WO2006007937A1 - Methods and assays for detecting guanylate cyclase activity - Google Patents

Methods and assays for detecting guanylate cyclase activity Download PDF

Info

Publication number
WO2006007937A1
WO2006007937A1 PCT/EP2005/006887 EP2005006887W WO2006007937A1 WO 2006007937 A1 WO2006007937 A1 WO 2006007937A1 EP 2005006887 W EP2005006887 W EP 2005006887W WO 2006007937 A1 WO2006007937 A1 WO 2006007937A1
Authority
WO
WIPO (PCT)
Prior art keywords
sgc
cell
cre
guanylate cyclase
vector encoding
Prior art date
Application number
PCT/EP2005/006887
Other languages
French (fr)
Inventor
Stefan Lohmer
Sabrina Corazza
Chiara Liberati
Original Assignee
Axxam S.R.L
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 Axxam S.R.L filed Critical Axxam S.R.L
Priority to CA002573357A priority Critical patent/CA2573357A1/en
Priority to JP2007520697A priority patent/JP2008506371A/en
Priority to KR1020077000872A priority patent/KR20070033428A/en
Priority to AU2005263396A priority patent/AU2005263396A1/en
Publication of WO2006007937A1 publication Critical patent/WO2006007937A1/en
Priority to IL180666A priority patent/IL180666A0/en
Priority to IS8590A priority patent/IS8590A/en
Priority to HR20070011A priority patent/HRP20070011A2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/527Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving lyase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/25Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving enzymes not classifiable in groups C12Q1/26 - C12Q1/66
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes

Definitions

  • the present invention relates to soluble guanylate cyclase (sGC) enzymes. More specifically it provides a cell-based assay system which allows to detect with high efficiency and sensitivity the soluble guanylate cyclase enzymatic activity.
  • the invention provides a method of screening molecules that interact with sGC by stimulating or inhibiting its enzymatic activity and that can be used in the diagnosis or therapy of sGC-mediated dysfunctions or diseases.
  • the invention further provides gene-constructs, vectors, and cells for use in said method.
  • Adenylate cyclases and guanylate cyclases synthesize the intracellular second messengers cAMP and cGMP, respectively, in response to a variety of regulatory signals.
  • Mammalian adenylate cyclases are intrinsic plasma membrane proteins that contain a duplicated module consisting of a hexahelical transmembrane region followed by a roughly 40-kDa cytosolic domain (Sunahara, et al. (1996) Annu. Rev. Pharm. Toxicol. 36, 461-480). Highly conserved and homologous sequences within the two cytosolic domains contribute to the active site of the enzyme (Tang, et al. (1995) Science 268, 1769-1772). The x-ray crystal structure of this soluble domain of the enzyme complexed with both activators and inhibitors has also been solved (Tesmer, et al. (1997) Science 278, 1907-1916).
  • Guanylate cyclases exist as both soluble and membrane-bound species. Both types of the enzyme contain cytoplasmic domains similar to those of the adenylate cyclases (Garbers, D.L., et al. (1994) MoL Biol. Cell 5, 1-5). Membrane-bound guanylate cyclases are monomers stimulated by the natriuretic peptides, whereas the soluble guanylate cyclase (sGC) exists as a heterodimer consisting of an ⁇ and a ⁇ subunit (both of which are required for catalysis) containing heme as prosthetic group.
  • sGC soluble guanylate cyclase
  • GC Guanylate cyclase belongs to a family of enzymes which catalyzes the conversion of guanosine 5'- triphosphate (GTP) to cyclic guanosine 3 ', 5'- monophosphate (cGMP).
  • GTP guanosine 5'- triphosphate
  • cGMP cyclic guanosine 3 ', 5'- monophosphate
  • sGC plays an important role in a variety of extracellular signals and different physiological processes, i.e. vasodilatation, platelet aggregation and adhesion and neural transmission.
  • the most important physiological activators of sGC are NO and NO-related compounds, which activate the enzyme upon binding to the heme moiety.
  • nitric oxide NO
  • heme group Binding of nitric oxide (NO) to a prosthetic heme group causes marked activation of soluble guanylate cyclases.
  • the pathogenesis of various cardiovascular diseases has been linked to an inappropriate activation of sGC.
  • Soluble guanylate cyclase is detectable in organs such as, for example, the heart, lung, liver, kidney, and brain of all mammals, including humans.
  • organs such as, for example, the heart, lung, liver, kidney, and brain of all mammals, including humans.
  • the oxidation state of the heme group iron in soluble guanylate cyclase may play an essential part.
  • a higher proportion of soluble guanylate cyclase with oxidized heme group iron would result in the possibility of diminishing activation of soluble guanylate cyclase by endogenous NO.
  • the mutated guanylate cyclase enzyme in particular, has ATP as substrate without having influenced the responsiveness to the respective GC stimulators.
  • This mutated form of soluble guanylate cyclase is useful for the detection of Nitric Oxide induced cAMP production.
  • Object of the present invention is to provide a cell-based assay useful for the determination of the enzymatic activity of soluble guanylate cyclase (sGC).
  • a further object of the invention is the provision of a method of screening molecules that interact with sGC, utilizing said cell-based assay.
  • the sGC enzymatic activity is determined by coupling a sGC enzyme with modified substrate specificity to a cAMP sensitive reporter construct in a suitable eukaryotic or prokaryotic cell, whereby the activity of the reporter gene is indicative of the sGC enzymatic activity.
  • the sGC enzyme with modified substrate specificity is able to catalyze the conversion of ATP into cyclic AMP without affecting the allosteric regulation of the natural enzyme.
  • the sGC substrate specificity can be modified by replacing the amino acid residues involved in nucleotide binding with the corresponding residues present in the active site of adenylate cyclase. For example, when rat sGC is used, the amino acids Arg592 of ⁇ subunit, Glu473 and Cys541 of ⁇ subunit are respectively changed into GIn, Lys and Asp, to confer ATP substrate-specificity. More details on the modification of sGC substrate specificity can be found in Sunahara et al. (1998), which is hereby incorporated by reference.
  • the cAMP sensitive reporter construct contains a reporter gene functionally linked to a cAMP-responsive promoter sensitive to intracellular cAMP levels.
  • the cAMP-responsive promoter is a MRE/CRE (Multiple Response Element - Ray A, et al. 1989; cAMP Responsive Element: Fink, J. S. et al. 1988) inducible promoter containing from 1 to 4 MREs and from 1 to 5 CRE elements.
  • the MRE-CRE inducible promoter sensitive to increased intracellular cAMP levels contains 3 MRE and 5 CRE element repetitions.
  • the MREs correspond to the human Interlukin-6 promoter region which enables the cell line to respond to Ga, Gq and Gs signalling (MRE: Ray A,
  • Each MRE element is 32 bp and corresponds to the sequence : ATGCT AAAGG ACGTC AC ATTGC AC AATCTTAA.
  • TGACGTCA The CRE sequence
  • CGTCA conserved sequence motif
  • the reporter construct may further contain transcription-regulating elements, selectable markers and viral promoters.
  • Suitable reporter genes that may be used according to the invention include, but are not limited to, luciferase, Green Fluorescent Proteins (GFP) and photoprotein genes. This system, however, is very useful not only for reporter gene transcription, but also for the expression and induction of any gene of interest such as the G-protein coupled receptor families (GPCRs), the ion channels and the nuclear hormone receptors.
  • GPCRs G-protein coupled receptor families
  • the invention in addition provides vectors and host cells containing the sGC-encoding gene and/or the reporter gene construct.
  • the host cell is a CHO cell line (hereafter, the CHO cell line containing the described MRE- CRE elements driving the expression of a luciferase reporter gene will be referred to as CHO cAMPL).
