular
cytokine which is added to the basic culturing medium is a cytokine needed to
proliferate the cytokine dependent host cell that is utilized to derive the mammalian
cells of the present invention. Such a particular cytokine may be interleukin-3 when
BaF-3 is utilized as the cytokine dependent mammalian host cell. The normal
conditions to culture the mammalian cell is typically provided by an incubator in which
the temperature is set to provide a temperature of 37°C with 5% (v/v) CO2 gas.
To expose the test agent to the mammalian cell, the test agent is typically added
to a basic culturing medium utilized for the mammalian cells. For example, a basic
culturing medium containing the test agent may be fractioned into a 96 well plate. The cultured mammalian cells can then be fractioned into each of the wells and be further
cultured under the normal culturing conditions. The test agent is usually added to the basic culturing medium so that the concentration of the test agent therein is InM to
ImM, and preferably 50nM to 50μM. The concentration of the cultured cells in the
basic culturing medium containing the test agent, varies with the proliferating
capabilities of said mammalian cell, but may generally be 5x10 cells/ml to 5x10
cells/ml, and preferably 1x10 cells/ml to lxlO5 cell/ml.
If so desired, the cultured mammalian cells can be washed with the basic
culturing medium before being exposed to the test agent. Typically, such cells are
washed a plurality of times, such as 2 to 5 times. Such a process of washing the cultured mammalian cells substantially avoids carrying over to the step of being exposed to the
test agent, the OB or the particular cytokine utilized to culture said mammalian cells.
The proliferation of the mammalian cells can be measured by utilizing
conventional methods which can measure whether the mammalian cell has proliferated
or conventional methods which can measure the amount of proliferation of a
mammalian cell. Examples of such conventional methods include an alamar blue
assay, MTT assay, various DNA quantification methods and the like. Further, to
measure the proliferation of the mammalian cell, the proliferation value achieved from
exposing the mammalian cell with the test agent is compared to a control culture. As
such a control culture, there is mentioned a first control culture in which the mammalian
cell of the present invention is unexposed to a test agent, a second control culture in
which the cytokine dependent mammalian host cell is exposed to the test agent and a
third control culture in which the cytokine dependent mammalian host cell is
unexposed to the test agent. The first control culture can be cultured in the basic
culturing medium to which the particular cytokine needed to proliferate the cytokine dependent mammalian host cell proliferate is added. The second and third controls can be cultured by using the basic culturing medium to which the particular cytokine needed to proliferate the cytokine dependent mammalian host cell is added.
The methods of the present invention can be utilized to screen a test agent as a
OB-like activating agent. The OB-like activating abilities of the test agent can be
measured when the mammalian cell is exposed to the test agent and to substantially no
cytokines activating the chimeric receptor. In such cases, there is selected as the OB-
like activating agent, a test agent which can provide a significantly higher proliferation
with the mammalian cell than that of the first control culture. Further, such a test agent usually provides no significant difference of proliferation between the second control
culture and the third control culture. Such a OB-like activating agent is a substance other than OB, which has a physiological function substantially identical to OB, such as
binding to OBR and transducing the triggered signal into the cell.
The test agent having the OB-like activating agent, which can be screened from
the present invention, can be, for example, N-(2-methoxyethyl) O-(5-methoxy-2-
nitrophenyl) methylsulfonate thiomide.
Additionally, the methods of the present invention can be utilized to screen a
test agent as an OB inhibiting agent. The OB inhibiting abilities of the test agent can be
measured when mammalian cell is exposed to the test agent and OB. When exposing
the mammalian cell with OB, the concentration of OB in the utilized culturing medium
is typically from InM to ImM, and preferably 50nM to 50 M. In such cases, there is
selected as the OB inhibiting agent, a test agent which can provide a significantly lower
proliferation with the mammalian cell than that of the first control culture. The second
control in screening for the OB inhibiting agent is usually cultured with a corresponding
amount of OB in its culturing medium. Further, such a test agent usually provides no significant difference of proliferation between the second control culture and the third
control culture.
A pharmaceutical composition comprising as an active ingredient, the OB-like
activating agent, which is selected from the screening methods above, can be used as an
anti-obesity pharmaceutical compositions. Further, a pharmaceutical composition
comprising as an active ingredient, the OB inhibiting agent, which is selected from the screening methods above, can be used as an anti-emaciation pharmaceutical
compositions. Further, such pharmaceutical compositions may comprise as the active
ingredient a pharmaceutically acceptable salt of the OB-like activating agent or OB
inhibiting agent.
A mammal, such as a human, can be applied orally or non-orally with an
effective amount of the pharmaceutical compositions. For example, when applying
orally, the pharmaceutical compositions can be used as a typical formulation such as
tablet, capsule, syrup, suspension and the like. Further, when applying non-orally, such
compositions can be used as a typical liquid formulation such as a solution, emulsion,
suspension and the like. As methods of non-orally applying the above formulations, for
example, there is mentioned injection methods, methods of directly applying as a
suppository to the intestine, and the like.
The formulations can be combined with an acceptable carrier, vehicle, adhesive,
stabilizer, diluent or the like to produce the pharmaceutical compositions. Further, there