  • Host cell lines according to the invention are conveniently used as a tool for detection of intracellular NO-release.
  • the invention provides a method for determining the sGC enzymatic activity in a cultured eukaryotic or prokaryotic cell, which comprises:
  • the invention is directed to a method of identifying molecules that modulate sGC activity, which comprises:
  • Molecules that stimulate or inhibit sGC activity can be selected by measuring the reporter gene expression/activity.
  • the assay can be carried out in the presence or absence of known NO-releasing substances which activate the enzyme upon NO-binding to its heme moiety.
  • NO-releasing molecules and NO-independent sGC activators and inhibitors which are not specific for the catalytic site, e.g. ODQ
  • ODQ inhibitors which are not specific for the catalytic site
  • the selected compounds represent valuable candidate for the treatment of diseases related to sGC dysfunctions.
  • sGC-stimulating compounds can be used in the therapy of diseases involving inappropriate activation of sGC, especially diseases of the cardiovascular system.
  • the method of the invention is more advantageous in that a) it does not require radioactive reagents, b) it has a higher sensitivity, c) it can be easily adapted to a high throughput format and d) it is performed in a native environment.
  • the invention provides a method of modulating the expression of a gene of interest in cultured eukaryotic or prokaryotic cell, which comprises:
  • Rat wild type sGC ⁇ 3 and ⁇ l subunits were amplified from brain cDNA by RT-PCR and used as templates for site directed mutagenesis.
  • the amino acids Arg 592 of ⁇ subunit, GIu 473 and Cys 541 of ⁇ subunit were respectively changed into GIn, Lys and Asp, which confer specificity for adenosine.
  • Responsive Element Fink, J. S. et al. 1988) inducible promoter at 5' of the luciferase gene of the pMAMneoluc vector (Clontech).
  • MRE element 3
  • Both vectors were transfected in a stable way in CHO-Kl cells and positive clones were selected on the basis of the best Luciferase induction obtained upon 4 hrs. incubation with the NO donor compound SNAP (Fig. 1).
  • the most responsive clone coming from 3 rounds of limiting dilutions was used to test the activity of different compounds such as NO-donors and NO-independent activators, in the presence or absence of the known sGC inhibitor ODQ (Fig. 2-4).
  • Luciferase activity was detected after injection of 50 ⁇ l Bright-GloTM (Promega) at the CCD camera (60" measurement, high sensitivity). Published EC 50 1,7x10 "6 M.
  • Figure 6 Kinetic and dose response experiments with SNAP 5000 c/w were seeded in 96 wp. 48 h after seeding the cells were incubated with increasing concentration of SNAP: after times of incubation indicated the compound was replaced with medium serum free. Total incubation time 4 h.
  • Rat soluble Guanylate Cyclase ( ⁇ 3 and ⁇ l subunits): alpha 3 subunit NM O 17090; coding region: 690 aa long beta 1 subunit NM O 12769; coding region: 619 aa long Rat brain cDNA was purchased from Clontech (cat. N° 637312). 2 ⁇ l of cDNA was used as template in PCR analysis for non quantitative expression analysis. In addition a negative control was performed with no template. Standard PCR procedure were as indicated by Perkin Elmer. PCR protocol was as follows: Primers:
  • Primer sGCalpha UP (A) : 5 ' CGGJCTA
  • Amplification products were analysed by electrophoresis on 1% agarose gel in IxTAE running buffer following standard procedure, as described by Maniatis et al.
  • Mutations into the ⁇ subunit of sGC were inserted by PCR using the wild type gene as template, in particular primers sGCbeta UP and sGCbetaE473K LOW were used to amplify the 5' portion of the gene, primers sGCbetaE473K UP and sGCbetaC541D LOW were used to amplify a central fragment, primers sGCbetaC541D UP and sGCbeta LOW were used to amplify the 3' portion of the gene. To reconstitute the full length mutated gene, these three discrete fragments were used as template in a final PCR reaction performed with primers sGCbeta UP sGCbeta LOW.
  • Amplification protocol performed in Perkin Elmer 9700 thermocycler 1 time the following step: pre PCR 2' at 94°C 20 times the following steps: denaturation 30" at 94°C annealing 1 ' at 52 0 C elongation 30" at 68°C
  • the mutation R592Q into the ⁇ subunit of sGC was inserted by site directed mutagenesis (QuikChange XL Site-Directed Mutagenesis Kit from Stratagene), using primers sGCalphaR592Q UP and sGCalphaR592Q LOW.
  • Amplification protocol performed in Perkin Elmer 9700 thermocycler 1 time the following step: pre PCR 1 ' at 95°C 18 times the following steps: denaturation 30" at 95 0 C annealing 1 ' at 55 0 C elongation 13' at 68 0 C 1 time the following step: elongation 7' at 68°C PCR reaction mix:
  • pIRES bi-cistronic mammalian expression vector
  • the mutated ⁇ subunit of sGC described above was cloned into EcoRI unique restriction site of pIRES.
  • the pIRESsGC ⁇ vector was subsequently digested with Smal to insert the mutated ⁇ subunit of sGC downstream the IRES element. All the constructs obtained were verified by full-length dideoxy sequencing.
  • G418 (Calbiochem cod.345812).
  • Preculture conditions Cells were seeded for experiments when 70-80% confluent.
  • CHO cAMPL previously generated by Axxam, were transfected with pIRESsGC ⁇ mut.
  • the stable transfected cells were put in 1st limiting dilution (1st LD) at 1 c/w in 96 wp format.
  • the four best responding clones were put in 3rd LD (0.3 c/w, 96 wp format). The final clone was chosen after the 3rd LD selected with SNAP 10 ⁇ M.
  • SNAP (Tocris cat# 0598) was dissolved in DMSO at a concentration of 100 mM and stored in aliquots at -20 0 C. Working solution was freshly prepared in Serum Free Medium.
  • SIN (Tocris cat# 0 756) was dissolved in water at a concentration of 100 mM and stored in aliquots at -20 0 C.
  • NOC18 (Calbiochem cat# 487957) was dissolved in water at a concentration of 100 mM and stored in aliquots at -20 0 C.
  • ODQ (Calbiochem cat# 495320) was dissolved in DMSO at a concentration of 100 mM and stored in aliquots at -20 0 C.
  • B A Y41-2272 was purchased from Alexis, dissolved in DMSO at a concentration of 10 mM and stored in aliquots at -20 0 C.
  • Tyrode Buffer composition NaCl 130 mM, KCl 5 mM, CaCl 2 2 mM, MgCl 2 1 mM, NaHCO 3 5 mM e HEPES 20 mM pH 7.4.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