may be added to the formulations when used as a injection formulation, a buffer,
dilution auxiliary, isotoning agent and the like, to produce the pharmaceutical
compositions.
Effective amounts of the applying the compositions may differ according to the
age, sex, weight, severity of symptoms and the like, but may be for an average adult mammal when orally applying the composition, in an amount of from lmg to 2g. When
injecting the compositions to an average adult mammal, the effective amount of the
provided composition may be O.lmg to 500mg. Further, such an effective amount may
be applied 1 to several times a day.
EXAMPLES
Example 1: Production of the mammalian cells of the present invention containing a
polynucleotide encoding the ECD of an OBR subunit and the ICD of a TPOR subunit
In the present Example 1, polynucleotides encoding the ECD of an OBR subunit
and the ICD of a TPOR subunit are introduced respectively to BaF-3 cells. The first of
the polynucleotides encodes the ECD of an OBR subunit as well as the TMD and ICD
of a TPOR subunit. The second of the polynucleotides encodes the ECD and TMD of
an OBR subunit and the ICD of a TPOR subunit. This can be accomplished according
to the following subsections (1.1) to (1.5).
(1.1) Preparation of polynucleotides encoding specified domains of an OBR subunit
and a TPOR subunit
A human brain cDNA library (CLONTECH) and a TF-1 cell cDNA library are
utilized to PCR amplify specified domains of an OBR subunit and a TPOR subunit. A
first set of the primers is designed, based on the nucleotide sequence of a OBR subunit
(GenBank Accession No. U43168). As such primers in the first set, Primers 1, 2 and 3
are synthesized with a DNA automatic synthesizer (PE Applied Biosystems) so that
Primer 1 has the nucleotide sequence shown in SEQ ID:1, Primer 2 has the nucleotide
sequence shown in SEQ ID:2 and Primer 3 has the nucleotide sequence shown in SEQ ID:3. A second set of primers is designed, based on the nucleotide sequence of a TPOR subunit (GenBank Accession No. M90102). As such primers in the second set, Primers
4, 5 and 6 are synthesized with a DNA automatic synthesizer (PE Applied Biosystems)
so that Primer 4 has the nucleotide sequence shown in SEQ ID: 4, Primer 5 has the
nucleotide sequence shown in SEQ ID:5 and Primer 6 has the nucleotide sequence
shown in SEQ ID: 6. Further, the first and second sets of the primers are synthesized to
additionally contain a particular restriction enzyme site, so that restriction enzyme sites
are added to the amplified product.
A PCR amplification is conducted with 50μl of a PCR mixture containing lOng
of the human brain cDNA library, lOOpmol of Primer 1, lOOpmol of Primer 2, 1.25U of Pyrobest DNA polymerase (Takara Shuzo), 4μl (2.5mM) of the dNTP mixture
provided with the Pyrobest DNA polymerase (Takara Shuzo) and 5μl of the buffer
provided with the Pyrobest DNA polymerase (Takara Shuzo). In the PCR
amplification, there is repeated 30 times, after an initial incubation of 94"C for 1
minute, an incubation cycle including incubations at 94°C for 30 seconds followed by
55T for 1 minute and then 72°C for 3 minutes. After repeating the incubation cycle,
the PCR amplification has an incubation at 72°C for 10 minutes. The resulting PCR
product thereof is subjected to agarose gel electrophoresis to obtain the amplified DNA
thereof (hereinafter referred to as DNA 1), which moves in the agarose gel as a size
corresponding to about 2.5kbp.
Another PCR amplification is similarly conducted under the conditions above
with a PCR mixture containing lOng of the human brain cDNA library, lOOpmol of
Primer 1, lOOpmol of Primer 3, 1.25U of the Pyrobest DNA polymerase, 4μl (2.5mM)
of the dNTP mixture provided with the Pyrobest DNA polymerase and 5μl of the buffer provided with the Pyrobest DNA polymerase. The resulting PCR product thereof is subjected to agarose gel electrophoresis to obtain that the amplified DNA (hereinafter
referred to as DNA 2), which moves in the agarose gel as a size corresponding to about
2.5kbp.
A cDNA library is prepared from TF-1 cells (ATCC No. CRL-2003 T.
Kitamura et al., 1989, J. Cell. Physiol. 140:323-334) according to a method described in
J. Sambrook, E.F. Frisch, T. Maniatis, Molecular Cloning 2nd Edition (Cold Springs
Harbor Laboratory press). A PCR amplification is similarly conducted under the
conditions above with a PCR mixture containing lOng of the cDNA library obtained
from the TF-1 cells, lOOpmol of Primer 4, lOOpmol of Primer 6, 1.25U of the Pyrobest
DNA polymerase, 4μl (2.5mM) of the dNTP mixture provided with the Pyrobest DNA
polymerase and 5μl of the buffer provided with the Pyrobest DNA polymerase. The
resulting PCR product thereof is subjected to agarose gel electrophoresis to obtain the
amplified DNA (hereinafter referred to as DNA 3), which moves in the agarose gel as a
size corresponding to about 440bp. Further, another PCR amplification is similarly conducted under the conditions
above with a PCR mixture containing lOng of the cDNA library obtained from the TF-1
cells, lOOpmol of Primer 5, lOOpmol of Primer 6, 1.25U of the Pyrobest DNA
polymerase, 4μl (2.5mM) of the dNTP mixture provided with the Pyrobest DNA
polymerase and 5μl of the buffer provided with the Pyrobest DNA polymerase. The
resulting PCR product thereof is subjected to agarose gel electrophoresis to obtain the amplified DNA (hereinafter referred to as DNA 4), which moves in the agarose gel as a
size corresponding to 370bp.
Each of the amplified DNAs 1, 2, 3 and 4 are phenol-chloroform extracted and
collected through ethanol precipitation. Each of the DNAs 1, 2, 3 and 4 are then dried and have added thereto 20μl of water to produce respectively aqueous solutions of the
DNAs 1, 2, 3 and 4.
(1.2) Preparation of vector pMX-neo
Based on the nucleotide sequence of pMAM-neo (GenBank Accession No.
U02432), Primer 7 having the sequence shown in SEQ ID:7 and Primer 8 having the
nucleotide sequence shown in SEQ ID:8 are synthesized with a DNA automatic
synthesizer (PE applied Biosystems). Further, Primers 7 and 8 are synthesized to additionally contain a particular restriction enzyme site.
A PCR amplification is then conducted with 50 l of a PCR solution containing
50pg of pMAM-neo (Clontech), lOOpmol of Primer 7, lOOpmol of Primer 8, 1.25U of
Pyrobest DNA Polymerase (Takara Shuzo), 4μl (2.5mM) of dNTP mixture provided
with the Pyrobest DNA Polymerase (Takara Shuzo) and 5μl of the buffer provided with
the Pyrobest DNA Polymerase (Takara Shuzo). In the PCR amplification, there is
repeated 30 times, after an initial incubation at 94°C for 1 minute, an incubation cycle
including incubations at 94°C for 30 seconds followed by 55°C for 1 minute and then
72°C for 3 minutes. After repeating the incubation cycle, the PCR amplification has an
incubation at 72°C for 10 minutes. The resulting PCR product thereof is subjected to
agarose gel electrophoresis to confirm that the amplified DNA (hereinafter referred to
as DNA 5) moves in the agarose gel as a size corresponding to about 1.5kbp. DNA 5 is
phenol-chloroform extracted and collected through ethanol precipitation. DNA 5 is
then dried and has added thereto 20μl of water.
Ten micro-liters (lOμl) of the resulting DNA 5 solution is restriction digested
for 2 hours at 37°C in a 50 fi L reaction buffer with 5U of Sal 1. One micro-gram (lμg) of vector pMX is constructed according to the method
disclosed in Exp. Hematol. 24:324-329 (1996). Vector pMX is then restriction digested
for 2 hours at 37°C in a lOμl reaction buffer with 5U Sal I.
The restriction digest products of DNA 5 and pMX are subjected to agarose gel
electrophoresis to separate, respectively, the DNA fragments of the restriction digest
products. The desired DNA fragments are obtained with Gene Clean Kit (BIO 101
Inc.). Ten micro-liters (lOμl) of water is added, respectively, to the fragment from DNA 5. Twenty micro-liters (20μl) of water is added to the fragment from pMX. For 1
hour, lμl of the solution containing the fragment from DNA 5 and lμl of the solution
containing the fragment from pMX are allowed to ligate together atl6°C in 30μl of a
ligation reaction mixture. A competent E. coli JM 109 strain is transformed with lOμl
of the resulting ligation reaction mixture. The E. coli JM 109 transformant is cultured
in 100ml of LB additionally containing 50μg/ml of ampicillin. Vector pMX-neo is then
prepared from the E. coli JM109 transformants with a QLAGEN Plasmid Maxi Prep.
(1.3) Construction of the chimeric receptor subunit expression vector
Ten micro-liters (lOμl) of each of the aqueous solutions of DNA 1, DNA 2,
DNA 3 and DNA 4 (prepared in (1.1)) are subjected, respectively, to a restriction digest
in 50μl of a reaction mixture for 2 hours at 37°C with the particular restriction enzymes
shown in Table 1. Further, lμg of vector pMX-neo is subjected to a restriction digest at
37°C for 2 hours in lOμl of a reaction mixture with 2U of each of BamH I and EcoR I.
The resulting products from the restriction digest are subjected to agarose gel
electrophoresis. The desired DNA fragments from DNA 1, DNA 2, DNA 3, DNA 4 and pMX-neo are then prepared from the agarose gel with Gene Clean Kit (BIO 101 Inc.). Ten micro-liters (lOμl) of water is added, respectively, to each of the prepared
DNA fragments to produce aqueous solutions of the prepared DNAs 1, 2, 3, and 4.
Twenty micro-liters (20μl) of water is added to the prepared pMX-neo fragment to
produce an aqueous solution of the prepared pMX-neo fragment.
One micro-liter (lμl) of the aqueous solution of the prepared pMX-neo
fragment, 2μl of the aqueous solution of the prepared DNA 1 fragment and 2μl of the
aqueous solution of the prepared DNA 3 fragment are allowed to ligate together in 50μl
of a ligation reaction mixture by utilizing Ligation Kit ver.l (Takara Shuzo) at 16°C for
1 hour. Ten micro-liters (lOμl) of the resulting reaction mixture is then utilized to transform a competent E. coli JM109 strain. The transformants are cultured in 100ml of
LB additionally containing 50μg/ml of ampicillin. The expression vector pMX-LTl
(Fig. 1) is then prepared from colonies of the transformant with QIAGEN Plasmid Maxi
Prep.
An expression vector pMX-LT2 (Fig. 2) is similarly prepared with lμl of the
aqueous solution of the prepared pMX-neo fragment and 2μl of the aqueous solution of
the prepared DNA 2 fragment and 2μl of the aqueous solution of the prepared DNA 4
fragment.
The expression vectors pMX-LTl and pMX-LT2 are analyzed with a DNA
automatic sequencer (PE Applied Biosystems) using a Thermo Sequenase II Die
terminator kit (Amersham Pharmacia Biotech). As a result, pMX-LTl is confirmed to
contain a polynucleotide encoding amino acid 1 to 841 (from the N-terminus) of a
human OBR subunit (GenBank Accession No. AAA93015) and amino acid 492 to 635
(from the N-terminus) of a human TPOR subunit (GenBank Accession No.
AAA69971). As such, it is confirmed that pMX-LTl contains a polynucleotide encoding the ECD of an OBR subunit as well as the TMD and ICD of a TPOR subunit. PMX-LT2 is confirmed to contain a polynucleotide encoding amino acid 1 to 863 (from
the N-terminus) of a human OBR subunit (GenBank Accession No. AAA93015) and
amino acid 514 to 635 (from the N-terminus) of a human TPOR subunit (GenBank
Accession No. AAA69971). As such, it is confirmed that pMX-LT2 contains a
polynucleotide encoding the ECD and TMD of an OBR subunit as well as ICD of a
TPOR subunit.
Table 1
Primers Restriction Enzymes
DNA 1 SEQ ID:1 and SEQ ID:2 BamH \ and Hind \\\
DNA 2 SEQ ID:1 and SEQ ID:3 BamH I and Xba I
DNA 3 SEQ ID:4 and SEQ ID:6 Hind \\\ and EcoR \
DNA 4 SEQ ID:6 and SEQ ID:6 Xba I and EcoR I
(1.4) The preparation of a viral medium
The expression vectors prepared in (1.3), pMX-LTl and pMX-LT2, are transfected into virus packaging BOSC23 cells by using lipofectamine (GibcoBRL).
For pMX-LTl, a mixture is produced by combining a solution containing 3μl of
pMX-LTl and 200μl of OPTI-MEM (GibcoBRL) with a solution containing 18μl of
lipofectamine and 200μl of the OPTI-MEM and is then incubated. Subsequently,
1600μl of the OPTI-MEM is added and mixed into the resulting mixture.
In a 60mm dish, 2xl06 cells of BOSC23 cells are placed therein and are cultured
overnight with 5% CO at 37°C in Dulbecco's modified Eagle's medium (GibcoBRL;
hereinafter referred to as D-MEM) additionally containing 10% (v/v) of fetal bovine serum (GibcoBRL; hereinafter referred to as FBS). After removing the supernatant D-
MEM additionally containing 10% (v/v) of FBS, the BOSC23 cells are washed with the