The present invention relates to soluble guanylate cyclase (sGC) enzymes. In particular it provides a cell-based assay system which allows to detect with high efficiency and sensitivity the soluble guanylate cyclase enzymatic activity. In a preferred embodiment, the invention provides a method of screening molecules that interact with sGC by stimulating or inhibiting its enzymatic activity and that can be used in the diagnosis or therapy of sGC-mediated dysfunctions or diseases. The invention further provides gene-constructs, vectors, and cells for use in said method.

Description

METHODS AND ASSAYS FOR DETECTING GUANYLATE CYCLASE ACTIVITY
The present invention relates to soluble guanylate cyclase (sGC) enzymes. More specifically it provides a cell-based assay system which allows to detect with high efficiency and sensitivity the soluble guanylate cyclase enzymatic activity. In a preferred embodiment, the invention provides a method of screening molecules that interact with sGC by stimulating or inhibiting its enzymatic activity and that can be used in the diagnosis or therapy of sGC-mediated dysfunctions or diseases. The invention further provides gene-constructs, vectors, and cells for use in said method. BACKGROUND OF THE INVENTION Adenylate cyclases and guanylate cyclases synthesize the intracellular second messengers cAMP and cGMP, respectively, in response to a variety of regulatory signals. Mammalian adenylate cyclases are intrinsic plasma membrane proteins that contain a duplicated module consisting of a hexahelical transmembrane region followed by a roughly 40-kDa cytosolic domain (Sunahara, et al. (1996) Annu. Rev. Pharm. Toxicol. 36, 461-480). Highly conserved and homologous sequences within the two cytosolic domains contribute to the active site of the enzyme (Tang, et al. (1995) Science 268, 1769-1772). The x-ray crystal structure of this soluble domain of the enzyme complexed with both activators and inhibitors has also been solved (Tesmer, et al. (1997) Science 278, 1907-1916).
Guanylate cyclases exist as both soluble and membrane-bound species. Both types of the enzyme contain cytoplasmic domains similar to those of the adenylate cyclases (Garbers, D.L., et al. (1994) MoL Biol. Cell 5, 1-5). Membrane-bound guanylate cyclases are monomers stimulated by the natriuretic peptides, whereas the soluble guanylate cyclase (sGC) exists as a heterodimer consisting of an α and a β subunit (both of which are required for catalysis) containing heme as prosthetic group.
Guanylate cyclase (GC) belongs to a family of enzymes which catalyzes the conversion of guanosine 5'- triphosphate (GTP) to cyclic guanosine 3 ', 5'- monophosphate (cGMP).
By formation of cGMP as second messenger, sGC plays an important role in a variety of extracellular signals and different physiological processes, i.e. vasodilatation, platelet aggregation and adhesion and neural transmission.
The most important physiological activators of sGC are NO and NO-related compounds, which activate the enzyme upon binding to the heme moiety.
Binding of nitric oxide (NO) to a prosthetic heme group causes marked activation of soluble guanylate cyclases. The pathogenesis of various cardiovascular diseases has been linked to an inappropriate activation of sGC.
The concept of NO dependent sGC activation as a mechanism of action has been widely validated by the clinical use of NO donors.
Soluble guanylate cyclase is detectable in organs such as, for example, the heart, lung, liver, kidney, and brain of all mammals, including humans. In pathological processes or in processes relevant for pathological events, the oxidation state of the heme group iron in soluble guanylate cyclase may play an essential part. A higher proportion of soluble guanylate cyclase with oxidized heme group iron would result in the possibility of diminishing activation of soluble guanylate cyclase by endogenous NO. This might lead, inter alia, to an increase in blood pressure, activation of platelets, increased proliferation of cells or enhanced adhesion of cells, to permanent high blood pressure, stable or unstable angina pectoris, thromboses, myocardial infarct, strokes, pulmonary edemas, erectile dysfunction, uncontrolled tissue growth with tumor formation, diabetes, renal dysfunctions, hepatic dysfunctions, or vascular dysfunctions. A mutated GC enzyme in which the three residues of the active site of guanylate cyclase are substituted with those of adenylate cyclase has been shown to have a completely changed nucleotide specificity (Sunahara, R.K. et al 1998). The mutated guanylate cyclase enzyme, in particular, has ATP as substrate without having influenced the responsiveness to the respective GC stimulators. This mutated form of soluble guanylate cyclase is useful for the detection of Nitric Oxide induced cAMP production.
Traditional methods for the detection of soluble guanylate cyclase modulators are: - radioactive methods in which nucleotide precursors are used cellular systems based on calcium channels sensitive to cyclic nucleotides in-vivo methods in which the effect of stimulators of GC are tested on tissues or in animal models. DISCLOSURE OF THE INVENTION
Object of the present invention is to provide a cell-based assay useful for the determination of the enzymatic activity of soluble guanylate cyclase (sGC). A further object of the invention is the provision of a method of screening molecules that interact with sGC, utilizing said cell-based assay. According to the invention, the sGC enzymatic activity is determined by coupling a sGC enzyme with modified substrate specificity to a cAMP sensitive reporter construct in a suitable eukaryotic or prokaryotic cell, whereby the activity of the reporter gene is indicative of the sGC enzymatic activity. The sGC enzyme with modified substrate specificity is able to catalyze the conversion of ATP into cyclic AMP without affecting the allosteric regulation of the natural enzyme. The sGC substrate specificity can be modified by replacing the amino acid residues involved in nucleotide binding with the corresponding residues present in the active site of adenylate cyclase. For example, when rat sGC is used, the amino acids Arg592 of α subunit, Glu473 and Cys541 of β subunit are respectively changed into GIn, Lys and Asp, to confer ATP substrate-specificity. More details on the modification of sGC substrate specificity can be found in Sunahara et al. (1998), which is hereby incorporated by reference.
The cAMP sensitive reporter construct contains a reporter gene functionally linked to a cAMP-responsive promoter sensitive to intracellular cAMP levels. In a preferred embodiment, the cAMP-responsive promoter is a MRE/CRE (Multiple Response Element - Ray A, et al. 1989; cAMP Responsive Element: Fink, J. S. et al. 1988) inducible promoter containing from 1 to 4 MREs and from 1 to 5 CRE elements. Preferably, the MRE-CRE inducible promoter sensitive to increased intracellular cAMP levels contains 3 MRE and 5 CRE element repetitions.
The MREs correspond to the human Interlukin-6 promoter region which enables the cell line to respond to Ga, Gq and Gs signalling (MRE: Ray A,
Sassone-Corsi P, Sehgal PB., 1989; Fitzgerald, LR, Manan, IJ, Dytko, GM,
Wu, HL, Nambi,P, 1999). Each MRE element is 32 bp and corresponds to the sequence : ATGCT AAAGG ACGTC AC ATTGC AC AATCTTAA.
The CRE sequence ("TGACGTCA") contains one or multiple copies of the conserved sequence motif CGTCA (Fink, J. S. et al. 1988). This element is similar to sequences in other genes known to be regulated by cAMP and to sequences in several viral enhancers.