OPTI-MEM. The above mixture containing pMX-LTl, OPTI-MEM and lipofectamine
is added to the BOSC23 cells. The BOSC23 cells are then cultured for 5 hours in
similar culturing conditions as above. Subsequently, 2ml of D-MEM additionally
containing 20% (v/v) of FBS is added to the BOSC23 cells to further culture the
BOSC23 cells for 19 hours. The culturing medium is then replaced with 3ml of D-
MEM additionally containing 10% (v/v) of FBS. The BOSC23 cells are cultured for 24
hours under similar culturing conditions as above, to produce an innoculation medium. The innoculation medium is then centrifuged at 3000rpm for 5 minutes to obtain a first
viral medium. The expression vector pMX-LT2 is subjected under similar conditions
to obtain a second viral medium.
(1.5) Cloning of the mammalian cell of the present invention
BaF-3 cells are cultured in a basic culturing medium additionally containing
lng/ml of mouse IL-3 (R&D System) at 37°C with 5% CO2 gas. The cultured BaF-3
cells are centrifuged at lOOOrpm for 3 minutes to collect the cultured BaF-3 cells.
Polyprene (hexadimethrine bromide, Sigma) is added to the concentration of lOμg/ml
and mouse IL-3 is added to the concentration of lng/ml to the first and second viral
mediums prepared above in (1.4). The collected BaF-3 cells are suspended,
respectively, in 1ml of the resulting first and second viral mediums and are then
transferred to a 24 well plate. After 5 hours of culturing, 1 ml of a basic culturing medium additionally containing lng/ml of mouse IL-3 is added thereto. The cultured
BaF-3 cells are then further cultured for 1 hour to produce first infected BaF-3 cells (hereinafter referred to as cl cells) and second infected BaF-3 cells (hereinafter referred
to as c2 cells).
The cl cells and c2 cells are cultured, respectively, in a basic culturing medium
additionally containing lng/ml of mouse IL-3 and lmg/ml of GENETICIN
(GibcoBRL). The cl cells and c2 cells are suspended to 5 cells/ml with a basic culturing
medium additionally containing lOpM of OB, are transferred to a 96 well plate at
lOOμl/well and are cultured for 5 days at 37°C with 5% CO gas. The cl cells and c2
cells are then observed under a microscope to select the successfully cloned cl and c2 cells. The clones are then scaled up, respectively, to obtain 7 clones of the cl cells and
11 clones of the c2 cells.
Example 2: Production of the mammalian cells of the present invention containing a
polynucleotide encoding the ECD of an OBR subunit as well as the TMD and ICD of an
EPOR subunit
A human brain cDNA library (CLONTECH) and a TF-1 cell cDNA library is
utilized to PCR amplify specified domains of an OBR subunit and a TPOR subunit.
Two sets of primers are designed to amplify the desired nucleotide sequence in the
human brain cDNA library. A first set of the primers is designed, based on the
nucleotide sequence of an OBR subunit (GenBank Accession No. U43168). As such
primers, Primers 1 and 9 are synthesized with a DNA automatic synthesizer (PE
Applied Biosystems) so that Primer 1 has the nucleotide sequence shown in SEQ ID:1
and Primer 9 has the nucleotide sequence shown in SEQ ID:9. A second set of primers
is designed, based on the nucleotide sequence of an EPOR subunit (GenBank Accession No. M34986). As such primers, Primers 10 and 11 are synthesized with a DNA automatic synthesizer (PE Applied Biosystems) so that Primer 10 has the nucleotide sequence shown in SEQ ID: 10 and Primer 11 has the nucleotide sequence
shown in SEQ ID: 11. Further, the first and second sets of the primers are synthesized to
additionally contain a particular restriction enzyme site, so that restriction enzyme sites
are added to the amplified product.
A PCR amplification is conducted similarly to the conditions of (1.1) with 50μl
of a PCR mixture containing lOng of the human brain cDNA library, lOOpmol of
Primer 1, lOOpmol of Primer 9, 1.25U of Pyrobest DNA polymerase (Takara Shuzo),
4μl (2.5mM) of the dNTP mixture provided with the Pyrobest DNA polymerase
(Takara Shuzo) and 5μl of the buffer provided with the Pyrobest DNA polymerase (Takara Shuzo). The resulting PCR product thereof is subjected to agarose gel
electrophoresis to obtain the amplified DNA (hereinafter referred to as DNA 6), which
moves to a length corresponding to a length of about 2.5kbp.
Another PCR is conducted similarly to the conditions of (1.1) with 50μl of a
PCR mixture containing lOng of the TF-1 cell cDNA library, lOOpmol of Primer 10,
lOOpmol of Primer 11, 1.25U of Pyrobest DNA polymerase (Takara Shuzo), 4μl
(2.5mM) of the dNTP mixture provided with the Pyrobest DNA polymerase (Takara
Shuzo) and 5μl of the buffer provided with the Pyrobest DNA polymerase (Takara
Shuzo). The resulting PCR product thereof is subjected to agarose gel electrophoresis
and Gene Clean Kit (BIO 101 Inc.) to obtain the amplified DNA (hereinafter referred to
as DNA 7), which moves in the agarose gel to a size corresponding to about 780bp.
Forty-five micro-liters (45μl) of each of the PCR products for DNA 6 and
DNA7 are phenol-chloroform extracted and are collected by ethanol precipitation. Each
of the DNAs 6 and 7 are then dried and have 20μl of water added, to produce,
respectively aqueous solutions of the DNAs 6 and 7. Ten micro-liters (lOμl) of the aqueous solution of DNA 6 and lOμl of the aqueous solution of DNA 7 are restriction digested for 2 hours at 37°C in a 50μl reaction mixture with 5U of the particular
restriction enzyme shown in Table 2. Further, lμg of vector pMX-neo is subjected to a
restriction digest at 37°C for 2 hours in lOμl of a reaction mixture with 2U of each of
BamH I and EcoR I. The resulting products from the restriction digest are subjected to
agarose gel electrophoresis. The desired DNA fragments from DNA 6, DNA 7 and
pMX-neo are then prepared from the agarose gel with Gene Clean Kit (BIO 101 Inc.).
Ten microliters (lOμl) of water is added, respectively, to each of the prepared DNA 6
and DNA 7 fragments to produce, respectively, aqueous solutions of the prepared
DNAs 6 and 7. Twenty microliters (20μl) of water is added to the prepared pMX-neo fragment to produce an aqueous solution of the pMX-neo fragment.
Table 2
Primers Restriction Enzymes
DNA 6 SEQ ID:1 and SEQ ID:9 BamH \
DNA 7 SEQ ID:10 and SEQ ID:1 1 EcoR \
One micro-liter (lμl) of the aqueous solution of the prepared pMX-neo
fragment, 2μl of the aqueous solution of the prepared DNA 6 fragment, and 2μl of the
aqueous solution of the prepared DNA 7 fragment are allowed to ligate together in 50μl
of a ligation reaction mixture by utilizing Ligation Kit ver.l (Takara Shuzo) at 16°C for
1 hour. Ten micro-liters (lOμl) of the resulting reaction mixture is then utilized to
transform a competent E. coli JM109 strain. The transformants are cultured in 100ml of
LB additionally containing 50μg/ml of ampicillin. The expression vector pMX-LE is then prepared from colonies of the transformant with QIAGEN Plasmid Maxi Prep. The expression vector pMX-LE is analyzed with a DNA automatic sequencer
(PE Applied Biosystems) using a Thermo Sequenase II Die terminator kit (Amersham
Pharmacia Biotech). As a result, pMX-LE is confirmed to contain a polynucleotide
encoding amino acid 1 to 841 (from the N-terminus) of a human OBR subunit
(GenBank Accession No. AAA93015) and amino acid 251 to 508 (from the N-
terminus) of a human EPOR subunit (GenBank Accession No. AAA52401). As such, it
is confirmed that pMX-LE contains a polynucleotide encoding the ECD of an OBR
subunit as well as the TMD and ICD of an EPOR subunit.
A viral medium is produced similarly to the procedures of (1.4) with pMX-LE.
TF-1 cells are cultured in a basic culturing medium additionally containing
lng/ml of human granulocyte macrophage colony-stimulating factor receptor
(hereinafter referred to as human GM-CSF) with 5% C0 gas and at 37°C . The cultured
TF-1 cells are centrifuged at lOOOrpm for 3 minutes to collect 1 x 105 cells thereof.
Polyprene (hexadimethrine bromide, Sigma) is added to the concentration of lOμg/ml and human GM-CSF is added to the concentration of lng/ml to the viral medium. The
TF-1 cells are suspended in 1ml of the resulting viral medium and are then transferred
to a 24 well plate. After 5 hours of culturing, the TF-1 cells have added thereto a basic
culturing medium additionally containing lng/ml of human GM-CSF and are cultured overnight to obtain infected TF-1 cells (hereinafter referred to as c3 cells).
The c3 cells are cultured in a basic culturing medium additionally containing
lng/ml of human GM-CSF and lmg/ml of GENETICIN (GibcoBRL). The c3 cells are
suspended to 5 cells/ml in a basic culturing medium additionally containing lOpM of
OB, are transferred to a 96 well plate at lOOμl/well and are cultured for 5 days at 37°C
with 5% C02 gas. The c3 cells are then observed under a microscope to select the successfully cloned c3cells. The clones are then scaled up, respectively, to obtain a c3
cell clone.
Example 3: Proliferation dependency of the mammalian cells of the present invention
In a 100mm dish, the 7 clones of the cl cells and the 11 clones of the c2 cells,
prepared in (1.5), are cultured to 4xl05 to 8xl05 cells/ml at 37°C with 5% C02 gas in
10ml of a basic culturing medium additionally containing lng/ml of mouse IL-3 and
lmg/ml of GENEΗCIN. The cl cells and c2 cells are collected by centrifuging said cl
cells and c2 cells for 3 minutes at lOOOrpm. After the cl cells and c2 cells are washed 3
times with the basic culturing medium, the cl cells and the c2 cells are suspended in a
basic culturing medium.
The basic culturing medium and a basic culturing medium additionally
containing lOnM of OB are added, respectively, to the wells of a 96 well plate ([3603]
Costar) at lOμl/well. The suspensions of the cl cells are then added, respectively, at
90μl/well (5xl03 cells/well) to the basic culturing medium (final OB concentration of
OpM) and to the basic culturing medium additionally containing OB (final OB
concentration of InM). The suspensions of the c2 cells are also added, respectively, at
90μl/well (5xl03 cells/well) to the basic culturing medium (final OB concentration of OpM) and to the basic culturing medium additionally containing OB (final OB
concentration of InM). The cl cells and the c2 cells are cultured for 48 hours with 5%
C02 gas at 37°C. Ten micro-liters (lOμl) of Alamar blue (BIOSOURCE) are added to
each of the wells. After the cl cells and the c2 cells are then further cultured for 24
hours, the fluorescence thereof are measured with spectrofluoro (TECAN) under the
conditions in which the gain is set to 50, the excited wavelength is set to 595nm and the detection wavelength is set to 595nm. The experiment is repeated 40 or 80 times. When
the fluorescence value provided by the cultures containing no OB is subtracted from the
fluorescence value provided by the cultures containing OB, the resulting value for clone
8 of the cl cells (hereinafter referred to as the cl-8 cell) is 13,000 and the for clone 7 of
the c2 cells (hereinafter referred to as the c2-7 cell) is 11,000.
Subsequently, the cl-8 cells in a 100mm dish are cultured in lOμl of a basic
culturing medium additionally containing lng/nl of mouse IL-3 and lmg/ml of
GENETICIN with 5% C02 gas at 37°C. After the cl-8 cells are washed 3 times with
basic culturing medium, the cl-8 cells are suspended in basic culturing medium.
Culturing mediums are prepared by adding OB to basic culturing medium to
amount to, respectively, lOnM, InM, 500pM, lOOpM and 50pM. Subsequently, basic
culturing medium and the prepared mediums containing specified amounts of OB are
added, respectively to the wells of a 96 well plate at lOμl/well. The suspension of the
cl-8 cells is added at 90μl/well (5x10 cells/well) to the basic culturing medium (final
OB concentration of OpM) and to the basic culturing medium additionally containing
OB (final OB concentration of InM, 100pm, 50pM, lOpM and 5pM). The cl-8 cells
are then cultured for 48 hours at 37°C with 5% C0 gas. Ten microliters (lOμl) of
Alamar blue is added to each of the wells. After further culturing the cl-8 cells for 24
hours, the fluorescence thereof is measured similarly to the above.
Additionally, the procedures above of exposing OB is repeated with BaF-3
cells, instead of the cl-8 cells. The results of the BaF-3 cells and cl-8 cells are shown in
Table 4.The values in Table 4 illustrate the resulting value from correspondingly
subtracting fluorescence value provided by the cultures containing no OB from the fluorescence value provided by the cultures containing OB. Table 4 c1-8 cells BaF-3 cells