The reporter construct may further contain transcription-regulating elements, selectable markers and viral promoters. Suitable reporter genes that may be used according to the invention include, but are not limited to, luciferase, Green Fluorescent Proteins (GFP) and photoprotein genes. This system, however, is very useful not only for reporter gene transcription, but also for the expression and induction of any gene of interest such as the G-protein coupled receptor families (GPCRs), the ion channels and the nuclear hormone receptors.
The invention in addition provides vectors and host cells containing the sGC-encoding gene and/or the reporter gene construct. Preferably, the host cell is a CHO cell line (hereafter, the CHO cell line containing the described MRE- CRE elements driving the expression of a luciferase reporter gene will be referred to as CHO cAMPL). Host cell lines according to the invention are conveniently used as a tool for detection of intracellular NO-release.
In a preferred embodiment, the invention provides a method for determining the sGC enzymatic activity in a cultured eukaryotic or prokaryotic cell, which comprises:
A) introducing into the cell: i) a vector encoding a reporter gene functionally linked to a CRE or MRE-CRE inducible promoter, and, simultaneously or separately, ii) a vector encoding a sGC with substrate specificity for ATP, i.e able to catalyze the conversion of ATP into cyclic AMP;
B) incubating the cell with a compound that stimulates or inhibits sGC enzymatic activity; C) determining the activity of the reporter gene.
In a preferred embodiment, the invention is directed to a method of identifying molecules that modulate sGC activity, which comprises:
A) introducing into the cell: i) a vector encoding a reporter gene functionally linked to a CRE or MRE-CRE inducible promoter, and, simultaneously or separately, ii) a vector encoding sGC with substrate specifcity for ATP,
B) incubating the cell with a candidate molecule; C) determining the activity of the reporter gene.
Molecules that stimulate or inhibit sGC activity can be selected by measuring the reporter gene expression/activity. The assay can be carried out in the presence or absence of known NO-releasing substances which activate the enzyme upon NO-binding to its heme moiety. Both NO-releasing molecules and NO-independent sGC activators (and inhibitors which are not specific for the catalytic site, e.g. ODQ) can be conveniently assayed with the method of the invention. The selected compounds represent valuable candidate for the treatment of diseases related to sGC dysfunctions. In particular, sGC-stimulating compounds can be used in the therapy of diseases involving inappropriate activation of sGC, especially diseases of the cardiovascular system.
Compared to known assays used so far for determining the sGC activity, the method of the invention is more advantageous in that a) it does not require radioactive reagents, b) it has a higher sensitivity, c) it can be easily adapted to a high throughput format and d) it is performed in a native environment.
In a further embodiment, the invention provides a method of modulating the expression of a gene of interest in cultured eukaryotic or prokaryotic cell, which comprises:
A) introducing into the cell: i) a vector encoding the target gene functionally linked to a
CRE or MRE-CRE inducible promoter; and, simultaneously or separately, ii) a vector encoding sGC with substrate specificity for ATP,
B) incubating the cell with a molecule able to stimulate sGC in a NO-dependent or NO-independent manner.
A typical assay according to the invention, the conditions and reagents therein utilized, are disclosed below in greater detail. EXPERIMENTAL SECTION
Rat wild type sGC α3 and β l subunits were amplified from brain cDNA by RT-PCR and used as templates for site directed mutagenesis. The amino acids Arg 592 of α subunit, GIu 473 and Cys 541 of β subunit were respectively changed into GIn, Lys and Asp, which confer specificity for adenosine.
An expression vector for the rat mutagenized sGC cDNA was prepared
(pIRES from Clontech). A reporter vector was constructed by cloning the MRE-CRE (Multiple Response Element: Ray A, et al. 1989; cAMP
Responsive Element: Fink, J. S. et al. 1988) inducible promoter at 5' of the luciferase gene of the pMAMneoluc vector (Clontech).
The structure of the MRE-CRE inducible element sensitive to increased intracellular cAMP levels, is the following: MRE element: 3
CRE element: 5
Figure imgf000008_0001
Both vectors were transfected in a stable way in CHO-Kl cells and positive clones were selected on the basis of the best Luciferase induction obtained upon 4 hrs. incubation with the NO donor compound SNAP (Fig. 1). The most responsive clone coming from 3 rounds of limiting dilutions was used to test the activity of different compounds such as NO-donors and NO-independent activators, in the presence or absence of the known sGC inhibitor ODQ (Fig. 2-4).
The results are illustrated in the attached Figures, where: Figure 1: 3rd LD clone re-test
12 best clones coming from 3rd LD were incubated in 50 μl serum free medium with SNAP for 4h: luciferase activity was measured after injection of 50 μl Bright-GloTM (Promega) at the CCD camera (60" measurement, high sensitivity).
Experimental conditions: 5000 c/w in 96 wp format, 48 h after seeding. Clone 7 was chosen as final clone for further characterization. Figure 2: (a) EC50 calculation for the NO-donor SNAP 5000 c?w were seeded in 96 wp. 48 h after seeding the cells were incubated 4 h with 50 μl of SNAP in serum free medium (SFM). (b) The experiment was performed in parallel also on not transfected cells.
Luciferase activity was detected after injection of 50 μl Bright-GloTM (Promega) at the CCD camera (60" measurement, high sensitivity). Published EC50 1,7x10"6M.
Figure 3; (a) EC50 calculation for the NO-donor SIN-I 1500 c/w were seeded in 384 wp. 48 h after seeding the cells were incubated 4 h with SIN-I in Tyrode buffer (25 μl). Luciferase activity was detected after injection of 25 μl of Bright-GloTM (Promega) at the CCD camera (60" measurement, high sensitivity). Published EC50 10"7-10"6M. (b) Inhibition of SIN-I activity by ODQ
1500 c/w were seeded in 384 wp; 48 h after seeding a dose response of SIN in the absence or presence of ODQ (300 nM, 3 μM, 30 μM) was performed. Total volume of incubation 25 μl in Tyrode buffer. Figure 4: EC50 calculation for the NO-donor NOC- 18.
5000 c/w were seeded in 96 wp. 48 h after seeding the cells were incubated 4 h with 50 μl of NOC 18 in serum free medium (SFM). Luciferase activity was detected after injection of 50 μl Bright-GloTM (Promega) at the CCD camera (60" measurement, high sensitivity). Figure 5: (a) EC50 calculation for the sGC stimulator BAY41-2272
1500 c/w were seeded in 384 wp. 48 h after seeding the cells were incubated 4 h with BAY41-2272 in Tyrode buffer 25 μl. Luciferase activity was detected after injection of 25ul Bright-GloTM (Promega) at the CCD camera (60" measurement, high sensitivity). Published EC50 6x10'7M. (b) Inhibition of B AY41-2272 activity by ODQ
1500 c/w were seeded in 384 wp; 48h after seeding a dose response of BAY41-2272 in the absence or presence of ODQ (300 nM, 3 μM, 30 μM) was performed. Total volume of incubation 25 μl in Tyrode buffer. BAY41-2272 stimulating effect on sGC is competed by ODQ.
Figure 6: Kinetic and dose response experiments with SNAP 5000 c/w were seeded in 96 wp. 48 h after seeding the cells were incubated with increasing concentration of SNAP: after times of incubation indicated the compound was replaced with medium serum free. Total incubation time 4 h.
MATERIALS AND METHODS PCR analysis
Rat soluble Guanylate Cyclase (α3 and β l subunits): alpha 3 subunit NM O 17090; coding region: 690 aa long beta 1 subunit NM O 12769; coding region: 619 aa long Rat brain cDNA was purchased from Clontech (cat. N° 637312). 2 μl of cDNA was used as template in PCR analysis for non quantitative expression analysis. In addition a negative control was performed with no template. Standard PCR procedure were as indicated by Perkin Elmer. PCR protocol was as follows: Primers:
Primer sGCalpha UP (A) : 5 ' CGGJCTA|ATA]AGGJAGG]AAAJCCAJC3 ' Primer sGCalpha LOW (B): 5'AGASAAC|\TGJ:GG|CTCKCT)\AT(:TA3 ' Primer sGCbeta UP (C): 5 'CGG|\CAJCCA|TGT)\CG|GTT)ΓTGSΓGA3 '
Primer sGCbeta LOW (D) 5 'GGc|rGCJ3Aτfrc4GTTfrTC|ATc|cTG3 ' PCR reaction mix: 2 μl template 5 μl 10 x Pfx Buffer (GIBCO-LifeTechnologies)
1.5 μl 1O mM dNTPs
1 μl 50 mM MgSO4 (GIBCO-LifeTechnologies)
2.5 μl primer A (IO μM) 2.5 μl primer B (10 μM)
2.5 U Platinum Pfx (GIBCO-LifeTechnologies)
35 μl H2O
Amplification protocol performed in Perkin Elmer 9700 thermocycler: 1 time the following step: pre PCR 2' at 94°C
40 times the following steps: denaturation 30" at 94°C annealing l ' at 56°C elongation 2' 10" at 680C 1 time the following step: elongation 7' at 68°C
Expected length of specific PCR product: 1309 bp (690 bp for A/B amplification; 619 bp for C/D).
Amplification products were analysed by electrophoresis on 1% agarose gel in IxTAE running buffer following standard procedure, as described by Maniatis et al.
Site Directed Mutagenesis
Mutations into the β subunit of sGC were inserted by PCR using the wild type gene as template, in particular primers sGCbeta UP and sGCbetaE473K LOW were used to amplify the 5' portion of the gene, primers sGCbetaE473K UP and sGCbetaC541D LOW were used to amplify a central fragment, primers sGCbetaC541D UP and sGCbeta LOW were used to amplify the 3' portion of the gene. To reconstitute the full length mutated gene, these three discrete fragments were used as template in a final PCR reaction performed with primers sGCbeta UP sGCbeta LOW.
Amplification protocol performed in Perkin Elmer 9700 thermocycler: 1 time the following step: pre PCR 2' at 94°C 20 times the following steps: denaturation 30" at 94°C annealing 1 ' at 520C elongation 30" at 68°C
1 time the following step: elongation 7' at 680C
The mutation R592Q into the α subunit of sGC was inserted by site directed mutagenesis (QuikChange XL Site-Directed Mutagenesis Kit from Stratagene), using primers sGCalphaR592Q UP and sGCalphaR592Q LOW.
Amplification protocol performed in Perkin Elmer 9700 thermocycler: 1 time the following step: pre PCR 1 ' at 95°C 18 times the following steps: denaturation 30" at 950C annealing 1 ' at 550C elongation 13' at 680C 1 time the following step: elongation 7' at 68°C PCR reaction mix:
1 μl template
5 μl 10 x Turbo Pfu Buffer (Stratagene)
1 μl 2OmM dNTPs 2.5 μl primer A (10 μM)
2.5 μl primer B (10 μM)
1 μl 2.5 U/μl Turbo Pfu (Stratagene)
37 μl H2O
Figure imgf000013_0001
Cloninε procedure: pIRESsGCβαmut
Each of the two mutated subunits of sGC was cloned into a bi-cistronic mammalian expression vector (pIRES, purchased from Clontech) that allows the simultaneous translation of two consecutive ORF (Open Reading Frame) from the same messenger RNA, through an Internal Ribosome Entry Site
(IRES) element, thus generating the so-called pIRESsGCβα vector.
The mutated β subunit of sGC described above was cloned into EcoRI unique restriction site of pIRES. The pIRESsGCβ vector was subsequently digested with Smal to insert the mutated α subunit of sGC downstream the IRES element. All the constructs obtained were verified by full-length dideoxy sequencing.
Cell culture:
Culture medium, seeding and incubation: DMEM/F 12 with Glutamax (GIBCO cod.31331-028), 10% FBS, 1% Pen./Strep. (Invitrogen cod.15140-
122), 25 mM Hepes Buffer Solution (GIBCO cod.15630-056), 1.0 mM
Sodium Pyruvate (GIBCO cod.11360-039), 1.5 g/L Sodium Bicarbonate
(GIBCO cod. 25080-060), 0.5 mg/ml G418 (Calbiochem cod.345812).
Preculture conditions: Cells were seeded for experiments when 70-80% confluent.
Cell culture conditions: Split twice a week: 3.OxIO5 cells/ffask T75 (recovery: 8x106 cells).
Clone Selection Process:
CHO cAMPL, previously generated by Axxam, were transfected with pIRESsGCβαmut.
After eight days of G418 selection, the stable transfected cells were put in 1st limiting dilution (1st LD) at 1 c/w in 96 wp format.
After two weeks, plates were incubated 4h with SNAP 100 μM and measured at CCD camera after injection of Bright-GloTM (Promega). The 9 best responding clones were re-tested in a dose response experiment as counted cells.
The 3 best responding clones were put in 2nd LD (0.3 c/w, 96wp).
After two weeks, plates were replicated and after 48 h incubated 4 h with SNAP 100 μM and read at CCD camera after injection of Bright-GloTM (Promega). The best responding 12 clones were re-tested in a dose response experiment as counted cells.
The four best responding clones were put in 3rd LD (0.3 c/w, 96 wp format). The final clone was chosen after the 3rd LD selected with SNAP 10 μM.
Complete optimization of the assay was performed on the best responding clone 7. Reference Compounds:
SNAP (Tocris cat# 0598) was dissolved in DMSO at a concentration of 100 mM and stored in aliquots at -200C. Working solution was freshly prepared in Serum Free Medium.
SIN (Tocris cat# 0 756) was dissolved in water at a concentration of 100 mM and stored in aliquots at -200C.
NOC18 (Calbiochem cat# 487957) was dissolved in water at a concentration of 100 mM and stored in aliquots at -200C.
ODQ (Calbiochem cat# 495320) was dissolved in DMSO at a concentration of 100 mM and stored in aliquots at -200C. B A Y41-2272 was purchased from Alexis, dissolved in DMSO at a concentration of 10 mM and stored in aliquots at -200C.
Working solutions for SIN- I and BAY41-2272 and B AY58-2667 were freshly prepared in standard Tyrode buffer.
Tyrode Buffer composition: NaCl 130 mM, KCl 5 mM, CaCl2 2 mM, MgCl2 1 mM, NaHCO3 5 mM e HEPES 20 mM pH 7.4.
Bright-Glo™: Promega cat#E2620.
REFERENCES
Garbers, D.L., Koesling, D., and Schultz, G. (1994) MoI. Biol. Cell 5, 1-5.
Dessauer, C. W., Scully, T.T., and Gilman, A.G. (1997) J. Biol. Chem. 272, 22272-22277.
Fink JS, Verhave M, Kasper S, Tsukada T, Mandel G, Goodman RH., Proc
Natl Acad Sci U S A. 1988 Sep;85(18):6662-6.
Fitzgerald, LR, Manan, IJ, Dytko, GM, Wu, HL, Nambi,P, (1999) Analytical
Biochemistry, 275, 54-61. Liu, Y., Ruoho, A.E., Rao, V.D., and Hurley, J.H. (1997) Proc. Natl. Acad.
ScL U.S.A. 94, 13414-13419.
Ray A, Sassone-Corsi P, Sehgal PB. MoI Cell Biol. 1989 Dec; 9(12):5537-47.
Schmidt P.M., Schramm M., Schroeder H, Wunder F. and Stasch J.P., J Biol.
Chem., 2004, vol. 279, p. 3025-3032. Stasch J.P., Nature, 2001, vol. 410, p. 212-215.
Sunahara, R.K., Dessauer, C. W., and Gilman, A.G. (1996) Annu. Rev. Pharm.
Toxicol. 36, 461-480.
Sunahara, R.K., Dessauer, C. W., Whisnant, R.E., Kleuss, C, and Gilman,
A.G. (1997) J. Biol. Chem. 272, 22265-22271. Sunahara, R.K., Beuve, A., Tesmer, JJ. G., Sprang, S.R., Garbers, D.L., and
Gilman, A.G., J Biol. Chem., 1998, vol. 237, p. 16332-16338.
Tang, W.-J., and Gilman, A.G. (1995) Science 268, 1769-1772.
Tesmer, J.J.G., Sunahara, R.K., Gilman, A.G., and Sprang, S.R. (1997)
Science 278, 1907-1916.