1 nM OB 13,000 30
100pM OB 12,900 -6
50pM OB 12,700 20
10pM OB 12,000 2
5pM OB 9,900 4 no OB 0 0
Example 4: Screening of a OB-like activating agent
(4.1) The screening method
In a 100mm dish, cl-8 cells are cultured with 5% C02 gas at 37°C in lOμl of a
basic culturing medium additionally containing lng/nl of mouse IL-3 and lmg/ml of
GENETICIN. After the cl-8 cells are washed 3 times with a basic culturing medium,
the cl-8 cells are suspended in a basic culturing medium.
Culturing mediums are prepared by adding a test agent to a basic culturing
mediums to amount to lOOμM or lOμM. As the test agent, there is utilized the
compound shown in the following general formula (I), N-(2-methoxyethyl) 0-(5- methoxy-2-nitrophenyl) methylsulfonatethioamide.
Figure imgf000040_0001
(I)
Subsequently, the basic culturing medium and the prepared mediums containing
specified amounts of the test agent are added, respectively, to the wells of a 96 well
plate at lOμl/well. The suspension of the cl-8 cells is added at 90μl/well (5xl03
cells/well) to the basic culturing medium (final test agent concentration of OpM) and to
the basic culturing mediums additionally containing the test agent (final test agent
concentration of lOμM and lμM). The cl-8 cells are then cultured for 48 hours at 37 C
with 5% C0 gas. Ten micro-liters (lOμl) of Alamar blue is added to each of the wells.
After further culturing the cl-8 cells for 24 hours, the fluorescence thereof is measured
similarly to the above. The resulting value from subtracting fluorescence value
provided by the cultures containing no test agent from the fluorescence value provided
by the cultures containing lμM of the test agent is 880.
Additionally, the procedures above of exposing the test agent is repeated with
BaF-3 cells, instead of the cl-8 cells. The resulting value from subtracting fluorescence
value provided by the cultures containing no test agent from the fluorescence value provided by the cultures containing lμM of the test agent is 35. (4.2) Production of N-(2-methoxyethyl) 0-(5-methoxy-2-nitrophenyl)
methylsulfonatethioamide
One and eighty-eight hundredths milliliters (1.88ml) of triethylamine and 7ml
of a toluene solution of 5-methoxy-2-nitrophenol (2.30g, 13.6 millimoles) is added at
drop increments to a toluene solution of methylphosphonothiophosphate dichloride (2.02g, 13.6 millimoles). The resulting solution is then allowed to react by being stirred
at room temperature for 12 hours. After lowering the temperature of the reaction
solution to 10°C or less, there is added thereto in drop increments, 2-
methoxyethylamine (2.04g, 27.2 millimoles) to produce a second reaction solution. The
second reaction solution is allowed to react by being stirred at room temperature for 12
hours. The second reaction solution is poured into water and is extracted with ethyl
acetate. The organic layer therefrom is washed with saturated salt water and dried with
anhydrous magnesium sulfate. The organic layer is then concentrated under pressurized
conditions and the resulting residue is subjected to silica gel chromatography to produce 2.37g of N-(2-methoxyethyl) 0-(5-methoxy-2-nitrophenyl)
methylsulfonatethioamide .
nD 25'8 1.5784
1H-NMR (300MHz, CDC13/TMS) : <5 (ppm)= 8.02 (d, J=9.2Hz, IH), 7.31 (d, J=3.1Hz,
IH), 6.75 (dd, J=9.2,3.1Hz, IH), 3.89 (s, 3H), 3.62-3.75 (m, IH), 3.30-3.40 (m, 2H),
3.19-3.28 (m, 2H), 3.26 (s, 3H), 2.07 (d, J=15.3Hz, 3H)
Example 5: Screening of an OB inhibiting agent In a 100mm dish, cl-8 cells are cultured with 5% C0 gas at 37°C in lOμl of a
basic culturing medium additionally containing lng/nl of mouse IL-3 and lmg/ml of
GENETICIN. After the cl-8 cells are washed 3 times with basic culturing medium, the
cl-8 cells are suspended in a basic culturing medium additionally containing 50pM of
OB.
Culturing mediums are prepared by adding to basic culturing mediums, OB to
amount to 50pM and a test agent to amount to lOOμM or lOμM. Subsequently, the basic culturing medium and the prepared mediums containing specified amounts of OB
and the test agent are added, respectively, to the wells of a 96 well plate at lOμl/well. The suspension of the cl-8 cells is also added at 90μl/well (5xl03 cells/well) to the
basic culturing medium (final test agent concentration of OpM) and to the basic
culturing mediums additionally containing OB and the test agent (final test agent
concentration of lOμM and lμM). The cl-8 cells are then cultured for 48 hours at 37°C
with 5% C0 gas. Ten microliters (lOμl) of Alamar blue is added to each of the wells.
After further culturing the cl-8 cells for 24 hours, the fluorescence thereof is measured
similarly to the above.
Additionally, the procedures above of exposing OB and the test agent are
repeated with BaF-3 cells, instead of the cl-8 cells.
An OB inhibiting agent can be then screened by selecting a test agent which
provides a low level of proliferation and no substantial differences between the BaF-3
cells cultured with the test agent and the BaF-3 cells cultured with no test agent. In
providing the low level of proliferation, the test agent provides a level of proliferation
which is lower than the cl-8 cells cultured with no test agent. All of the documents, patents, patent applications and publications mentioned above are incorporated herein by reference in their entirety.
Sequence Free Text
SEQ ID:1
An oligonucleotide primer designed for PCR amplification.
SEQ ID:2
An oligonucleotide primer designed for PCR amplification.
SEQ ID:3
An oligonucleotide primer designed for PCR amplification.
SEQ ID:4
An oligonucleotide primer designed for PCR amplification.
SEQ ID:5
An oligonucleotide primer designed for PCR amplification.
SEQ ID:6
An oligonucleotide primer designed for PCR amplification.
SEQ ID:7
An oligonucleotide primer designed for PCR amplification.
SEQ ID:8
An oligonucleotide primer designed for PCR amplification. SEQ ID:9
An oligonucleotide primer designed for PCR amplification.
SEQ ID: 10
An oligonucleotide primer designed for PCR amplification.
SEQ ID:11
An oligonucleotide primer designed for PCR amplification.
SEQ ID: 12
An oligonucleotide primer designed for PCR amplification.
SEQ ID: 13
An oligonucleotide primer designed for PCR amplification.