Claims

1. A method for determining the soluble guanylate cyclase (sGC) enzymatic activity in a cultured eukaryotic or prokaryotic cell, which comprises:
A) introducing into the cell: i) a vector encoding a reporter gene functionally linked to a CRE or MRE-CRE inducible promoter, and, simultaneously or separately, ii) a vector encoding a soluble guanylate cyclase able to catalyze the conversion of ATP into cyclic AMP;
B) contacting the cell with a compound that stimulates or inhibits sGC enzymatic activity;
C) determining the activity of the reporter gene.
2. A method of identifying molecules that modulate sGC activity in a cultured eukaryotic or prokaryotic cell, which comprises:
A) introducing into the cell: i) a vector encoding a reporter gene functionally linked to a CRE or MRE-CRE inducible promoter, and, simultaneously or separately, ii) a vector encoding a soluble guanylate cyclase able to catalyze the conversion of ATP into cyclic AMP;
B) contacting the cell with a candidate molecule;
C) determining the activity of the reporter gene.
3. A method of modulating the expression of a target gene in a cultured eukaryotic or prokaryotic cell, which comprises: A) introducing into the cell: i) a vector encoding the target gene functionally linked to a CRE or MRE-CRE inducible promoter, and, simultaneously or separately, ii) a vector encoding a soluble guanylate cyclase able to catalyze the conversion of ATP into cyclic AMP; B) contacting the cell with a molecule able to stimulate sGC enzymatic activity in a NO-dependent or NO-independent manner;
4. A method according to claims 1-3, wherein the MRE-CRE inducible promoter contains 3 MRE and 5 CRE elements.
5. A method according to claims 1 -3, wherein a vector encoding rat soluble guanylate cyclase is used.
6. A method according to claim 5, wherein said rat sGC carries Arg592— >Gln mutation on the α subunit and Glu473-→Lys and Cys541→Asp mutations on the β subunit.
7. A method according to claims 1-2, wherein the reporter gene codes for a protein selected from luciferase, Green Fluorescent Protein (GFP) and a
Photoprotein.
8. A method according to claim 2, wherein the candidate molecule is able to stimulate or inhibit sGC activity in a NO-dependent or NO-independent modality.
9. A method according to claim 3, wherein the target gene is selected from ion channels, GPCRs and Nuclear Hormone Receptors.
10. A prokaryotic or eukaryotic host cell containing i) a vector encoding a reporter gene functionally linked to a CRE or MRE-CRE inducible promoter and ii) a vector encoding a soluble guanylate cyclase able to catalyze the conversion of ATP into cyclic AMP.
11. A eukaryotic host cell according to claim 8, which is a CHO cell.
12. The use of a soluble guanylate cyclase able to catalyze the conversion of ATP into cyclic AMP for the screening of compounds that modulate sGC enzymatic activity.
13. The use of a host cell according to claims 10- 11 as a tool for detection of intracellular NO-release.
PCT/EP2005/006887 2004-07-16 2005-06-27 Methods and assays for detecting guanylate cyclase activity WO2006007937A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CA002573357A CA2573357A1 (en) 2004-07-16 2005-06-27 Methods and assays for detecting guanylate cyclase activity
JP2007520697A JP2008506371A (en) 2004-07-16 2005-06-27 Methods and assays for detecting guanylate cyclase activity
KR1020077000872A KR20070033428A (en) 2004-07-16 2005-06-27 Methods and assays for detecting guanylate cyclase activity
AU2005263396A AU2005263396A1 (en) 2004-07-16 2005-06-27 Methods and assays for detecting guanylate cyclase activity
IL180666A IL180666A0 (en) 2004-07-16 2007-01-11 Methods and assays for detecting guanylate cyclase activity
IS8590A IS8590A (en) 2004-07-16 2007-01-12 Methods and tests to detect guanylate cyclase activity
HR20070011A HRP20070011A2 (en) 2004-07-16 2007-01-12 Methods and assays for detecting guanylate cyclase activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP04016774A EP1616965A1 (en) 2004-07-16 2004-07-16 Methods and assays for detecting guanylate cyclase activity
EP04016774.4 2004-07-16

Publications (1)

Publication Number Publication Date
WO2006007937A1 true WO2006007937A1 (en) 2006-01-26

Family

ID=34925781

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/006887 WO2006007937A1 (en) 2004-07-16 2005-06-27 Methods and assays for detecting guanylate cyclase activity

Country Status (11)

Country Link
EP (1) EP1616965A1 (en)
JP (1) JP2008506371A (en)
KR (1) KR20070033428A (en)
CN (1) CN1985003A (en)
AU (1) AU2005263396A1 (en)
CA (1) CA2573357A1 (en)
HR (1) HRP20070011A2 (en)
IL (1) IL180666A0 (en)
IS (1) IS8590A (en)
RU (1) RU2007101290A (en)
WO (1) WO2006007937A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102006038942A1 (en) * 2006-08-18 2008-02-21 Bayer Healthcare Ag Method of identifying nitric oxide modulators
AU2011242527B2 (en) * 2010-04-23 2016-05-19 University Of Florida Research Foundation, Inc. rAAV-guanylate cyclase compositions and methods for treating Leber's congenital amaurosis-1 (LCA1)
SG185777A1 (en) * 2010-05-27 2012-12-28 Merck Sharp & Dohme Soluble guanylate cyclase activators

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5783402A (en) * 1991-10-01 1998-07-21 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method of identifying ligands and anatgonists of G-protein coupled receptor
EP0863214A2 (en) * 1997-02-25 1998-09-09 Smithkline Beecham Corporation Reporter gene construct for measuring G protein coupled receptor activation
US6221617B1 (en) * 1996-12-06 2001-04-24 Julie Heinrich Bioassay for growth hormone releasing hormone

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5783402A (en) * 1991-10-01 1998-07-21 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method of identifying ligands and anatgonists of G-protein coupled receptor
US6221617B1 (en) * 1996-12-06 2001-04-24 Julie Heinrich Bioassay for growth hormone releasing hormone
EP0863214A2 (en) * 1997-02-25 1998-09-09 Smithkline Beecham Corporation Reporter gene construct for measuring G protein coupled receptor activation

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BEUVE A: "Conversion of a Guanylyl Cyclase to an Adenylyl Cyclase", METHODS : A COMPANION TO METHODS IN ENZYMOLOGY, ACADEMIC PRESS INC., NEW YORK, NY, US, vol. 19, no. 4, December 1999 (1999-12-01), pages 545 - 550, XP004466856, ISSN: 1046-2023 *
CHEN W ET AL: "A COLORIMETRIC ASSAY FOR MEASURING ACTIVATION OF GS-AND GQ-COUPLED SIGNALING PATHWAYS", ANALYTICAL BIOCHEMISTRY, ACADEMIC PRESS, SAN DIEGO, CA, US, vol. 226, 1995, pages 349 - 354, XP002051981, ISSN: 0003-2697 *
FITZGERALD LAURA RYDELEK ET AL: "Measurement of responses from Gi-, Gs-, or Gq-coupled receptors by a multiple response element/cAMP response element-directed reporter assay", ANALYTICAL BIOCHEMISTRY, ACADEMIC PRESS, NEW YORK, NY, US, vol. 275, no. 1, 1 November 1999 (1999-11-01), pages 54 - 61, XP002320214, ISSN: 0003-2697 *
SUNAHARA ROGER K ET AL: "Exchange of substrate and inhibitor specificities between adenylyl and guanylyl cyclases", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 273, no. 26, 26 June 1998 (1998-06-26), pages 16332 - 16338, XP002325858, ISSN: 0021-9258 *

Also Published As

Publication number Publication date
AU2005263396A1 (en) 2006-01-26
HRP20070011A2 (en) 2007-04-30
RU2007101290A (en) 2008-07-20
EP1616965A1 (en) 2006-01-18
CA2573357A1 (en) 2006-01-26
JP2008506371A (en) 2008-03-06
IL180666A0 (en) 2007-06-03
IS8590A (en) 2007-01-12
KR20070033428A (en) 2007-03-26
CN1985003A (en) 2007-06-20

Similar Documents

Publication Publication Date Title
Beadling et al. Interleukin‐2 activation of STAT5 requires the convergent action of tyrosine kinases and a serine/threonine kinase pathway distinct from the Raf1/ERK2 MAP kinase pathway.
JPH09500023A (en) Identification of ligands by selective amplification of cells transfected with the receptor
Cox et al. Regulation and functional characterization of a rat recombinant dopamine D3 receptor
Verzi et al. N-twist, an evolutionarily conserved bHLH protein expressed in the developing CNS, functions as a transcriptional inhibitor
AU736899B2 (en) Co-expression of Gia protein and Gi protein coupled receptor to enhance signal transduction responses
WO2014142984A1 (en) Coincidence reporter gene system
WO2006007937A1 (en) Methods and assays for detecting guanylate cyclase activity
RU2139936C1 (en) Method of determining modulation effect of substance on interleukin-5 receptor-linked signal-transmission route in human or animal cell
CA2506619A1 (en) Cell lines and host nucleic acid sequences related to infectious disease
Fujie et al. Nitric oxide synthase and soluble guanylyl cyclase underlying the modulation of electrical oscillations in a central olfactory organ
JP2003534765A (en) Isolated VSHK-1 receptor polypeptide and methods of using the same
Kanasaki et al. cAMP responsive element-mediated regulation of the gene transcription of the alpha 1B adrenergic receptor by thyrotropin.
JP3643288B2 (en) G protein-coupled receptor ligand screening method and G protein-coupled receptor expression cloning method
JPH119282A (en) Melatonin receptor expression cell and its use
US20020164754A1 (en) Compositions and methods for treatment of Toxoplasma gondii and other apicomplexans
JP2005118050A (en) Screening method of g protein conjugating receptor ligand and expression cloning method of g protein conjugating receptor
MacDonald Exploring the Pharmacophore of Novel Synthetic Peptide Activators of type I-alpha cGMP-Dependent Protein Kinase
JP4400183B2 (en) Mutant estrogen receptor α and its gene
JP2002045188A (en) Dioxin receptor gene and its use
US20020119440A1 (en) Peripheral nerve type sodium channels
JP2000354494A (en) Dioxin receptor gene and its utilization
JP2004254601A (en) Variant androgen receptor and cell for evaluating its activity
JP2004008140A (en) Variant androgen receptor and cell for evaluating its activity
JP2004008141A (en) Variant androgen receptor and cell for evaluating its activity
JP2001078782A (en) Dioxin receptor gene and its use

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

WWE Wipo information: entry into national phase

Ref document number: 2573357

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 180666

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: P20070011A

Country of ref document: HR

Ref document number: 2007101290

Country of ref document: RU

Ref document number: 200700360

Country of ref document: ZA

Ref document number: 200580023476.6

Country of ref document: CN

Ref document number: 1020077000872

Country of ref document: KR

Ref document number: 2007520697

Country of ref document: JP

Ref document number: 2005263396

Country of ref document: AU

Ref document number: 162/KOLNP/2007

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2005263396

Country of ref document: AU

Date of ref document: 20050627

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005263396

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1020077000872

Country of ref document: KR

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