Claims

CLAIMS:
1. A method of evaluating whether a test agent is an agent affecting the activity of
OBR, said method comprising:
culturing a mammalian cell having:
an OB bindable chimeric receptor comprising an extracellular domain of an OBR
subunit, a transmembrane domain of a cytokine receptor subunit and an
intracellular domain of a receptor subunit selected from a non-OBR homodimeric
cytokine receptor subunit, gpl30 and IL-2R β subunit; and
a proliferation dependency on OB;
exposing the test agent to the mammalian cell; and
measuring the proliferation of the mammalian cell.
2. The method according to claim 1, wherein the intracellular domain is an
intracellular domain of a non-OBR homodimeric cytokine receptor subunit.
3. The method according to claim 1, wherein the intracellular domain is an
intracellular domain of a receptor subunit selected from gpl30 and IL-2R β subunit.
4. The method according to claim 1, wherein the intracellular domain is an
intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR
subunit, G-CSFR subunit, GHR subunit and PRLR subunit.
5. The method according to claim 1 , wherein the mammalian cell is cultured with a culturing medium containing substantially no cytokine activating the chimeric receptor.
6. The method according to claim 1, wherein the mammalian cell is cultured with a
culturing medium comprising OB.
7. A mammalian cell having:
a polynuceotide encoding a chimeric receptor subunit comprising an extracellular
domain of an OBR subunit, a transmembrane domain of a cytokine receptor subunit and
an intracellular domain of a receptor subunit selected from a non-OBR homodimeric
cytokine receptor subunit, gpl30 and IL-2R β subunit; and
a proliferation dependency on OB.
8. The mammalian cell according to claim 7, wherein the mammalian cell is
derived from a hematopoietic mammalian cell.
9. The mammalian cell according to claim 7, wherein the mammalian cell is
derived from a mammalian lymphocyte.
10. The mammalian cell according to claim 7, wherein the mammalian cell is
derived from a cell selected from BaF-3, TALL-101, OCl-Ly9, OCl-Lyl3.1, FDC-Pl,
TF-1, UT-7 and F-36-P.
11. The mammalian cell according to claim 10, wherein the mammalian cell is
derived from BaF-3.
12. The mammalian cell according to claim 7, wherein the intracellular domain is an intracellular domain of a non-OBR homodimeric cytokine receptor subunit.
13. The mammalian cell according to claim 7, wherein the intracellular domain is
an intracellular domain of a receptor subunit selected from gpl30 and IL-2R β subunit.
14. The mammalian cell according to claim 7, wherein the intracellular domain is
an intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR subunit, G-CSFR subunit, GHR subunit and PRLR subunit.
15. The mammalian cell according to claim 7, wherein said transmembrane domain
is a transmembrane domain of a receptor subunit selected from a homodimeric cytokine
receptor subunit, gpl30 and IL-2R β subunit.
16. The mammalian cell according to claim 7, wherein the transmembrane domain
is a transmembrane domain of an OBR subunit.
17. A mammalian cell having:
a polynucleotide encoding a chimeric receptor subunit comprising an extracellular
domain of an OBR subunit and intracellular and transmembrane domains of a receptor
subunit selected from a non-OBR homodimeric cytokine receptor subunit, gpl30 and
IL-2R j3 subunit; and
a proliferation dependency on OB.
18. The mammalian cell according to claim 17, wherein the intracellular and
transmembrane domains are intracellular and transmembrane domains of a receptor
subunit selected from a TPOR subunit, EPOR subunit, G-CSFR subunit, GHR subunit
and PRLR subunit.
19. A mammalian cell having:
an expression vector comprising a polynucleotide encoding a chimeric receptor subunit
comprising an extracellular domain of an OBR subunit, a transmembrane domain of a cytokine receptor subunit and an intracellular domain of a receptor subunit selected
from a non-OBR homodimeric cytokine receptor subunit, gpl30 and IL-2R β subunit;
and
a proliferation dependency on OB.
20. The mammalian cell according to claim 19, wherein the intracellular domain is
an intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR
subunit, G-CSFR subunit, GHR subunit and PRLR subunit.
21. A mammalian cell having:
a chimeric receptor anchored to a cell membrane thereof, wherein the chimeric receptor
comprises an extracellular domain of an OBR subunit, a transmembrane domain of a
cytokine receptor subunit and an intracellular domain of a receptor subunit selected
from a non-OBR homodimeric cytokine receptor subunit, gρl30 and IL-2R β subunit;
and
a proliferation dependency on OB.
22. A polynucleotide encoding a chimeric receptor subunit, wherein the chimeric
receptor subunit comprises an extracellular domain of an OBR subunit, a
transmembrane domain of a cytokine receptor subunit and an intracellular domain of a
receptor subunit selected from a TPOR subunit, EPOR subunit, GHR subunit, PRLR
subunit, gpl30 and IL-2R β subunit.
23. A chimeric receptor subunit comprising an extracellular domain of an OBR
subunit, a transmembrane domain of a cytokine receptor subunit and an intracellular domain of a receptor subunit selected from a TPOR subunit, EPOR subunit, GHR
subunit, PRLR subunit, gpl30 and IL-2R β subunit.
24. A screening method comprising:
conducting the method claim 1 and
selecting a test agent which proliferates the mammalian cell.
25. An OBR activating agent comprising the test agent selected from the screening
method of claim 24.
26. An anti-obesity pharmaceutical comprising as an active ingredient, the OBR
activating agent of claim 25.
27. A screening method comprising:
conducting the method of claim 1 and selecting a test agent which proliferates the mammalian cell.
28. An OBR inhibiting agent comprising the test agent selected from the screening
method of claim 27.
29. An anti-emaciation pharmaceutical comprising as an active ingredient, the OBR
inhibiting agent of claim 28.
PCT/JP2000/006814 1999-10-06 2000-09-29 Methods of evaluating whether a test agent is an agent affecting a leptin receptor WO2001025797A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU74515/00A AU7451500A (en) 1999-10-06 2000-09-29 Methods of evaluating whether a test agent is an agent affecting a leptin receptor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP11/285396 1999-10-06
JP28539699 1999-10-06

Publications (2)

Publication Number Publication Date
WO2001025797A2 true WO2001025797A2 (en) 2001-04-12
WO2001025797A3 WO2001025797A3 (en) 2002-05-10

Family

ID=17691001

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2000/006814 WO2001025797A2 (en) 1999-10-06 2000-09-29 Methods of evaluating whether a test agent is an agent affecting a leptin receptor

Country Status (2)

Country Link
AU (1) AU7451500A (en)
WO (1) WO2001025797A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7732133B2 (en) * 2000-07-17 2010-06-08 Chugai Seiyaku Kabushiki Kaisha Screening methods for biologically active ligands

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999004264A1 (en) * 1997-07-14 1999-01-28 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Method for identifying modifiers of the leptin: leptin receptor interaction
WO1999023493A1 (en) * 1997-10-31 1999-05-14 The Rockefeller University Methods of identifying agents that modulate leptin activity

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999004264A1 (en) * 1997-07-14 1999-01-28 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Method for identifying modifiers of the leptin: leptin receptor interaction
WO1999023493A1 (en) * 1997-10-31 1999-05-14 The Rockefeller University Methods of identifying agents that modulate leptin activity

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BENNET ET AL: "A role for leptin and its cognate receptor in hematopoiesis" CURRENT BIOLOGY, vol. 6, no. 9, - 17 July 1996 (1996-07-17) pages 1170-1180, XP000673008 *
HOUSEKNECHT, PORTOCARRERO: "Leptin and its receptors: regulators of whole-body energy homeostasis" DOMESTIC ANIMAL ENDOCRINOLOGY, vol. 15, no. 6, - November 1998 (1998-11) pages 457-475, XP001008003 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7732133B2 (en) * 2000-07-17 2010-06-08 Chugai Seiyaku Kabushiki Kaisha Screening methods for biologically active ligands

Also Published As

Publication number Publication date
WO2001025797A3 (en) 2002-05-10
AU7451500A (en) 2001-05-10

Similar Documents

Publication Publication Date Title
Raines et al. Identification and molecular cloning of a soluble human granulocyte-macrophage colony-stimulating factor receptor.
Miyazaki et al. The integrity of the conserved ‘WS motif’common to IL‐2 and other cytokine receptors is essential for ligand binding and signal transduction.
Arcasoy et al. Expression of erythropoietin receptor splice variants in human cancer
Matsui et al. Identification of two NF-κB sites in mouse CD95 ligand (Fas ligand) promoter: functional analysis in T cell hybridoma
Skoda et al. Murine c‐mpl: a member of the hematopoietic growth factor receptor superfamily that transduces a proliferative signal.
Greaves et al. CCR6, a CC chemokine receptor that interacts with macrophage inflammatory protein 3α and is highly expressed in human dendritic cells
Mignotte et al. Structure and transcription of the human c-mpl gene (MPL)
JP3737793B2 (en) IL-13 receptor polypeptide
US5521295A (en) Nucleic acids encoding hybrid receptor molecules
Nelson et al. A membrane-proximal region of the interleukin-2 receptor γc chain sufficient for Jak kinase activation and induction of proliferation in T cells
Jenkins et al. Activating point mutations in the common beta subunit of the human GM‐CSF, IL‐3 and IL‐5 receptors suggest the involvement of beta subunit dimerization and cell type‐specific molecules in signalling.
Elson et al. Cytokine-like factor-1, a novel soluble protein, shares homology with members of the cytokine type I receptor family
Seldin et al. Mutational analysis of a critical signaling domain of the human interleukin 4 receptor.
Nitz et al. Modular organization of Interleukin-6 and Interleukin-11 α-receptors
US20040091876A1 (en) Method for screening ligand having biological activity
Ihle et al. Signal transduction through the receptor for erythropoietin
Avalos et al. Point mutations in the conserved box 1 region inactivate the human granulocyte colony-stimulating factor receptor for growth signal transduction and tyrosine phosphorylation of p75c-rel
Kamogawa et al. Cutting edge: a conditionally active form of STAT6 can mimic certain effects of IL-4
US6007998A (en) Leptin assay
FI111016B (en) A method for screening for agents that modulate interleukin-5 receptor-dependent signaling pathways in cells
Gearing et al. Reconstitution of high affinity leukaemia inhibitory factor (LIF) receptors in haemopoietic cells transfected with the cloned human LIF receptor
Yamamura et al. Distinct downstream signaling mechanism between erythropoietin receptor and interleukin‐2 receptor.
Koga et al. Identification of differentially expressed molecules in adult T‐cell leukemia cells proliferating in vivo
WO2001025797A2 (en) Methods of evaluating whether a test agent is an agent affecting a leptin receptor
AU765010B2 (en) Eukaryotic cell-based gene interaction cloning

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase