Screening methods for compounds that modulate the activity of G-protein coupled receptors

Krohn; Michael ;   et al.

Patent Application Summary

U.S. patent application number 11/810402 was filed with the patent office on 2009-01-01 for screening methods for compounds that modulate the activity of g-protein coupled receptors. Invention is credited to Michael Krohn, Holger Zinke.

Application Number20090004651 11/810402
Document ID /
Family ID37243512
Filed Date2009-01-01

United States Patent Application 20090004651
Kind Code A1
Krohn; Michael ;   et al. January 1, 2009

Screening methods for compounds that modulate the activity of G-protein coupled receptors

Abstract

The present invention relates to a screening system for modulators of GPCRs. Further it relates to recombinant vector systems for the heterologous expression of heterodimeric g-protein coupled receptors (GPCRs) in eucaryotic host cells. Preferably the functional expression of engineered GPCRs for the perception of sweet and L-amino acid taste or more preferably the use of said receptors for the identification of functional ligands is also encompassed.


Inventors: Krohn; Michael; (Lorsch, DE) ; Zinke; Holger; (Zwingenberg, DE)
Correspondence Address:
    PROPAT, L.L.C.
    425-C SOUTH SHARON AMITY ROAD
    CHARLOTTE
    NC
    28211-2841
    US
Family ID: 37243512
Appl. No.: 11/810402
Filed: June 5, 2007

Current U.S. Class: 435/6.12 ; 435/320.1; 435/354; 435/358; 435/366; 435/6.13; 530/350; 536/23.5
Current CPC Class: G01N 2333/705 20130101; G01N 2500/10 20130101; C12N 15/79 20130101; C12Q 1/025 20130101; G01N 33/566 20130101
Class at Publication: 435/6 ; 536/23.5; 530/350; 435/320.1; 435/366; 435/358; 435/354
International Class: C12Q 1/68 20060101 C12Q001/68; C07H 21/00 20060101 C07H021/00; C07K 14/435 20060101 C07K014/435; C12N 15/63 20060101 C12N015/63; C12N 5/10 20060101 C12N005/10

Foreign Application Data

Date Code Application Number
Jun 7, 2006 EP 06011710.8

Claims



1.) A method for the identification of modulators of GPCRs, comprising the steps of: a. transforming a eucaryotic host cell with genetic sequences coding for one or more than one GPCR(s), b. cultivation of the transformed host cells under conditions sufficient for the functional expression of said one or more GPCRs, c. contacting the cultivated host cell expressing the one or more GPCRs in a functional manner with a potential modulator of the selected one or more GPCRs, d. measuring an selected cellular response of the transformed host cell on exposure to the potential modulator, and e. selecting an identificate, which induces a specific response.

2.) A method according to claim 1, wherein the host cell is selected from the group consisting of: HEK293 (human embryo kidney), Hela (Human Negroid cervix epitheloid carcinoma), HT29 (Human Caucasian colon adenocarcinoma grade II), A431 (human squamous carcinoma), IMR 32 (human caucasian neuroblastoma), K562 (Human Caucasian chronic myelogenous leukaemia), U937 (Human Caucasian histiocytic lymphoma), MDA-MB-231 (Human Caucasian breast adenocarcinoma), SK-N-BE(2) (Human Caucasian neuroblastoma), SH-SY5Y (Human neuroblastoma), HL60 (human promyelocytic leukemia) or eukaryotic non-human cell lines like CHO-K1 (Hamster Chinese ovary), COS-7 (Monkey African green kidney, SV40 transformed), S49 (mouse lymphoma), Ltk (Mouse C34/An connective tissue), NG108-15 (Mouse neuroblastoma.times.Rat glioma hybrid), B50 (Rat nervous tissue neuronal, ECACC), C6 (Rat glial tumour), Jurkat (Human leukaemic T cell lymphoblast), BHK (Hamster Syrian kidney), Neuro-2a (Mouse Albino neuroblastoma), NIH/3T3 (mouse embryo fibroblast).

3.) A method according to claim 1, wherein the one or more GPCR(s) is a T1R-type GPCR.

4.) A method according to claim 1, wherein two or more GPCRs are expressed in a heterologous co-expression of at least two different T1R-type GPCRs.

5.) A method according to claim 1, wherein the transformation is accomplished with a multicistronic expression vector.

6.) A method according to claim 5, wherein the multicistronic expression vector comprises a multicistronic expression unit comprising downstream from a promoter for the expression in an eucaryotic host and functionally linked thereto, the following cistrons: a. GPCR.sub.1 b. GPCR.sub.2 and a c. selection marker, wherein the promoter is a strong promoter suitable for use in the selected host cell, wherein the GPCR, and the GPCR.sub.2 are independently from another being selected from the group consisting of T1R or T2R taste receptors, wherein the selection marker is being selected from the group consisting of hygromycin.sup.r, zeocin.sup.r, neomycin.sup.r, blasticidin.sup.r or puromycin.sup.r, wherein both the GPCR1 and the GPCR2 as well as the selection marker are functionally connected by intervening IRES selected from the group consisting of IRES.sub.EMCV, derived from encephalomyocarditis virus (synonym: CITE.sub.EMCV); IRES.sub.GTX, derived from the GTX homeodomain mRNA; IRES.sub.Rbm3, derived from cold-inducible Rbm3; IRES.sub.PV, derived of polioviral origin, IRES.sub.RV, derived from rhinovirus, IRESFMDV, derived from food and mouth disease virus; IRE.sub.HV, derived from hepatitis C virus, IRES.sub.CSFV, derived from classic swine fever virus, IRES.sub.BVDV, derived from bovine viral diarrhea virus; IRES.sub.FMLV, derived from friend murine leukemia virus gag mRNA; IRES.sub.MMLV, derived from moloney murine leukemia virus gag mRNA; IRES.sub.HIV, derived from human immunodefiency virus env mRNA; IRES.sub.PSIV, derived from Plautia stali intestine virus; IRES.sub.RPV, derived from Rhopalosiphum padi virus; IRES.sub.KSH, derived from Karposi's sarcoma-associated herpesvirus, preferably the IRES.sub.EMCV being derived from encephalomyocarditis virus (synonym: CITE.sub.EMCV), and wherein the multicistronic expression unit is terminated by a polyadenylation signal.

7.) A method according to claim 5, wherein the multicistronic cloning vector additionally comprises a cistron coding for a G-protein.

8.) A method according to claim 1, wherein the modulator is selected from the group consisting of small molecules and/or peptides.

9.) A method according to claim 1, wherein the cellular response is measured by measuring the change of intracellular calcium levels relative to intracellular calcium levels without contacting the cell to the modulator.

10.) A method according to claim 9, wherein the change is an increase.

11.) A method according to claim 9, wherein the change is a decrease relative to intracellular calcium levels when the cell is contacted to a sweet compound instead of the modulator.

12.) Multiparameter optimized nucleic acid molecule coding for a GPCR.

13.) Protein consisting of the amino acids sequences according to SEQ ID NO: 1 and/or SEQ ID NO: 2 or functionally equivalent proteins.

14.) Nucleic acid molecules according to claim 12, as depicted in one of SEQ ID NO 3 or SEQ ID NO: 4.

15.) Nucleic acid molecules functionally equivalent to the molecules according to claim 14.

16.) Multicistronic expression vector comprising more than one cistron coding for a GPCR.

17.) Multicistronic expression vector comprising downstream from a promoter for the expression in an eucaryotic host and functionally linked thereto, the following cistrons: d. GPCR.sub.1 e. GPCR.sub.2 and a f. selection marker, wherein the promoter preferably is a strong promoter suitable for use in the selected host cell, wherein the GPCR.sub.1 and the GPCR.sub.2 are independently from another selected from the group consisting of T1R or T2R taste receptors, wherein the selection marker is selected from the group consisting of hygromycin.sup.r, zeocin.sup.r, neomycin.sup.r, blasticidin.sup.r puromycin.sup.r, wherein both the GPCR1 and the GPCR2 as well as the selection marker are functionally connected by intervening IRES selected from the group consisting of IRES.sub.EMCV, derived from encephalomyocarditis virus (synonym: CITE.sub.EMCV); IRES.sub.GTX, derived from the GTX homeodomain mRNA; IRES.sub.Rbm3, derived from cold-inducible Rbm3; IRES.sub.PV, derived of polioviral origin, IRES.sub.RV, derived from rhinovirus, IRESFMDV, derived from food and mouth disease virus; IRE.sub.HV, derived from hepatitis C virus, IRES.sub.CSFV, derived from classic swine fever virus, IRES.sub.BVDV, derived from bovine viral diarrhea virus; IRES.sub.FMLV, derived from friend murine leukemia virus gag mRNA; IRES.sub.MMLV, derived from moloney murine leukemia virus gag mRNA; IRES.sub.HIV, derived from human immunodefiency virus env mRNA; IRES.sub.PSIV, derived from Plautia stali intestine virus; IRES.sub.RPV, derived from Rhopalosiphum padi virus; IRES.sub.KSH, derived from Karposi's sarcoma-associated herpesvirus, preferably the IRES.sub.EMCV being derived from encephalomyocarditis virus (synonym: CITE.sub.EMCV), and wherein the multicistronic expression unit is terminated by a polyadenylation signal.

18.) A vector according to claim 17, wherein the multicistronic cloning vector additionally comprises a cistron coding for a g-protein or an equivalent thereof.

19.) Cell lines stably transfected with a nucleic acid molecule according to claim 14.

20.) A method of identifying a GPCR modulator comprising a method according to claim 1.

21.) A method according to claim 1, wherein the host cell is HEK293 (human embryo kidney), Hela (Human Negroid cervix epitheloid carcinoma), CHO-K1 (Hamster Chinese ovary) or Neuro-2a (Mouse Albino neuroblastoma).

22.) A method according to claim 1, wherein the one or more GPCR(s) is T1R1, T1R2 and/or T1R3.

23.) A method according to claim 1, wherein two or more GPCRs are expressed in a heterologous co-expression of T1R1/T1R3.

24.) A method according to claim 1, wherein two or more GPCRs are expressed in a heterologous co-expression of T1R2/T1R3.

25.) A method according to claim 1, wherein the transformation is accomplished with a tricistronic or a tetracistronic cloning vector.

26.) A method according to claim 5, wherein the promoter is selected from the group consisting of: cytomegalovirus promoter (P-CMV), human elongation factor 1 alpha promoter (P-E1.alpha.), human ubiquitin promoter (P-ubi), simian virus promoter (P-SV40), and Rous sarcoma virus long terminal repeat promoter (P--RSV-LTR).

27.) A method according to claim 5, wherein the GPCR.sub.1 and the GPCR.sub.2 are independently from another selected from the group of T1R receptors.

28.) A method according to claim 5, wherein the GPCR.sub.1 and the GPCR.sub.2 are independently from another selected from a combination of T1R1-T1R3 or T1R2-T1R3.

29.) A method according to claim 5 wherein the multicistronic cloning vector additionally comprises a cistron coding for G-alpha 15.

30.) A method according to claim 7, wherein the g-protein is located between the last GPCR and the selection marker and is functionally connected to both via an IRES.

31.) A method according to claim 11, wherein the sweet compound is selected from the group consisting of glucose, fructose, saccharose, acesulfam K, saccharin, cyclamat, aspartam, xylitol, stevioside, sucralose, thaumatin, monellin, brazzein, perillartine, glycyrrhizin, sucronic acid, P-4000, SC45647, NC174, neohesperidin and sweet tasting amino acids.

32.) Multiparameter optimized nucleic acid molecule according to claim 12, wherein the GPCR, is a T1R-type GPCR.

33.) Multiparameter optimized nucleic acid molecule according to claim 28, wherein the T1R-type GPCR, is a T1R1, T1R2 or T1R3 or a functionally equivalent receptor protein.

34.) Multicistronic expression vector according to claim 17, wherein the promoter is selected host cell, more preferably being selected from the group consisting of: cytomegalovirus promoter (P-CMV), human elongation factor 1 alpha promoter (P-E1.alpha.), human ubiquitin promoter (P-ubi), simian virus promoter (P-SV40), Rous sarcoma virus long terminal repeat promoter (P--RSV-LTR).

35.) Multicistronic expression vector according to claim 17, wherein the GPCR.sub.1 and the GPCR.sub.2 are independently from another selected from the group consisting of T1R receptors.

36.) Multicistronic expression vector according to claim 31, wherein the GPCR.sub.1 and GPCR.sub.2 are a combination of T1R1-T1R3 or T1R2-T1R3.

37.) A vector according to claim 17, wherein the multicistronic cloning vector additionally comprises a cistron coding for G-alpha 15 or an equivalent thereof, the g-protein preferably being fused in-frame to above GPCR.sub.1 and/or GPCR.sub.2.

38.) Cell lines stably transfected with a vector according to claim 16.

39.) A vector according to claim 17, wherein the g-protein is fused in-frame to the GPCR.sub.1 and/or GPCR.sub.2
Description



CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims priority through its parent application, European Patent Application No. 06011710.8 filed Jun. 7, 2006 hereby incorporated by reference herein in its entirety.

FIELD OF THE INVENTION

[0002] The present invention relates to a screening method for the identification of modulators (agonists as well as antagonists) of selected GPCRs and the thus identified modulators. In a preferred embodiment of the present invention these modulators may be taste modulators. The present invention relates further to recombinant vector systems for the stable, heterologous expression of selected heterodimeric g-protein coupled receptors (GPCRs) in eucaryotic host cells. The functional expression of engineered GPCRs for the perception of sweet and L-amino acid taste and the use of said receptors for the identification of functional ligands is disclosed.

BACKGROUND OF THE INVENTION

[0003] In this specification, a number of documents are cited. The disclosure of these documents, including manufacturer's manuals and patent applications or patents, is herewith incorporated by reference in its entirety.

[0004] GPCRs represent the largest family of cell surface receptors with an estimated number of up to 1000 genes within the human genome characterized by a seven-transmembrane configuration as their main feature. (Bockaert and Pin, 1999; Pierce et al., 2002). GPCRs are activated by a multitude of different ligands, including peptides, proteins, lipids, small molecules, ions or even photons. Activated GPCRs alter their conformation allowing it to catalyze the exchange of guanosine diphosphate (GDP) for guanosine triphosphate (GTP) on the .alpha.-subunit of a heterotrimeric g-protein coupled to the GPCR. Heterotrimeric G-proteins composed of one out of 18 different .alpha.-subunits, one out of 5 different .beta.-subunits and one out of 11 different .gamma.-subunits are usually classified by the nature of their .alpha.-subunit and generally grouped into four main classes: G.sub.s, which activates adenylyl cyclase; G.sub.i, which inhibits adenylyl cyclase; G.sub.q, which activates phospholipase C; and G.sub.12/13 with heterologous functions. In addition to the .alpha.-subunit dependent signaling the .beta./.gamma.-subunits can function as signaling molecules on their own. GPCR dependent signaling becomes even more complex if it is considered that these receptors can exist as homo-oligomeric or hetero-oligomeric complexes. (George et al., 2002; Milligan et al., 2003; Salahpour et al., 2000). Hence, it is not surprising that GPCRs are responsible for the regulation of a wide variety of different physiological processes.

[0005] Recently the role of GPCRs in human senses like vision, olfaction and taste has been subject of intensified investigations. While the participation of the GPCR rhodopsin in visual sensing is one of the most comprehensively examined g-protein coupled receptor signaling examples of the last 30 years (Maeda et al., 2003), the role of GPCRs in olfaction and bitter taste as well as sweet taste was discovered in the 1990ies. (Buck and Axel, 1991; Firestein, 2001; Lindemann, 1996b; Lindemann, 2001).

[0006] The discovery of GPCR signaling in taste perception is closely connected to the discovery of tastant specific signaling in vertebrate taste cells. In electrophysical and biochemical studies it was apparent that tastant derived signaling resulted in typical GPCR dependent second messenger induction, e.g. cyclic nucleosides (cAMP, cGMP), inositol tri-phosphate (IP3) or calcium. (Kinnamon und Cummings, 1992; Kinnamon und Margolskee, 1996; Lindemann, 1996a). The participation of GPCRs in taste perception was further approved by the finding of the g-protein gustducin specifically expressed in vertebrate taste cells. (McLaughlin et al., 1992; Wong et al., 1996). On the other hand it was known from genetic mouse studies that the ability to sense sweet taste of e.g. saccharin was linked to the so called sac locus on mouse chromosome 4. (Bachmanov et al., 2001; Lush, 1989; Lush et al., 1995). Based on these data it was obvious to search for GPCR sequence tags in taste cell derived subtracted cDNA libraries or by performing genomic sequence scanning to further narrow down the mouse sac locus for the identification of GPCR analogs as putative taste receptors. These two approaches led to the rat, mouse and human receptor DNA sequences for the taste GPCRs T1R1 and T1R2 (Hoon et al., 1999; Hoon and Ryba, 1997) as well as T1R3. (Kitagawa et al., 2001; Li et al., 2001; Max et al., 2001; Montmayeur et al., 2001; Sainz et al., 2001). Homology alignments revealed that these taste receptors like the homodimeric metabotrophic glutamate receptor (mGluR), the heterodimeric .gamma.-aminobutyric acid type B receptor (GABA.sub.BR) and homodimeric extracellular calcium receptors are members of the small family of class C GPCRs. As a common characteristic most of the class C receptors exhibit a large extracellular amino terminal domain composed of a so called venus flytrap module (VFTM) and a cysteine rich domain (CRD) that connects the VFTM to the heptahelical domain. (Pin et al., 2003). Besides that homo- as well as hetero-oligomerisation was described for several of these class C receptors. (Bai et al., 1998; Kaupmann et al., 1998; Kunishima et al., 2000; White et al., 1998). Consequently, the characteristic feature of GPCR oligomerisation of class C receptors was tested for the putative sweet taste receptors T1R1, T1R2 and T1R3.

[0007] By recombinant heterologous expression in eucaryotic cell systems a functional expression and tastant specific activation of an artificially linked G-Protein dependent signaling cascade was demonstrated by calcium imaging. T1R receptors assemble to build up functional taste receptors. As a result of several investigations it was shown that the heterodimeric T1R1/T1R3 functions as a glutamate (umami) and L-amino acid receptor whereas the heterodimeric T1R2/T1R3 functions as a high affinity sugar and artificial sweetener receptor. Particularly, heterodimeric co-expression of T1R1 and T1R3 results in taste receptors that respond to umami taste and monosodium glutamate stimuli whereas heterodimeric co-expression of T1R2 and T1R3 results in taste receptors that respond to sweet stimuli like diverse sugars (e.g. glucose and sucrose), artificial sweetener (e.g. acesulfam K, cyclamat, saccharin) and sweet proteins like monellin, thaumatin, brazzein (Li et al., 2002; Nelson et al., 2002; Nelson et al., 2001; Zhao et al., 2002). A similar chronicle could be generated for the identification of GPCRs for the perception of bitter taste with the exception that so far no homo- or oligomerisation has been reported for these so called T2R-GPCRs. (Meyerhof et al., 2005).

[0008] The above discussed identification of genes coding for receptors responsible e.g. for taste perception, together with cloning said genes into appropriate vectors for the expression of said proteins in eukaryotic cells and the transformation of said cells with said vectors raised the expectation that screening systems and/or screening methods for GPCR modulators, i.e. agonists and antagonists of the above detailed receptors should be easy to be developed within a reasonable time.

[0009] This is reflected by a huge and still growing number of patent applications in this field.

[0010] The cloning of T1R1 is disclosed in different patent applications, e.g. in WO 03/025137; in WO 00/06952 (wherein it is designated GPCR-B3) US020040191862A1 and WO2005/033125.

[0011] The cloning of T1R2 is disclosed in patent applications WO 03/025137, US020040191862A1 and US020030040045A1

[0012] The cloning of T1R3 is disclosed in patent applications WO 03/025137, WO 03/025137, US020040191862A1 and US020030040045A1

[0013] A system for the expression of said proteins in eukaryotic cells is disclosed in patent applications WO 03/025137, WO 00/06952, US20040191862A1, WO2004069191 and US20030040045A1.

[0014] A screening system for putative taste modulators is disclosed in patent applications WO 00/06952, WO2004069191 and US20030040045A1.

[0015] Yet, nothing is to be told about the successful identification of new modulators, e.g. new artificial taste modulators such as new sweeteners utilizing such screening methods/systems.

[0016] The ongoing debate on obesity in developed countries and the growing health consciousness of consumers lead to an increasing demand of food and beverages with significant calorie reduction compared to products fully sweetened with carbohydrates such as sucrose, glucose, fructose or syrups such as HFCS 55 or 42. As the consumer usually is not willing to compromise on taste products should have similar sweetness intensity and taste quality as products regularly sweetened with these carbohydrates.

[0017] High intensity sweeteners are substances, which have no or virtually no calories and a sweetness potency several times higher than sugar. High intensity sweeteners or blends of high intensity sweeteners are used in food and beverages to achieve a sweet taste without adding calories to the products.

[0018] Most commonly used high intensity sweeteners are not from natural origin; They were discovered accidentally and are chemically synthesized. Most of them have a widespread approval in a large number of countries. Examples are substances such as acesulfame K, alitame, aspartame, cyclamate, neohesperidine dihydrochalcone, neotame, saccharin, and sucralose.

[0019] However, no high-intensity sweetener matches the taste profile of sugar completely. They differ in characteristics such as sweetness profile, side taste and off-taste characteristics. Proper blending of different high intensity sweeteners is known to overcome part of the taste limitations of single high-intensity sweeteners. But even if a more sugar-like sweetness profile is achieved in products with high-intensity sweeteners only, they still can be distinguished sensorically from their counterparts with just sugar or other carbohydrates by lack of mouthfeel and reduced flavour characteristics. Therefore a need exists for new high-intensity sweeteners which offer either alone or in blends with existing sweeteners sweetness profiles and flavour characteristics much closer to sugar than the existing products can offer.

[0020] Besides calorie reduction many of today's consumers are seeking for food and beverage products either without artificial additives or even being fully organic. Theoretically natural high-intensity sweeteners could fulfil this demand. A number of natural high-intensity sweeteners were discovered throughout past years such as stevioside, rebaudioside, brazzein, thaumatin, mogroside, glycyrrhizin, monatin, abrusoside, monellin, phyllodulcin and others. These are substances which naturally occur in different plants and can be obtained by selective extraction measures. Besides very limited approvals and in some cases difficulties to extract products on an industrial scale none of these products can claim to offer a sugar-like taste. In fact, all of these substances show a sweetness with a far slower onset than sucrose and a very lingering sweetness. Most of these products have strong side-taste and aftertaste characteristics such as bitter, mentholic or liquorice notes or show even strong cooling or numbing sensations. Therefore some of these products, e.g. thaumatin, can be rather regarded as being flavour enhancer than sweetener. Blending of two or more of these substances can not overcome these taste limitations. Therefore in the area of natural sweetener the need for new high-intensity sweeteners with a taste profile closer to sugar is even stronger than in the case of artificial sweeteners (O'Brien Nabors, 2001; Leatherhead Food RA, 2000; Grenby, 1996; von Rymon Lipinski und Schiweck, 1991).

[0021] Therefore, there still exists a need in the art to identify and isolate new substances which may be used as modulators of taste perception, e.g. as sweeteners. Notwithstanding the above, because of the high importance of these GPCRs in vivo, and the many different functions associated with said receptors, it has to be assumed that many of the modulators of GPCRs that might be identified by the method of the present invention may be of practical value.

[0022] Therefore, the availability of a simple and reliable screening system for modulators of said receptors would be of big importance.

[0023] In multicistronic expression vectors the coding sequences of different proteins are under the control of only one promoter and the different cistrons are connected via virus derived internal ribosomal entry sites (IRES) or cap independent translation enhancer (CITE). IRES or CITE elements confer a translation initiation independent from the otherwise necessary 5'-end of a messenger RNA, which is recognized by the eucaryotic ribosomes to start their scanning process for the first accessible translational start codon. (Fux et al., 2004; Hellen and Sarnow, 2001). So far multicistronic expression vectors have been described basically as dicistronic expression units for the coupled expression of a gene of interest linked via an cap-independent translation initiation site to a resistance marker (confering resistance to e.g. hygromycin, zeocin, neomycin) enabling selection of stable cell lines for heterologous mammalian expression studies. For this approach IRES or CITE dependent expression vectors are commercially available.

[0024] Reports on genuine multicistronic expression studies in mammalian systems with descriptions of tri-cistronic or even quadro-cistronic heterologous expression studies are rare and for the most part intended to improve inducible protein expression e.g. for gene therapy applications. However in this pioneering work multicistronic expressions have been mostly performed with small and soluble proteins like reporter genes (green fluorescent protein, yellow fluorescent protein, red fluorescent proteins, secreted alkaline phosphatase, secreted amylase) or engineered transactivators e.g. for macrolide- or streptogramin dependent expression or selection markers. Although these studies are aimed at potential therapeutic protein expression in gene therapy applications, only few genes with a therapeutic potential are mentioned within this studies (e.g. vascular endothelial growth factor (VEGF); the oncoprotein bcl-2). (Fussenegger et al., 1998; Hartenbach and Fussenegger, 2005; Kramer et al., 2003; Moser et al., 2000; Weber et al., 2005).

[0025] Concerning the expression of taste receptors there is one report on dicistronic expression of mouse taste receptors (mT2R8/5; mT1R3) each fused to green fluorescent protein and linked via an IRES element to red fluorescent protein. This approach was applied to trace and localize the expression pattern of taste receptors in neurons (Sugita and Shiba, 2005).

SUMMARY OF ADVANTAGEOUS EMBODIMENTS OF THE INVENTION

[0026] Therefore, the technical problem underlying the present invention was to provide a cell based screening system for modulators of GPCRs, i.e. establishing a method which makes it possible to successfully identify important modulators of some selected GPCRs, preferably T1R-type taste GPCRs. It was a further problem underlying the present invention to provide means and methods for the isolation of identified modulators without undue burden.

[0027] The solution to the above mentioned technical problems is achieved by providing the embodiments characterized in the claims.

[0028] Therefore, specially preferred embodiments of the present invention are means and methods for the expression of heterodimeric T1R-type taste GPCRs, wherein the expression of these heterodimeric T1R-type taste GPCRs is effected by modulating the GPCR coding sequences as well as their expression in multicistronic operons.

BRIEF DESCRIPTION OF THE FIGURES

[0029] FIG. 1 depicts different GPCR nucleotide sequences of the TIR class, presented in an alignment analysis, with the underlying distance matrix shown below;

[0030] FIG. 2 depicts an exemplary multicistronic eukaryotic expression vector pTrix-Eb-R2R3;

[0031] FIG. 3 depicts a Ca.sup.z+ assay with HEK_Ga15#17R2R3b #8 measuring several sweet tastants;

[0032] FIG. 4 depicts an alternative exemplary multicistronic eukaryotic expression vector p Trix-Eb-R2R3.

DETAILED DESCRIPTION OF ADVANTAGEOUS EMBODIMENTS OF THE INVENTION

[0033] The above problem can surprisingly yet easily be solved by providing a method for the identification of modulators of GPCRs, comprising the steps of: [0034] a. transforming eucaryotic host cells with genetic sequences coding for one or more than one GPCR(s), [0035] b. cultivation of the transformed host cells under conditions sufficient to ensure the functional expression of said one or more GPCRs, [0036] c. contacting the cultivated host cell expressing the one or more GPCRs in a functional manner with a potential modulator of the selected one or more GPCRs, [0037] d. measuring a specific cellular response on exposure of the transformed host cell to the potential modulator, and [0038] e. selecting an identificate, which induces a specific response.

[0039] Within the context of the present invention the term "modulator of GPCRs" is intended to mean a substance, which if applied in vivo, would (a) trigger an intracellular response by binding to the selected GPCR or (b) would inhibit such a response in the presence of a sweet compound. E.g. in the case of sweet receptors as the selected GPCR this modulator could be a substance which tastes sweet or which potentiates the sweetness of a sweet tasting compound without being sweet on its own or which inhibits the sweetness of sweet substances.

[0040] The term "conditions sufficient to ensure the functional expression" shall be understood as a summary of all conditions needed to ensure the functional expression of the GPCRs, rather than cell culture conditions. In a preferred embodiment of the present invention this term pertains to the vector used for transformation being a tricistronic vector. Further, this expression encompasses culturing cells which comprise a functional g-.protein. Under any circumstances, the functional expression of the GPCR(s) must be ensured.

[0041] The terms "functional expression" or "expressing in a functional manner" means within the context of the present invention that the selected receptor maintains its ability to interact specifically with the same substances the receptor would interact with in the in vivo situation.

[0042] By "selected cellular response" e.g. changes in intracellular calcium levels have to be understood, which can be measured easily e.g. by using dyes like Calcium 3, Fura-2, Fluo-4, Indo-1 or the calcium dependent reporter protein aequorin. Any cellular response related to the activity of the selected GPCRs is encompassed. Further examples of measuring the activity of GPCRs are (i) the activation of the second messenger cyclic adenosine monophosphate (cAMP) which can be quantified in GPCR dependent cell based assays with luminescent tags or with cAMP dependent reporter gene expression (e.g. Luciferase, SEAP or similar) or with the melanophore technology using frog skin cells; (ii) the measurement of .beta.-arrestin binding to the targeted GPCR after ligand induced activation of the GPCR via bioluminescence resonance energy transfer (BRET) measurements or GFP-tagged .beta.-arrestin movement imaging; (iii) measuring the agonist occupation of a GPCR and respectively the activation of the associated G-Protein, which can be quantified by using the radioactive, non-hydrolyzable analogue of GTP, [35S]GTP.gamma.S; (IV) the use of response element controlled reporter gene assays for the readout of GPCR activated interconnected pathways, including those involving MAP kinases, nonreceptor tyrosine kinases, receptor tyrosine kinases, phosphatidylinositol 3-kinases, and JNKs. (Eglen, 2005; Filmore, 2004; Milligan, 2003).

[0043] The term "identificate" is directed to a potential modulator after being identified as a true modulator of GPCR or the GPCR specific signaling cascade.

[0044] In a preferred embodiment the method is characterized in that the host cell is selected from the group consisting of: HEK293 (human embryo kidney), Hela (Human Negroid cervix epitheloid carcinoma), HT29 (Human Caucasian colon adenocarcinoma grade II), A431 (human squamous carcinoma), IMR 32 (human caucasian neuroblastoma), K562 (Human Caucasian chronic myelogenous leukaemia), U937 (Human Caucasian histiocytic lymphoma), MDA-MB-231 (Human Caucasian breast adenocarcinoma), SK-N-BE(2) (Human Caucasian neuroblastoma), SH-SY5Y (Human neuroblastoma), HL60 (human promyelocytic leukemia) or eukaryotic non-human cell lines like CHO-K1 (Hamster Chinese ovary), COS-7 (Monkey African green kidney, SV40 transformed), S49 (mouse lymphoma), Ltk (Mouse C34/an connective tissue), NG108-15 (Mouse neuroblastoma.times.Rat glioma hybrid), B50 (Rat nervous tissue neuronal, ECACC), C6 (Rat glial tumour), Jurkat (Human leukaemic T cell lymphoblast), BHK (Hamster Syrian kidney), Neuro-2a (Mouse Albino neuroblastoma), NIH/3T3 (mouse embryo fibroblast), prefarably HEK293 (human embryo kidney), Hela (Human Negroid cervix epitheloid carcinoma), CHO-K1 (Hamster Chinese ovary) or Neuro-2a (Mouse Albino neuroblastoma).

[0045] It is essential that the host cell expresses a functional g-protein, preferably G-alpha15, either naturally or by means of genetic alteration of the host cell. Means for genetic alteration of eucaryotic cells are well known in the art, and need not to be discussed in depth here. The DNA-Sequences of G-proteins, e.g. G-alpha15 have already been described in the art.

[0046] A further preferred embodiment of the method of the present invention is characterized in that the one or more GPCR(s) is (are) selected from the group consisting of T1R or T2R taste receptors, preferably a T1R-type GPCR, especially T1R1, T1R2 and/or T1R3.

[0047] A further preferred embodiment of the method of the present invention is characterized in that two or more GPCRs are expressed in a heterologous co-expression of at least two different T1R-type GPCRs, preferably T1R1/T1R3, more preferably T1R2/T1R3.

[0048] A further preferred embodiment of the method of the present invention is characterized in that the transformation is accomplished with a multicistronic vector, preferably a tricistronic vector.

[0049] The vector of the present invention is an expression vector. By the term "expression vector" it is meant that the vector is used to transform a selected eucaryotic host cell, which, after transformation, expresses the gene or genes encoded by said vector. Expression vectors may be e.g. cloning vectors, binary vectors or integrating vector. Expression comprises transcription of the encoded nucleic acids into a functional (translatable) mRNA. Therefore, the respective control elements should be present, e.g. a sequence promoting transcription of the messenger (a promoter) and (optionally) a polyadenylation signal. Means for the expression of heterologous genes in eukaryotic cells are very well studied.

[0050] For the propagation of such vectors, usually prokaryotic cells such as E. coli are used. Therefore, although the vectors of the present invention are designed to work as expression vectors in eukaryotic cells, they also carry elements for propagation in prokaryotic cells, e.g. an origin of replication (ori) and an antibiotics resistance gene, e.g. amp.sup.r, kan.sup.r and similar. Means for the propagation of vectors in prokaryotes are well known in the art.

[0051] A list of possible eukaryotic expression vectors usable in the present invention, optionally usable for propagation in a prokaryotic host, and already commercially available comprises: pCR1000, pCDM8, pcDNA1, pcDNA1.1, pcDNA1/Amp, pcDNA1.1/Amp and pcDNA1/Neo, pcDNA3, pcDNA3.1, pcDNA3.2, pcDNA6.2, pDEST26, pDEST27, pCR3.1, pcDNA3.1 His, pDisplay (Invitrogen); pTriEx (pTriEx-2-Hygro) (Novagen); pSI, pCI (pCI-neo), pTargeT (Promega); pERV3, pFB-ERV, pCFB-EGSH, pDual, pCMV-Script (Stratagene); pNEBR (New England Biolabs), pEAK (Edge Biosystems).

[0052] A further preferred embodiment of the method of the present invention is characterized in that the multicistronic vector comprises a multicistronic expression unit comprising downstream from a promoter for the expression in an eucaryotic host and functionally linked thereto, the following cistrons: [0053] a. GPCR.sub.1 [0054] b. GPCR.sub.2 and [0055] c. a selection marker, wherein the promoter preferably is a strong promoter suitable for use in the selected host cell, more preferably being selected from the group consisting of cytomegalovirus promoter (P-CMV), human elongation factor 1 alpha promoter (P-E1.alpha.), human ubiquitin promoter (P-ubi), simian virus promoter (P-SV40), Rous sarcoma virus long terminal repeat promoter (P-RSV-LTR) and similar, wherein the GPCR, and the GPCR.sub.2 are independently from another being selected from the group consisting of T1R or T2R taste receptors, preferably of the group of T1R receptors, more preferably a combination of T1R1-T1R3 or T1R2-T1R3, wherein the selection marker is being selected from the group consisting of hygromycin.sup.r, zeocin.sup.r, neomycin.sup.r, blasticidin.sup.r or puromycin.sup.r, and wherein both the GPCR1 and the GPCR2 as well as the selection marker are functionally connected by intervening IRES selected from the group consisting of IRES.sub.EMCV, derived from encephalomyocarditis virus (synonym: CITE.sub.EMCV); IRES.sub.GTX, derived from the GTX homeodomain mRNA; IRES.sub.Rbm3, derived from cold-inducible Rbm3; IRES.sub.PV, derived of polioviral origin, IRES.sub.RV, derived from rhinovirus, IRESFMDV, derived from food and mouth disease virus; IRE.sub.HV, derived from hepatitis C virus, IRES.sub.CSFV, derived from classic swine fever virus, IRES.sub.BVDV, derived from bovine viral diarrhea virus; IRES.sub.FMLV, derived from friend murine leukemia virus gag mRNA; IRES.sub.MMLV, derived from moloney murine leukemia virus gag mRNA; IRES.sub.HIV, derived from human immunodefiency virus env mRNA; IRES.sub.PSIV, derived from Plautia stali intestine virus; IRES.sub.RPV, derived from Rhopalosiphum padi virus; IRES.sub.KSH, derived from Karposi's sarcoma-associated herpesvirus, preferably the IRES.sub.EMCV being derived from encephalomyocarditis virus (synonym: CITE.sub.EMCV) and wherein the multicistronic expression unit is terminated by a polyadenylation signal.

[0056] The term "functionally linked thereto" means within the context of the present invention that the components described are linked together to function in their intended manner.

[0057] A further preferred embodiment of the method of the present invention is characterized in that the multicistronic vector additionally comprises a genetic sequence coding for a g-protein or an equivalent thereof, preferably G-alpha 15 or an equivalent thereof, which is coded as an monocistronic unit consisting of a promoter, the gene of the selected g-protein and a polyadenylation site.

[0058] A further preferred embodiment of the method of the present invention is characterized in that the multicistronic vector additionally comprises a genetic sequence coding for a g-protein or an equivalent thereof, preferably G-alpha 15 or an equivalent thereof, which is coded as a fourth cistron in a quadrocistronic arrangement via an additional IRES element.

[0059] A further preferred embodiment of the method of the present invention is characterized in that the multicistronic vector additionally comprises a genetic sequence coding for a g-protein or an equivalent thereof, preferably G-alpha 15 or an equivalent thereof, the g-protein preferably being fused in-frame to above GPCR.sub.1 and/or GPCR.sub.2.

[0060] According to the present invention G proteins such as G-alpha15 or G-alpha16 or other promiscuous G proteins or G protein variants, or an endogenous G protein like gustducin, or another g-protein that when expressed in association with the multicistronically encoded GPCR(s) produces a functional read out may be used. In addition, G-beta and G-gamma proteins may also be used.

[0061] Subvariants of G-alpha 15 and/or G-alpha 16 with modified N-termini are also well known in the art, and can be used accordingly.

[0062] Essentially any chemical compound can be employed as a potential modulator or ligand in the assays according to the present invention. Compounds tested as G-protein coupled receptor modulators can be any small chemical compound, or biological entity (e.g., protein, sugar, nucleic acid, lipid). Test compounds will typically be small chemical molecules and peptides. Generally, the compounds used as potential modulators can be dissolved in aqueous or organic (e.g., DMSO-based) solutions. The assays are designed to screen large chemical libraries by automating the assay steps and providing compounds from any convenient source. Assays are typically run in parallel, for example, in microtiter formats on microtiter plates in robotic assays. There are many suppliers of chemical compounds, including Sigma (St. Louis, Mo.), Aldrich (St. Louis, Mo.), Sigma-Aldrich (St. Louis, Mo.), Fluka Chemika-Biochemica Analytika (Buchs, Switzerland), for example. Also, compounds may be synthesized by methods known in the art.

[0063] So-called high throughput screening methods typically involve providing a combinatorial chemical or peptide library containing a large number of potential therapeutic compounds (e.g., ligand or modulator compounds). Such combinatorial chemical libraries or ligand libraries are then screened in one or more assays to identify those library members (e.g., particular chemical species or subclasses) that display a desired characteristic activity. The compounds so identified can serve as conventional lead compounds, or can themselves be used as potential or actual therapeutics.

[0064] A combinatorial chemical library is a collection of diverse chemical compounds generated either by chemical synthesis or biological synthesis, by combining a number of chemical building blocks (i.e., reagents such as amino acids). As an example, a linear combinatorial library, e.g., a polypeptide or peptide library, is formed by combining a set of chemical building blocks in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide or peptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks.

[0065] The preparation and screening of combinatorial chemical libraries is well known to those having skill in the pertinent art. Combinatorial libraries include, without limitation, peptide libraries (e.g. U.S. Pat. No. 5,010,175; Furka, 1991, Int. J. Pept. Prot. Res., 37:487-493; and Houghton et al., 1991, Nature, 354:84-88). Other chemistries for generating chemical diversity libraries can also be used. Nonlimiting examples of chemical diversity library chemistries include, peptides (PCT Publication No. WO 91/019735), encoded peptides (PCT Publication No. WO 93/20242), random bio-oligomers (PCT Publication No. WO 92/00091), benzodiazepines (U.S. Pat. No. 5,288,514), diversomers such as hydantoins, benzodiazepines and dipeptides (Hobbs et al., 1993, Proc. Natl. Acad. Sci. USA, 90:6909-6913), vinylogous polypeptides (Hagihara et al., 1992, J. Amer. Chem. Soc., 114:6568), nonpeptidal peptidomimetics with glucose scaffolding (Hirschmann et al., 1992, J. Amer. Chem. Soc., 114:9217-9218), analogous organic synthesis of small compound libraries (Chen et al., 1994, J. Amer. Chem. Soc., 116:2661), oligocarbamates (Cho et al., 1993, Science, 261:1303), and/or peptidyl phosphonates (Campbell et al., 1994, J. Org. Chem., 59:658), nucleic acid libraries (see Ausubel, Berger and Sambrook, all supra), peptide nucleic acid libraries (U.S. Pat. No. 5,539,083), antibody libraries (e.g., Vaughn et al., 1996, Nature Biotechnology, 14(3):309-314) and PCT/US96/10287), carbohydrate libraries (e.g., Liang et al., 1996, Science, 274-1520-1522) and U.S. Pat. No. 5,593,853), small organic molecule libraries (e.g., benzodiazepines, Baum C&EN, Jan. 18, 1993, page 33; and U.S. Pat. No. 5,288,514; isoprenoids, U.S. Pat. No. 5,569,588; thiazolidinones and metathiazanones, U.S. Pat. No. 5,549,974; pyrrolidines, U.S. Pat. Nos. 5,525,735 and 5,519,134; morpholino compounds, U.S. Pat. No. 5,506,337; and the like).

[0066] Devices for the preparation of combinatorial libraries are commercially available (e.g., 357 MPS, 390 MPS, Advanced Chem Tech, Louisville Ky.; Symphony, Rainin, Woburn, Mass.; 433A Applied Biosystems, Foster City, Calif.; 9050 Plus, Millipore, Bedford, Mass.). In addition, a large number of combinatorial libraries are commercially available (e.g., ComGenex, Princeton, N.J.; Asinex, Moscow, Russia; Tripos, Inc., St. Louis, Mo.; ChemStar, Ltd., Moscow, Russia; 3D Pharmaceuticals, Exton, Pa.; Martek Biosciences, Columbia, Md., and the like).

[0067] In one embodiment, the invention provides solid phase based in vitro assays in a high throughput format, where the cell or tissue expressing an ion channel is attached to a solid phase substrate. In such high throughput assays, it is possible to screen up to several thousand different modulators or ligands in a single day. In particular, each well of a microtiter plate can be used to perform a separate assay against a selected potential modulator, or, if concentration or incubation time effects are to be observed, every 5-10 wells can test a single modulator. Thus, a single standard microtiter plate can assay about 96 modulators. If 1536 well plates are used, then a single plate can easily assay from about 100 to about 1500 different compounds. It is possible to assay several different plates per day; thus, for example, assay screens for up to about 6,000-20,000 different compounds are possible using the described integrated systems.

[0068] In another aspect, the present invention encompasses screening and small molecule (e.g., drug) detection assays which involve the detection or identification of small molecules that can bind to a given protein, i.e., a taste receptor polypeptide or peptide. Particularly preferred are assays suitable for high throughput screening methodologies.

[0069] In such binding-based detection, identification, or screening assays, a functional assay is not typically required. All that is needed is a target protein, preferably substantially purified, and a library or panel of compounds (e.g., ligands, drugs, small molecules) or biological entities to be screened or assayed for binding to the protein target. Preferably, most small molecules that bind to the target protein will modulate activity in some manner, due to preferential, higher affinity binding to functional areas or sites on the protein.

[0070] An example of such an assay is the fluorescence based thermal shift assay (3-Dimensional Pharmaceuticals, Inc., 3DP, Exton, Pa.) as described in U.S. Pat. Nos. 6,020,141 and 6,036,920 to Pantoliano et al.; see also, J. Zimmerman, 2000, Gen. Eng. News, 20(8)). The assay allows the detection of small molecules (e.g., drugs, ligands) that bind to expressed, and preferably purified, ion channel polypeptide based on affinity of binding determinations by analyzing thermal unfolding curves of protein-drug or ligand complexes. The drugs or binding molecules determined by this technique can be further assayed, if desired, by methods, such as those described herein, to determine if the molecules affect or modulate function or activity of the target protein.

[0071] Compounds which are identified according to the methods provided herein, and which modulate or regulate the biological activity or physiology of the T1R polypeptides according to the present invention are a preferred embodiment of this invention. It is contemplated that such modulatory compounds may be employed as outlined above.

[0072] Assays that may be utilized with one or more T1Rs according to the invention include by way of example assays that utilize a genetic selection for living cells; assays that utilize whole cells or membrane fragments or purified taste receptor proteins; assays that utilize second messengers such as cAMP and IP3, assays that detect the translocation of arrestin to the cell surface, assays that detect the loss of receptor expression on the cell surface (internalization) by tested ligands, direct ligand-binding assays, competitive-binding assays with inhibitors, assays using in vitro translated protein, assays that detect conformational changes upon the binding of a ligand (e.g., as evidenced by proteolysis, fluorescence, or NMR), behavioral assays that utilize transgenic non-human animals that express a taste GPCR or a combination thereof, such as flies, worms, or mice, assays that utilize cells infected with recombinant viruses that contain taste GPCR genes, preferably measuring the change of intracellular calcium levels relative to intracellular calcium levels without contacting the cell to the modulator.

[0073] A further preferred embodiment of the method of the present invention is characterized in that the change is an increase.

[0074] A further preferred embodiment of the method of the present invention is characterized in that the change is an increase relative to intracellular calcium levels when the cell is contacted by a modulator in the peresence of a sweet compound thereby enhancing the calcium level above the level of the level generated by the sweet compound alone. In this case the modulator can either be sweet on its own or either be a tasteless compound without having an bioactive potential to activate T1Rs or combinations thereof on its own. The sweet compound preferably being selected from the group consisting of glucose, fructose, saccharose, acesulfam K, saccharin, cyclamat, aspartam, xylitol, stevioside, sucralose, thaumatin, monellin, brazzein, perillartine, glycyrrhizin, sucronic acid, P-4000, SC45647, NC174, neohesperidin and sweet tasting amino acids, more prefarably selected from the group consisting of glucose, fructose, saccharose or xylitol.

[0075] A further preferred embodiment of the method of the present invention is characterized in that the change is a decrease relative to intracellular calcium levels when the cell is contacted by a sweet compound instead of the modulator, the sweet compound preferably being selected from the non-exclusive group consisting of glucose, fructose, saccharose, acesulfam K, saccharin, cyclamat, aspartam, xylitol, stevioside, sucralose, thaumatin, monellin, brazzein, perillartine, glycyrrhizin, sucronic acid, P-4000, SC45647, NC174, neohesperidin and sweet tasting amino acids.

[0076] A still further preferred embodiment of the present invention are multiparameter optimized nucleic acid molecules coding for a GPCR, preferably a T1R-type GPCR, more preferably a T1R1, T1R2 or T1R3 or a functionally equivalent receptor protein, and even more preferably coding for proteins consisting of the amino acids sequences according to SEQ ID NO: 1 and/or SEQ ID NO: 2 or, optionally, functionally equivalent proteins.

[0077] These nucleic acid molecules in a preferred embodiment have the sequence as depicted in SEQ ID NOs 3 and 4.

[0078] Nucleic acid molecules which are functionally equivalent to the molecules as depicted in SEQ ID NOs 3 and 4 are also encompassed by the present invention.

[0079] Within the context of the present invention, functionally equivalent proteins have the same or a very similar function in-vivo. Preferably, functionally equivalent proteins share at least 60%, more preferably at least 80%, especially at least 90%, advantageously at least 99% identity in their amino acid sequence.

[0080] A functionally equivalent nucleic acid codes for a functionally equivalent protein and it is optimized for expression in a eucaryotic cell. According to a multiparameter optimization the codon usage was adapted to the codon bias of Homo sapiens genes. In addition, regions of very high (>80%) or very low (<30%) GC content have been avoided where possible. During the optimization process following cis-acting sequence motifs were avoided:

a) internal TATA-boxes, chi-sites and ribosomal entry sites b) AT-rich or GC-rich sequence stretches c) ARE, INS, CRS sequence elements d) repeat sequences and RNA secondary structures, as well as e) (cryptic) splice donor and acceptor sites, branch points.

[0081] The optimization process, starting from the human wild type sequences (wt_hT1R) revealed several sequences; surprisingly the sequences which offered the best performance (sh_T1R) in generating functional stable cell lines on the basis of the multicistronic expression approach were not those with the theoretically optimal sequence (opt_hT1R) nor the wild type sequences. The differences of the here examplified sequences are illustrated as a phylogenetic alignment (generated by the bioinformatic software clustal X) with an accompanying relational distance matrix in FIG. 1.

[0082] A very preferred embodiment of the present invention are multicistronic expression vectors comprising more than one cistron coding for a GPCR.

[0083] The said multicistronic expression vector preferably comprises downstream from a promoter for the expression in an eucaryotic host cell and functionally linked thereto, the following cistrons: [0084] a. GPCR.sub.1 [0085] b. GPCR.sub.2 and a [0086] c. selection marker, wherein the promoter preferably is a strong promoter, more preferably being selected from the group consisting of cytomegalovirus promoter (P-CMV), human elongation factor 1 alpha promoter (P-E1.alpha.), human ubiquitin promoter (P-ubi), simian virus promoter (P-SV40), Rous sarcoma virus long terminal repeat promoter (P-RSV-LTR) and similar wherein the GPCR.sub.1 and the GPCR.sub.2 are independently from another being selected from the group consisting of T1R or T2R taste receptors, preferably of the group of T1R receptors, more preferably a combination of T1R1-T1R3 or T1R2-T1R3, and wherein the selection marker is being selected from the group consisting of hygromycin.sup.r, zeocin.sup.r, neomycin.sup.r, blasticidin.sup.r or puromycin.sup.r, and wherein both the GPCR1 and the GPCR2 as well as the selection marker are functionally connected by intervening IRES selected from the group consisting of IRES.sub.EMCV, derived from encephalomyocarditis virus (synonym: CITE.sub.EMCV); IRES.sub.GTX, derived from the GTX homeodomain mRNA; IRES.sub.Rbm3, derived from cold-inducible Rbm3; IRES.sub.PV, derived of polioviral origin, IRES.sub.RV, derived from rhinovirus, IRESFMDV, derived from food and mouth disease virus; IRE.sub.HV, derived from hepatitis C virus, IRES.sub.CSFV, derived from classic swine fever virus, IRES.sub.BVDV, derived from bovine viral diarrhea virus; IRES.sub.FMLV, derived from friend murine leukemia virus gag mRNA; IRES.sub.MMLV, derived from moloney murine leukemia virus gag mRNA; IRES.sub.HIV, derived from human immunodefiency virus env mRNA; IRES.sub.PSIV, derived from Plautia stali intestine virus; IRES.sub.RPV, derived from Rhopalosiphum padi virus; IRES.sub.KSH, derived from Karposi's sarcoma-associated herpesvirus, preferably, the IRES.sub.EMCV, derived from encephalomyocarditis virus (synonym: CITE.sub.EMCV) and wherein the multicistronic expression unit is terminated by a polyadenylation signal.

[0087] Another preferred embodiment of the present invention is a multicistronic vector as defined above, the vector additionally comprising a cistron coding for a G-protein, preferably G-alpha 15, the g-protein preferably being located between the last GPCR and the selection marker and being functionally connected to both of them via an IRES element as defined supra.

[0088] A further preferred embodiment are cell lines transformed with the vectors of the present invention. Respective eukaryotic cell lines are amphibian, worm, insect or mammalian cells such as CHO, Hela, Hek-293 and the like, e.g., cultured cells, explants, and cells in vivo.

[0089] Preferably, the host cell is selected from the group consisting of: HEK293 (human embryo kidney), Hela (Human Negroid cervix epitheloid carcinoma), HT29 (Human Caucasian colon adenocarcinoma grade II), A431 (human squamous carcinoma), IMR 32 (human caucasian neuroblastoma), K562 (Human Caucasian chronic myelogenous leukaemia), U937 (Human Caucasian histiocytic lymphoma), MDA-MB-231 (Human Caucasian breast adenocarcinoma), SK-N-BE(2) (Human Caucasian neuroblastoma), SH-SY5Y (Human neuroblastoma), HL60 (human promyelocytic leukemia) or eukaryotic non-human cell lines like CHO-KL (Hamster Chinese ovary), COS-7 (Monkey African green kidney, SV40 transformed), S49 (mouse lymphoma), Ltk (Mouse C34/An connective tissue), NG108-15 (Mouse neuroblastoma.times.Rat glioma hybrid), B50 (Rat nervous tissue neuronal, ECACC), C6 (Rat glial tumour), Jurkat (Human leukaemic T cell lymphoblast), BHK (Hamster Syrian kidney), Neuro-2a (Mouse Albino neuroblastoma), NIH/3T3 (mouse embryo fibroblast), prefarably HEK293 (human embryo kidney), Hela (Human Negroid cervix epitheloid carcinoma), CHO-K1 (Hamster Chinese ovary) or Neuro-2a (Mouse Albino neuroblastoma).

[0090] Further, preferred embodiments of the present invention are the modulators of GPCRs identifiable by the methods of the present invention and further characterized by being selected from the group consisting of sugars, steroids, tannins and lignans, terpenes, quinons, macrocycles, heterocycles, N-heterocycles and O-heterocycles, aliphatics and polyketides, flavonoids, proteins, peptides and amino acids, alkaloids and arenes, as stated above.

[0091] The intense investigations of the present inventors that let to the establishment of the method of the present invention showed that the recombinant co-expression of said taste receptors as stable integrants of eucaryotic host cells is hampered by a common instability. Although the phenomenon is far away from being understood, it might be possible that the expression and specific signal transduction of these T1R-type receptors interferes in a negative fashion with the cell physiology of such stable host cells.

[0092] Within the context of the present invention, an instable integration is characterized by a rapid loss of functionality within the first 10 passages of a selected recombinant cell line clone. Within these first ten passages it becomes obvious that more and more cells of a selected recombinant clone loose their characteristic functional taste GPCR expression, indicated by passage dependent incremental decrease of sweetener induced calcium inductions (Fluo-4 measurements). By way of contrast, the stable integrants of the present invention are characterized in that they show a functional expression of said taste receptors at least over 50 passages, indicated by stable sweetener induced calcium inductions (Fluo-4 measurements) with a selected taste receptors expressing cell line clone.

[0093] As becomes clear from the present specification of the invention, the use and the hetero-oligomeric expression of the taste receptor combinations T1R1/T1R3 and more preferred T1R2/T1R3 to identify compounds as modulators of sweet taste in the field of tastants are especially preferred embodiments of the present invention. Accordingly, this invention relates to recombinantly engineered T1R-type GPCRs having activity in cell based tastant assays, obtainable by (a) improving the coding sequence of the human sweet taste receptors (T1Rs) and/or (b) cloning them in a multicistronic expression unit for coordinated expression and simultaneous selection for the generation of stable eucaryotic cell systems; and/or (c) tagging one or both GPCRs in the hetero-dimeric expression complex with G-proteins or G-Protein chimeras to enable an improved fluorescence read out for the identification of GPCR modulators, preferably sweet taste modulating compounds.

[0094] The receptor sequences applied for the here presented invention are furthermore examplified in Example 1. By adding a prefix the optimized receptors have been termed shT1R2 and shT1R3. The respective amino acid sequences are depicted in SEQ ID NOs 1 and 2, the respective nucleic acid sequences in SEQ ID NOs 3 and 4.

[0095] A further means of the present invention to overcome the instability of co-expressed GPCRs, preferably taste receptors T1R2 and T1R3 in stable cell line development is the construction of multicistronic transcription units. The method for obtaining a multicistronic expression vector for the simultaneous expression of taste receptors and a selection marker of the invention is further examplified in Example 2. As stated above the development of T1R2/T1R3 expressing stable cell lines e.g. Hek293, CHO, Hela for their use in cell based assay systems is hampered by upcoming instabilities within the ongoing eucaryotic cell cultivation process and passaging numbers. For the generation of stable cell lines in the art most often expression vectors are used were the selection marker, conferring resistance to e.g. neomycin, hygromycin, zeocin, blasticidin or puromycin, is in fact coded on the same plasmid vector under the control of a independent promoter.

[0096] This situation can be avoided if the receptors and the selection marker are encoded in a multicistronic transcription unit.

[0097] However, so far multicistronic expression vectors have been described only for the use of inducible protein expression e.g. for gene therapy applications (Fussenegger et al., 1998; Moser et al., 2000).

[0098] For the present invention the two taste receptors shT1R2 and shT1R3 have been cloned in the first and second position of a tricistronic expression unit, whereas the deaminase gene conferring resistance to the selection marker blasticidin was cloned into the third position of the expression unit. The tricistronic unit is under the control of the strong human elongation factor 1 alpha promoter and the genes in the second and third position are preceded by an IRES element to facilitate their translation initiation in the resulting mRNA.

[0099] So far an eight kb multicistronic expression system for the stable functional expression of membrane located GPCRs has not been described. Usually these multicistronic systems--already in expressions of much smaller gene products like fluorescent proteins--are characterized by a non-stoichiometric and decreasing expression with increased distance from the promoter. Since the taste GPCRs significantly exceed the size of GFP or genes so far used in tri- or quadrocistronic expression systems; and the fact that the GPCRs have to be co-expressed to form a heterodimeric protein complex to function as sweetener responsive receptors it was an unforeseeable and surprising result of the intense investigations leading to the present invention that only the use of multicistronic vectors, i.e. tri- or tetracistronic vectors let to satisfying results in establishing the screening method of the present invention.

[0100] In a further embodiment, this invention relates to methods using the before mentioned multicistronic expression vector to create stable cell lines suited to express T1R polypeptides to construct a cell based screening tool for the search of novel compounds in the field of sweet taste modulators. Preferably, the cells comprise a functional G protein, e.g., G-alpha-15, G-alpha-16 or chimeric G protein like the ones with altered c-terminus previously identified:

a) substitution of the last five amino acids of G-alpha 15: [0101] G-alpha.sub.--15 (EINLL), replaced with EYNLV (G-alpha q and G-alpha 11), EFNLV (G-alpha 14), QYELL (G-alpha s and G-alpha olf), DCGLF (G-alpha i1, G-alpha i2, G-alpha t1, G-alpha t2, and G-alpha gust), ECGLY (G-alpha i3), GCGLY (G-alpha o1 and G-alpha o2), YIGLC (G-alpha z), DIMLQ (G-alpha 12), QLMLQ (G-alpha 13) or G-alpha 16 replaced with ECGLY (G-alpha 16 i3) b) substitution of the last 44 carboxy-terminal amino acids of G-alpha-16: [0102] G-alpha 16gust44 (44 amino acids of the G-protein gustducin) [0103] G-alpha 16z44 (44 amino acids of G-alpha-z); [0104] G-alpha 16C44i2 (44 amino acids of the G-alpha-12); [0105] G-alpha 16C44i3 (44 amino acids of the G-alpha-13); or another G protein that is capable of coupling the chimeric receptor to an intracellular signaling pathway or to a signaling protein such as phospholipase C. (Li et al., 2002; Offermanns, 2003; Offermanns and Simon, 1995; Ueda et al., 2003). For this purpose HEK 293 cells comprising a functional G protein have been transfected with the vector pTrix-Eb_R2R3, cultured in the presence of blasticidine, and stable cell lines have been selected. As shown in Example 3 such cells have been found to exhibit responses to taste stimuli and in addition these cell lines have--compared to those derived with monocistronic T1R expression vectors--a pronounced stable expression of the T1R polypeptides. A stable cell line based on the expression vector pTrix-Eb_R2R3 (see example 3) shows calcium responses to several artificial sweeteners and a natural sweet protein. These responses can still be measured after more than 50 passages of the clone under selection with blasticidin and G418. Such results could not be achieved with monocistronic or bicistronic expressions. RT-PCR results of monocistronic clones revealed the loss of both receptors with the initial 5 to 10 passages under selection conditions. The stable cell line HEK_Ga15#17R2R3b #8 also shows dosis-dependent calcium responses for the above shown sweet tastants which correlated with physiological taste responses.

[0106] A further unexpected effect is that these multicistronic stable taste receptor expressing cell lines are well suited for natural compound screening using complex microbial extract preparations. For the isolation and preparation of such complex screening samples standard microbial methods can be applied which encompass the cultivation of microorganisms. Bioactive metabolites of interest from such cultivations may include intracellular molecules as well as those secreted into the cultivation media. Depending on the number of samples to be processed, a choice can generally be made from different options: (i) heating of the whole broth; (ii) filtration of broth supernatant through a high molecular weight exclusion filter with subsequent freeze-drying of the filtrate; (iii) extraction of either the whole broth (ultrasonication, french press, cell disaruption bombs or similar devices) or the broth supernantant with organic solvents of varying polarity (e.g. chloroform, aceton, methanol or ethyl acetat) followed by evaporation of the extracts to dryness. (iv) Mixing the whole broth or the broth supernantant with resins (e.g. AMBERLITE.RTM., XAD-2, XAD-4, XAD-9 or similar) with subsequent solid phase extraction with organic solvents (e.g. acetone, acetic acid, butanol, ethanol, ethyl acetate, methanol, polyglycols or similar) either fractionated by applying increasing solvent concentrations or as whole batch by applying the pure solvent without fractionation. Solid phase extracts can be used directly in suited assay systems or further evaporated to dryness and resolved under standardized conditions for subsequent testing, e.g. the activation of taste receptors in cell based assays. Activation of T1R receptors in such cells can be detected using any standard methods, such as by detecting changes in intracellular calcium by detecting Fluo-4 dependent fluorescence in the cell. Such an assay is the basis of the experimental findings presented in this application.

[0107] In a similar manner plant extracts well known to the skilled man may be used as well.

[0108] In FIG. 3 the activity of a stable cell line derived with the multicistronic expression plasmid pTrix-Eb-R2R3 in a G-alpha 15 G-Protein background is depicted. The data of FIG. 3 document that these cell line amongst several other isolated cell lines, shows T1R2/T1R3 dependent activity in cell based assays towards several tastants like cyclamate, D-phenylalanine, saccharin, acesulfam K, aspartam and thaumatin.

[0109] In a further embodiment, this invention relates to the use of T1R-G-protein fusion genes for the development of T1R-dependent cell based assays. It has been shown that GPCRs can be fused in frame with different polypeptides without loosing their functional activity (Milligan et al., 2003). From the present data it cannot be foreseen that a fusion of a functional G-Protein to any GPCR leads to an improved activity compared to the wild type unfused situation. However it is scheduled to develop T1R2- and/or T1R3-fusion proteins in which the T1R-GPCR is fused to a functional G-Protein which is able to connect the T1R dependent signaling to the calcium inducing pathways used for T1R dependent activity measurement in cell based assays as exemplified in Example 3. Preferably, the T1R-fusion proteins comprise a functional G protein, e.g., Galpha-15 or chimeric G protein like the ones previously identified, or another G protein that is capable of coupling the chimeric receptor to an intracellular signaling pathway or to a signaling protein such as phospholipase C. (Offermanns, 2003; Offermanns and Simon, 1995; Ueda et al., 2003) For the development of such cell based assays the fusion proteins will be subcloned into the multicistronic expression vector backbone leading to expression vector of the type pTrix-Eb-R2Gq-R3 depicted in FIG. 4.

[0110] The figures show:

[0111] FIG. 1: Bioinformatic alignment with Clustal X software: Different GPCR nucleotide sequences of the T1R class are presented in an alignment analysis. Human wild type cDNA sequences are depicted as wt_hT1Rs; the theoretically most optimized cDNA sequences after multi parameter optimization are depicted as opt_hT1Rs; partly optimized cDNAs used for stable cell line development are termed sh_T1Rs. The nucleotide alignments are presented as phylogenetic trees in which wt_T1R1 was defined as the outgroup. The underlying distance matrix is shown below.

[0112] FIG. 2: Multicistronic eucaryotic expression vector pTrix-Eb-R2R3: The expression of the taste receptor genes shT1R2, shT1R3 and blasticidin S deaminase (bsd) gene are under the control of the human elongation factor 1 alpha promoter (PEF1a). To confer multicistronic expression on the translational level two internal ribosomal entry sites (Cite I and Cite II) have been inserted. The multicistronic unit is terminated by a simian virus 40 polyadenylation site. The prokaryotic origin of replication (ori) and the kanamycin resistance gene serve for the propagation, amplification and selection of the plasmid vector in E. coli.

[0113] FIG. 3: The stable HEK_Ga15#17R2R3b #8 was selected with blasticidin and G418 after transfection of the stable HEK_Ga15#17 (G418) with pTriX_Eb_R2R3. This tricistronic expression was necessary for the selection and retrieval of stable clones. Clone #8 shows calcium responses to several artificial sweeteners and a natural sweet protein.

[0114] FIG. 4: Multicistronic eucaryotic expression vector pTrix-Eb-R2R3: The expression of the taste receptor genes shT1R2-Gq, shT1R3 and blasticidin S deaminase (bsd) gene are under the control of the human elongation factor 1 alpha promoter (PEF1a). To confer multicistronic expression on the translational level two internal ribosomal entry sites (Cite I and Cite II) have been inserted. The multicistronic unit is terminated by a simian virus 40 polyadenylation site. The prokaryotic origin of replication (ori) and the kanamycin resistance gene serve for the propagation, amplification and selection of the plasmid vector in E. coli.

Experimental Materials and Methods

Cell Culture

[0115] Transient transfection/selection of stable HEK293 cells--Transient and stable transfections can be performed with lipid complexes like calcium phosphate precipitation, Lipofectamine/PLUS reagent (Invitrogen), Lipofectamine 2000 (Invitrogen) or MIRUS TransIT293 (Mirus Bio Corporation) according to the manuals. Electroporation can also be a method of choice for stable transfection of eukaryotic cells.

[0116] The cells are seeded in 6-well plates at a density of 4.times.105 cells/well. HEK293 cells are transfected with linearised plasmids for stable expression of the genes of interest. After 24 hours, the selection with selecting reagents like zeocin, hygromycin, neomycin or blasticidin starts. About 50 .mu.l to 300 .mu.l trypsinized transfected cells from a 6-well are seeded in a 100 mm dish and the necessary antibiotic is added in an appropriate concentration. Cells are cultivated until clones are visible on the 100 mm cell culture plate. These clones are selected for further cultivation and calcium imaging. It takes about four to eight weeks to select cell clones which stably express the genes of interest.

Calcium Imaging

[0117] Fluo-4 AM assay with stable HEK293 cells--Stable cells are maintained in DMEM high-glucose medium. (Invitrogen) supplemented with 10% fetal bovine serum (Biochrom) and 4 mM L-glutamine (Invitrogen). Cells for calcium imaging are maintained in DMEM low-glucose medium supplemented with 10% FBS and 1.times. Glutamax-1 (Invitrogen) for 48 hours before seeding. These stable cells are trypsinized after 48 hours (either with Trypsin-EDTA, Accutase or TrypLE) and seeded onto poly-D-lysine coated 96-well assay plates (Corning) at a density of 45,000 cells/well in DMEM low-glucose medium supplemented with 10% FBS and 1.times. Glutamax-1.

[0118] After 24 hours, the cells were loaded in 100 .mu.l medium with additional 100 .mu.l of 4 .mu.M Fluo-4 (calcium sensing dye, 2 .mu.M end concentration; Molecular Probes) in Krebs-HEPES (KH)-buffer for 1 hour. The loading reagent is then replaced by 80 .mu.l KH-buffer per well. The Krebs-HEPES-buffer is a physiological saline solution including 1.2 mM CaCl2, 4.2 mM NaHCO3 and 10 mM HEPES.

[0119] The dye-loaded stable cells in plates were placed into a fluorescence microtiter plate reader to monitor fluorescence (excitation 488 nm, emission 520 nm) change after the addition of 20 .mu.l KH-buffer supplemented with 5.times. tastants. For each trace, tastant was added 11.5 seconds after the start of the scan and mixed two times with the buffer, scanning continued for an additional 32 seconds, and data were collected every second.

[0120] Data analysis/Data recording--Calcium mobilization was quantified as the change of peak fluorescence (.DELTA.F) over the baseline level (F). Data were expressed as the mean S.E. of the .DELTA.F/F value of replicated independent samples. The analysis was done with the software of the microtiter plate reader.

EXAMPLES

[0121] The following examples are provided to illustrate preferred embodiments and are intended to be illustrative and not limitative of the scope of the invention. In the DNA sequences presented herein, the one letter codes N or n refers to any of the of the four common nucleotide bases, A, T, C, or G. In the protein sequences presented herein, the one-letter code X or Xaa refers to any of the twenty common amino acid residues.

Example 1

Synthesis and Design of Synthetic Intronless hT1R2 and hT1R3 cDNAs

[0122] The nucleotide sequence of the human receptors hT1R2 and hT1R3 are based on their wild type coding DNAs and have been optimized according to a multiparameter analysis considering optimal codon usage, putative cryptic splice sites, putative repeated sequences as well as AT-rich or GC-rich sequence stretches. Gene optimization often has favorable effects on enhanced mRNA stability, translational efficiency and reduced RNA secondary structure to prevent transcriptional pausing or premature termination of transcriptional elongation.

[0123] Up to now the binding sites for only a few ligands of the sweet taste heterodimeric receptor T1R2/T1R3 are known. Hence, the encoded wild-type amino acid sequences of the receptors were left unchanged in this multiparameter optimization, to retain the receptors binding qualities.

[0124] Synthesis and construction of the receptor cDNAs was done via the assembly of synthetic oligonucleotides and subsequent cloning into a standard pUC-18 plasmid vector for further amplification. Alternatively, these nucleic acids can be cloned in vitro by well-known cloning techniques., (Ausubel et al., 1998; Pachuk et al., 2000; Sambrook et al., 1989; Stemmer et al., 1995). Double-stranded DNA fragments may then be obtained either by synthesizing the complementary strand and annealing the strands together under appropriate conditions, or by adding the complementary strand using DNA polymerase with an appropriate primer sequence.

[0125] Due to the synthetic generation of the cDNAs the receptors of the present invention have been described with the prefix "s" and termed shT1R2 and shT1R3. The nucleic acid and amino acid sequences for the above mentioned T1R cloned sequences as well as other full-length and partial T1R sequences are set forth in the sequence protocol:

Example 2

Multicistronic Vectors for the Expression of Taste Receptors

[0126] For the construction of a multicistronic expression unit the taste receptor sequences displayed in Example 1 have been used. As shown in FIG. 2 the tricistronic expression unit is under the control of the human elongation factor 1 alpha promoter. Using standard cloning techniques the cDNA for the receptors shT1R2 and shT1R3 and the cDNA for the blasticidin S deaminase gene have been cloned. To enable the translation initiation of each gene of this tricistronic unit two EMC-virus derived internal ribosomal entry sites (IRES--also termed Cap-independent translation enhancer (CITE)) have been inserted. (Jackson et al., 1990; Jang et al., 1988) The tricistronic expression unit is terminated by a simian virus 40 polyadenylation signal sequence. This composition permits the simultaneous expression of all three genes under the control of only one promoter. In contrast to monocistronic transcription units, which integrate independently from each other into different chromosomal locations during the process of stable cell line development, the tricistronic transcription unit integrates all containing genes in one and the same chromosomal locus. Due to the alignment of the genes, the blasticidin S deaminase gene is only transcribed in case a full length transcription takes place. Moreover the polarity of multicistronic transcription units (Moser et al., 2000) leads probably to a balanced stoichiometry of the receptor genes and their expression rates in the range of 1:0.7 up to 1:1 for the first two positions whereas the blasticidin S deaminase gene compared to the receptor genes in the third position is expressed to a lesser extend. Assuming that for the functional heterodimeric receptor shT1R2/shT1R3 a 1:1 stoichiometry is needed the lesser polarity effects for the receptor genes promote the desired stoichiometry whereas the reduced expression of the deaminase promotes an integration locus with enhanced transcriptional activity.

Example 3

Detection of T1R2/T1R3 Dependent Activity

[0127] In wild type taste cells--e.g. in the human taste bud--signal transduction is presumably transduced by the G-proteins gustducin and/or by G-Proteins of the Galpha-i type. Encountering sweet ligands the heterodimeric taste receptor T1R2/T1R3 reacts with induction of second messenger molecules; either induction of the cAMP level in response to most sugars or induction of the calcium level in response to most artificial sweeteners. (Margolskee, 2002)

[0128] To analyze the function and activity of the heterodimeric T1R2/T1R3 taste receptor a calcium dependent cell based assay has been utilized. Briefly, synthetic T1R type taste receptors (as shown in Example 1) have been transfected with the plasmid vector pTrix-Eb-R2R3 (see Example 2) in a HEK293 cell line stably expressing the mouse G-alpha-15 G-protein. Selection of T1R2/T1R3 expressing cells has been performed by culturing the transfected cells in the presence of blasticidine.

[0129] For measurement of T1R2/T1R3 taste receptor dependent activity HEK293 cells stably expressing G-alpha-15, shT1R2 and shT1R3 were 4.times.104 seeded in 96-well plates and labeled with the calcium sensitive fluorescence dye Fluo4-AM (2 .mu.M) in DMEM culture medium for one hour at 37.degree. C. For the measurement in a fluorescence plate reader the medium was exchanged for KH-buffer and incubated for another 15 minutes at 37.degree. C. Fluorescence measurement of the labeled cells was conducted in a Novostar fluorescence plate reader (BMG, Offenburg, Germany). Response to different tastants as depicted in FIG. 3 was recorded as Fluo4-AM fluorescence increase initiated through the T1R2/T1R3 dependent increase of the second messenger calcium. After obtaining calcium signals for each sample, calcium mobilization in response to tastants was quantified as the relative change (peak fluorescence F1--baseline fluorescence F0 level, denoted as dF) from its own baseline fluorescence level (denoted as F0). Though rel. RFU is dF/F0. Peak fluorescence intensity occurred about 20-30 sec after addition of tastants. The data shown were obtained from two independent experiments and done in triplicates.

REFERENCES

[0130] Ausubel, F. M., Brent, R., Kingston, R. E., Moore, D. D., Seidman, J. G., Smith, J. A., and Struhl, K. (1998). /Current protocols in molecular biolong. //V.B. Chanada, series ed. //New York: Wiley & Sons./ [0131] Bachmanov, A. A., Tordoff, M. G., and Beauchamp, G. K. (2001). Sweetener preference of C57BL/6ByJ and 129P3/J mice. /Chem Senses/ 26, 905-13. [0132] Bai, M., Trivedi, S., and Brown, E. M. (1998). Dimerization of the extracellular calcium-sensing receptor (CaR) on the cell surface of CaR-transfected HEK293 cells. /J Biol Chem/ 273, 23605-10. [0133] Bockaert, J., and Pin, J. P. (1999). Molecular tinkering of G protein-coupled receptors: an evolutionary success. /Embo J/ 18, 1723-9. [0134] Buck, L., and Axel, R. (1991). A novel multigene family may encode odorant receptors: a molecular basis for odor recognition. /Cell/ 65, 175-87. [0135] Eglen, R. (2005). An Overview of High Throughput Screening at G Protein Coupled Receptors. /Frontiers in Drug Design & Discovery/ 1, 97-111. [0136] Filmore, D. (2004). It's a GPCR world. /Modern Drug Discovery/, 24-28. [0137] Firestein, S. (2001). How the olfactory system makes sense of scents. /Nature/ 413, 211-8. [0138] Fussenegger, M., Mazur, X., and Bailey, J. E. (1998). pTRIDENT, a novel vector family for tricistronic gene expression in mammalian cells. /Biotechnol Bioeng/ 57, 1-10. [0139] Fux, C., Langer, D., Kelm, J. M., Weber, W., and Fussenegger, M. (2004). New-generation multicistronic expression platform: pTRIDENT vectors containing size-optimized IRES elements enable homing endonuclease-based cistron swapping into lentiviral expression vectors. /Biotechnol Bioeng/ 86, 174-87. [0140] George, S. R., O'Dowd, B. F., and Lee, S. P. (2002). G-protein-coupled receptor oligomerization and its potential for drug discovery. /Nat Rev Drug Discov/ 1, 808-20. [0141] Grenby, T. H. (1996): Advances in Sweeteners, 1.sup.st edition, Blackie Academic & Professional, London. [0142] Hartenbach, S., and Fussenegger, M. (2005). Autoregulated, bidirectional and multicistronic gas-inducible mammalian as well as lentiviral expression vectors. /J Biotechnol/. [0143] Hellen, C. U., and Sarnow, P. (2001). Internal ribosome entry sites in eukaryotic mRNA molecules. /Genes Dev/ 15, 1593-612. [0144] Hoon, M. A., Adler, E., Lindemeier, J., Battey, J. F., Ryba, N. J., and Zuker, C. S. (1999). Putative mammalian taste receptors: a class of taste-specific GPCRs with distinct topographic selectivity. /Cell/ 96, 541-51. [0145] Hoon, M. A., and Ryba, N. J. (1997). Analysis and comparison of partial sequences of clones from a taste-bud-enriched cDNA library. /J Dent Res/ 76, 831-8. [0146] Jackson, R. J., Howell, M. T., and Kaminski, A. (1990). The novel mechanism of initiation of picornavirus RNA translation. /Trends Biochem Sci/ 15, 477-83. [0147] Jang, S. K., Krausslich, H. G., Nicklin, M. J., Duke, G. M., Palmenberg, A. C., and Wimmer, E. (1988). A segment of the 5' nontranslated region of encephalomyocarditis virus RNA directs internal entry of ribosomes during in vitro translation. /J Virol/ 62, 2636-43. [0148] Kaupmann, K., Malitschek, B., Schuler, V., Heid, J., Froestl, W., Beck, P., Mosbacher, J., Bischoff, S., Kulik, A., Shigemoto, R., Karschin, A., and Bettler, B. (1998). GABA(B)-receptor subtypes assemble into functional heteromeric complexes. /Nature/ 396, 683-7. [0149] Kinnamon, S. C., and Cummings, T. A. (1992). Chemosensory transduction mechanisms in taste. /Annu Rev Physiol/ 54, 715-31. [0150] Kinnamon, S. C., and Margolskee, R. F. (1996). Mechanisms of taste transduction. /Curr Opin Neurobiol/ 6, 506-13. [0151] Kitagawa, M., Kusakabe, Y., Miura, H., Ninomiya, Y., and Hino, A. (2001). Molecular genetic identification of a candidate receptor gene for sweet taste. /Biochem Biophys Res Commun/ 283, 236-42. [0152] Kramer, B. P., Weber, W., and Fussenegger, M. (2003). Artificial regulatory networks and cascades for discrete multilevel transgene control in mammalian cells. /Biotechnol Bioeng/ 83, 810-20. [0153] Kunishima, N., Shimada, Y., Tsuji, Y., Sato, T., Yamamoto, M., Kumasaka, T., Nakanishi, S., Jingami, H., and Morikawa, K. (2000). Structural basis of glutamate recognition by a dimeric metabotropic glutamate receptor. /Nature/ 407, 971-7. [0154] Leatherhead Food RA (2000). Ingredients Handbook Sweeteners, 2.sup.nd edition, Leatherhead Publishing, Leatherhead, Surrey. [0155] Li, X., Inoue, M., Reed, D. R., Huque, T., Puchalski, R. B., Tordoff, M. G., Ninomiya, Y., Beauchamp, G. K., and Bachmanov, A. A. (2001). High-resolution genetic mapping of the saccharin preference locus (Sac) and the putative sweet taste receptor (T1R1) gene (Gpr70) to mouse distal Chromosome 4. /Mamm Genome/ 12, 13-6. [0156] Li, X., Staszewski, L., Xu, H., Durick, K., Zoller, M., and Adler, E. (2002). Human receptors for sweet and umami taste. /Proc Natl Acad Sci USA/ 99, 4692-6. [0157] Lindemann, B. (1996a). Chemoreception: tasting the sweet and the bitter. /Curr Biol/ 6, 1234-7. [0158] Lindemann, B. (1996b). Taste reception. /Physiol Rev/ 76, 718-66. [0159] Lindemann, B. (2001). Receptors and transduction in taste. /Nature/ 413, 219-25. [0160] Lush, I. E. (1989). The genetics of tasting in mice. VI. Saccharin, acesulfame, dulcin and sucrose. /Genet Res/ 53, 95-9. [0161] Lush, I. E., Hornigold, N., King, P., and Stoye, J. P. (1995). The genetics of tasting in mice. VII. Glycine revisited, and the chromosomal location of Sac and Soa. /Genet Res/ 66, 167-74. [0162] Maeda, T., Imanishi, Y., and Palczewski, K. (2003). Rhodopsin phosphorylation: 30 years later. /Prog Retin Eye Res/ 22, 417-34. [0163] Margolskee, R. F. (2002). Molecular mechanisms of bitter and sweet taste transduction. /J Biol Chem/ 277, 1-4. [0164] Max, M., Shanker, Y. G., Huang, L., Rong, M., Liu, Z., Campagne, F., Weinstein, H., Damak, S., and Margolskee, R. F. (2001). Tas1r3, encoding a new candidate taste receptor, is allelic to the sweet responsiveness locus Sac. /Nat Genet/ 28, 58-63. [0165] McLaughlin, S. K., McKinnon, P. J., and Margolskee, R. F. (1992). Gustducin is a taste-cell-specific G protein closely related to the transducins. /Nature/ 357, 563-9. [0166] Meyerhof, W., Behrens, M., Brockhoff, A., Bufe, B., and Kuhn, C. (2005). Human bitter taste perception. /Chem Senses/ 30 Suppl 1, i14-i15. Milligan, G. (2003). High-content assays for ligand regulation of G-protein-coupled receptors. /Drug Discov Today/ 8, 579-85. [0167] Milligan, G., Ramsay, D., Pascal, G., and Carrillo, J. J. (2003). GPCR dimerisation. /Life Sci/ 74, 181-8. [0168] Montmayeur, J. P., Liberles, S. D., Matsunami, H., and Buck, L. B. (2001). A candidate taste receptor gene near a sweet taste locus. /Nat Neurosci/ 4, 492-8. [0169] Moser, S., Schlatter, S., Fux, C., Rimann, M., Bailey, J. E., and Fussenegger, M. (2000). An update of pTRIDENT multicistronic expression vectors: pTRIDENTs containing novel streptogramin-responsive promoters. /Biotechnol Prog/ 16, 724-35. [0170] Nelson, G., Chandrashekar, J., Hoon, M. A., Feng, L., Zhao, G., Ryba, N. J., and Zuker, C. S. (2002). An amino-acid taste receptor. /Nature/ 416, 199-202. [0171] Nelson, G., Hoon, M. A., Chandrashekar, J., Zhang, Y., Ryba, N. J., and Zuker, C. S. (2001). Mammalian sweet taste receptors. /Cell/ 106, 381-90. [0172] O'Brien Nabors, L. (2001). Alternative Sweeteners, 3.sup.rd edition, Marcel Dekker, Inc., New York, Basel. [0173] Offermanns, S. (2003). G-proteins as transducers in transmembrane signalling. /Prog Biophys Mol Biol/ 83, 101-30. [0174] Offermanns, S., and Simon, M. I. (1995). G alpha 15 and G alpha 16 couple a wide variety of receptors to phospholipase C. /J Biol Chem/ 270, 15175-80. [0175] Pachuk, C. J., Samuel, M., Zurawski, J. A., Snyder, L., Phillips, P., and Satishchandran, C. (2000). Chain reaction cloning: a one-step method for directional ligation of multiple DNA fragments. /Gene/ 243, 19-25. [0176] Pierce, K. L., Premont, R. T., and Lefkowitz, R. J. (2002). Seven-transmembrane receptors. /Nat Rev Mol Cell Biol/ 3, 639-50. [0177] Pin, J. P., Galvez, T., and Prezeau, L. (2003). Evolution, structure, and activation mechanism of family 3/C G-protein-coupled receptors. /Pharmacol Ther/ 98, 325-54. [0178] Sainz, E., Korley, J. N., Battey, J. F., and Sullivan, S. L. (2001). Identification of a novel member of the T1R family of putative taste receptors. /J Neurochem/ 77, 896-903. [0179] Salahpour, A., Angers, S., and Bouvier, M. (2000). Functional significance of oligomerization of G-protein-coupled receptors. /Trends Endocrinol Metab/ 11, 163-8. [0180] Sambrook, J., Fritsch, E., and Maniatis, T. (1989). Molecular cloning. A Laboratory Manual. /In: second ed. //Cold Spring Harbor Laboratory Press. Cold Spring Harbor. N.Y./ [0181] Stemmer, W. P., Crameri, A., Ha, K. D., Brennan, T. M., and Heyneker, H. L. (1995). Single-step assembly of a gene and entire plasmid from large numbers of oligodeoxyribonucleotides. /Gene/ 164, 49-53. [0182] Sugita, M., and Shiba, Y. (2005). Genetic tracing shows segregation of taste neuronal circuitries for bitter and sweet. /Science/ 309, 781-5. [0183] Ueda, T., Ugawa, S., Yamamura, H., Imaizumi, Y., and Shimada, S. (2003). Functional interaction between T2R taste receptors and G-protein alpha subunits expressed in taste receptor cells. /J Neurosci/ 23, 7376-80. [0184] Von Rymon Lipinski, G.-W., Schiweck, H. (1991). Handbuch Su.beta.ungsmittel-Eigenschaften und Anwendung, Behr's Verlag, Hamburg. [0185] Weber, W., Malphettes, L., de Jesus, M., Schoenmakers, R., El-Baba, M. D., Spielmann, M., Keller, B., Weber, C. C., van de Wetering, P., Aubel, D., Wurm, F. M., and Fussenegger, M. (2005). Engineered Streptomyces quorum-sensing components enable inducible siRNA-mediated translation control in mammalian cells and adjustable transcription control in mice. /J Gene Med/ 7, 518-25. [0186] White, J. H., Wise, A., Main, M. J., Green, A., Fraser, N. J., Disney, G. H., Barnes, A. A., Emson, P., Foord, S. M., and Marshall, F. H. (1998). Heterodimerization is required for the formation of a functional GABA(B) receptor. /Nature/ 396, 679-82. [0187] Wong, G. T., Gannon, K. S., and Margolskee, R. F. (1996). Transduction of bitter and sweet taste by gustducin. /Nature/ 381, 796-800. [0188] Zhao, F. L., Lu, S. G., and Herness, S. (2002). Dual actions of caffeine on voltage-dependent currents and intracellular calcium in taste receptor cells. /Am J Physiol Regul Integr Comp Physiol/ 283, R115-29.

Sequence CWU 1

1

41839PRTHomo sapiens 1Met Gly Pro Arg Ala Lys Thr Ile Cys Ser Leu Phe Phe Leu Leu Trp1 5 10 15Val Leu Ala Glu Pro Ala Glu Asn Ser Asp Phe Tyr Leu Pro Gly Asp20 25 30Tyr Leu Leu Gly Gly Leu Phe Ser Leu His Ala Asn Met Lys Gly Ile35 40 45Val His Leu Asn Phe Leu Gln Val Pro Met Cys Lys Glu Tyr Glu Val50 55 60Lys Val Ile Gly Tyr Asn Leu Met Gln Ala Met Arg Phe Ala Val Glu65 70 75 80Glu Ile Asn Asn Asp Ser Ser Leu Leu Pro Gly Val Leu Leu Gly Tyr85 90 95Glu Ile Val Asp Val Cys Tyr Ile Ser Asn Asn Val Gln Pro Val Leu100 105 110Tyr Phe Leu Ala His Glu Asp Asn Leu Leu Pro Ile Gln Glu Asp Tyr115 120 125Ser Asn Tyr Ile Ser Arg Val Val Ala Val Ile Gly Pro Asp Asn Ser130 135 140Glu Ser Val Met Thr Val Ala Asn Phe Leu Ser Leu Phe Leu Leu Pro145 150 155 160Gln Ile Thr Tyr Ser Ala Ile Ser Asp Glu Leu Arg Asp Lys Val Arg165 170 175Phe Pro Ala Leu Leu Arg Thr Thr Pro Ser Ala Asp His His Val Glu180 185 190Ala Met Val Gln Leu Met Leu His Phe Arg Trp Asn Trp Ile Ile Val195 200 205Leu Val Ser Ser Asp Thr Tyr Gly Arg Asp Asn Gly Gln Leu Leu Gly210 215 220Glu Arg Val Ala Arg Arg Asp Ile Cys Ile Ala Phe Gln Glu Thr Leu225 230 235 240Pro Thr Leu Gln Pro Asn Gln Asn Met Thr Ser Glu Glu Arg Gln Arg245 250 255Leu Val Thr Ile Val Asp Lys Leu Gln Gln Ser Thr Ala Arg Val Val260 265 270Val Val Phe Ser Pro Asp Leu Thr Leu Tyr His Phe Phe Asn Glu Val275 280 285Leu Arg Gln Asn Phe Thr Gly Ala Val Trp Ile Ala Ser Glu Ser Trp290 295 300Ala Ile Asp Pro Val Leu His Asn Leu Thr Glu Leu Gly His Leu Gly305 310 315 320Thr Phe Leu Gly Ile Thr Ile Gln Ser Val Pro Ile Pro Gly Phe Ser325 330 335Glu Phe Arg Glu Trp Gly Pro Gln Ala Gly Pro Pro Pro Leu Ser Arg340 345 350Thr Ser Gln Ser Tyr Thr Cys Asn Gln Glu Cys Asp Asn Cys Leu Asn355 360 365Ala Thr Leu Ser Phe Asn Thr Ile Leu Arg Leu Ser Gly Glu Arg Val370 375 380Val Tyr Ser Val Tyr Ser Ala Val Tyr Ala Val Ala His Ala Leu His385 390 395 400Ser Leu Leu Gly Cys Asp Lys Ser Thr Cys Thr Lys Arg Val Val Tyr405 410 415Pro Trp Gln Leu Leu Glu Glu Ile Trp Lys Val Asn Phe Thr Leu Leu420 425 430Asp His Gln Ile Phe Phe Asp Pro Gln Gly Asp Val Ala Leu His Leu435 440 445Glu Ile Val Gln Trp Gln Trp Asp Arg Ser Gln Asn Pro Phe Gln Ser450 455 460Val Ala Ser Tyr Tyr Pro Leu Gln Arg Gln Leu Lys Asn Ile Gln Asp465 470 475 480Ile Ser Trp His Thr Val Asn Asn Thr Ile Pro Met Ser Met Cys Ser485 490 495Lys Arg Cys Gln Ser Gly Gln Lys Lys Lys Pro Val Gly Ile His Val500 505 510Cys Cys Phe Glu Cys Ile Asp Cys Leu Pro Gly Thr Phe Leu Asn His515 520 525Thr Glu Asp Glu Tyr Glu Cys Gln Ala Cys Pro Asn Asn Glu Trp Ser530 535 540Tyr Gln Ser Glu Thr Ser Cys Phe Lys Arg Gln Leu Val Phe Leu Glu545 550 555 560Trp His Glu Ala Pro Thr Ile Ala Val Ala Leu Leu Ala Ala Leu Gly565 570 575Phe Leu Ser Thr Leu Ala Ile Leu Val Ile Phe Trp Arg His Phe Gln580 585 590Thr Pro Ile Val Arg Ser Ala Gly Gly Pro Met Cys Phe Leu Met Leu595 600 605Thr Leu Leu Leu Val Ala Tyr Met Val Val Pro Val Tyr Val Gly Pro610 615 620Pro Lys Val Ser Thr Cys Leu Cys Arg Gln Ala Leu Phe Pro Leu Cys625 630 635 640Phe Thr Ile Cys Ile Ser Cys Ile Ala Val Arg Ser Phe Gln Ile Val645 650 655Cys Ala Phe Lys Met Ala Ser Arg Phe Pro Arg Ala Tyr Ser Tyr Trp660 665 670Val Arg Tyr Gln Gly Pro Tyr Val Ser Met Ala Phe Ile Thr Val Leu675 680 685Lys Met Val Ile Val Val Ile Gly Met Leu Ala Thr Gly Leu Ser Pro690 695 700Thr Thr Arg Thr Asp Pro Asp Asp Pro Lys Ile Thr Ile Val Ser Cys705 710 715 720Asn Pro Asn Tyr Arg Asn Ser Leu Leu Phe Asn Thr Ser Leu Asp Leu725 730 735Leu Leu Ser Val Val Gly Phe Ser Phe Ala Tyr Met Gly Lys Glu Leu740 745 750Pro Thr Asn Tyr Asn Glu Ala Lys Phe Ile Thr Leu Ser Met Thr Phe755 760 765Tyr Phe Thr Ser Ser Val Ser Leu Cys Thr Phe Met Ser Ala Tyr Ser770 775 780Gly Val Leu Val Thr Ile Val Asp Leu Leu Val Thr Val Leu Asn Leu785 790 795 800Leu Ala Ile Ser Leu Gly Tyr Phe Gly Pro Lys Cys Tyr Met Ile Leu805 810 815Phe Tyr Pro Glu Arg Asn Thr Pro Ala Tyr Phe Asn Ser Met Ile Gln820 825 830Gly Tyr Thr Met Arg Arg Asp8352852PRTHomo sapiens 2Met Leu Gly Pro Ala Val Leu Gly Leu Ser Leu Trp Ala Leu Leu His1 5 10 15Pro Gly Thr Gly Ala Pro Leu Cys Leu Ser Gln Gln Leu Arg Met Lys20 25 30Gly Asp Tyr Val Leu Gly Gly Leu Phe Pro Leu Gly Glu Ala Glu Glu35 40 45Ala Gly Leu Arg Ser Arg Thr Arg Pro Ser Ser Pro Val Cys Thr Arg50 55 60Phe Ser Ser Asn Gly Leu Leu Trp Ala Leu Ala Met Lys Met Ala Val65 70 75 80Glu Glu Ile Asn Asn Lys Ser Asp Leu Leu Pro Gly Leu Arg Leu Gly85 90 95Tyr Asp Leu Phe Asp Thr Cys Ser Glu Pro Val Val Ala Met Lys Pro100 105 110Ser Leu Met Phe Leu Ala Lys Ala Gly Ser Arg Asp Ile Ala Ala Tyr115 120 125Cys Asn Tyr Thr Gln Tyr Gln Pro Arg Val Leu Ala Val Ile Gly Pro130 135 140His Ser Ser Glu Leu Ala Met Val Thr Gly Lys Phe Phe Ser Phe Phe145 150 155 160Leu Met Pro Gln Val Ser Tyr Gly Ala Ser Met Glu Leu Leu Ser Ala165 170 175Arg Glu Thr Phe Pro Ser Phe Phe Arg Thr Val Pro Ser Asp Arg Val180 185 190Gln Leu Thr Ala Ala Ala Glu Leu Leu Gln Glu Phe Gly Trp Asn Trp195 200 205Val Ala Ala Leu Gly Ser Asp Asp Glu Tyr Gly Arg Gln Gly Leu Ser210 215 220Ile Phe Ser Ala Leu Ala Ala Ala Arg Gly Ile Cys Ile Ala His Glu225 230 235 240Gly Leu Val Pro Leu Pro Arg Ala Asp Asp Ser Arg Leu Gly Lys Val245 250 255Gln Asp Val Leu His Gln Val Asn Gln Ser Ser Val Gln Val Val Leu260 265 270Leu Phe Ala Ser Val His Ala Ala His Ala Leu Phe Asn Tyr Ser Ile275 280 285Ser Ser Arg Leu Ser Pro Lys Val Trp Val Ala Ser Glu Ala Trp Leu290 295 300Thr Ser Asp Leu Val Met Gly Leu Pro Gly Met Ala Gln Met Gly Thr305 310 315 320Val Leu Gly Phe Leu Gln Arg Gly Ala Gln Leu His Glu Phe Pro Gln325 330 335Tyr Val Lys Thr His Leu Ala Leu Ala Thr Asp Pro Ala Phe Cys Ser340 345 350Ala Leu Gly Glu Arg Glu Gln Gly Leu Glu Glu Asp Val Val Gly Gln355 360 365Arg Cys Pro Gln Cys Asp Cys Ile Thr Leu Gln Asn Val Ser Ala Gly370 375 380Leu Asn His His Gln Thr Phe Ser Val Tyr Ala Ala Val Tyr Ser Val385 390 395 400Ala Gln Ala Leu His Asn Thr Leu Gln Cys Asn Ala Ser Gly Cys Pro405 410 415Ala Gln Asp Pro Val Lys Pro Trp Gln Leu Leu Glu Asn Met Tyr Asn420 425 430Leu Thr Phe His Val Gly Gly Leu Pro Leu Arg Phe Asp Ser Ser Gly435 440 445Asn Val Asp Met Glu Tyr Asp Leu Lys Leu Trp Val Trp Gln Gly Ser450 455 460Val Pro Arg Leu His Asp Val Gly Arg Phe Asn Gly Ser Leu Arg Thr465 470 475 480Glu Arg Leu Lys Ile Arg Trp His Thr Ser Asp Asn Gln Lys Pro Val485 490 495Ser Arg Cys Ser Arg Gln Cys Gln Glu Gly Gln Val Arg Arg Val Lys500 505 510Gly Phe His Ser Cys Cys Tyr Asp Cys Val Asp Cys Glu Ala Gly Ser515 520 525Tyr Arg Gln Asn Pro Asp Asp Ile Ala Cys Thr Phe Cys Gly Gln Asp530 535 540Glu Trp Ser Pro Glu Arg Ser Thr Arg Cys Phe Arg Arg Arg Ser Arg545 550 555 560Phe Leu Ala Trp Gly Glu Pro Ala Val Leu Leu Leu Leu Leu Leu Leu565 570 575Ser Leu Ala Leu Gly Leu Val Leu Ala Ala Leu Gly Leu Phe Val His580 585 590His Arg Asp Ser Pro Leu Val Gln Ala Ser Gly Gly Pro Leu Ala Cys595 600 605Phe Gly Leu Val Cys Leu Gly Leu Val Cys Leu Ser Val Leu Leu Phe610 615 620Pro Gly Gln Pro Ser Pro Ala Arg Cys Leu Ala Gln Gln Pro Leu Ser625 630 635 640His Leu Pro Leu Thr Gly Cys Leu Ser Thr Leu Phe Leu Gln Ala Ala645 650 655Glu Ile Phe Val Glu Ser Glu Leu Pro Leu Ser Trp Ala Asp Arg Leu660 665 670Ser Gly Cys Leu Arg Gly Pro Trp Ala Trp Leu Val Val Leu Leu Ala675 680 685Met Leu Val Glu Val Ala Leu Cys Thr Trp Tyr Leu Val Ala Phe Pro690 695 700Pro Glu Val Val Thr Asp Trp His Met Leu Pro Thr Glu Ala Leu Val705 710 715 720His Cys Arg Thr Arg Ser Trp Val Ser Phe Gly Leu Ala His Ala Thr725 730 735Asn Ala Thr Leu Ala Phe Leu Cys Phe Leu Gly Thr Phe Leu Val Arg740 745 750Ser Gln Pro Gly Arg Tyr Asn Arg Ala Arg Gly Leu Thr Phe Ala Met755 760 765Leu Ala Tyr Phe Ile Thr Trp Val Ser Phe Val Pro Leu Leu Ala Asn770 775 780Val Gln Val Val Leu Arg Pro Ala Val Gln Met Gly Ala Leu Leu Leu785 790 795 800Cys Val Leu Gly Ile Leu Ala Ala Phe His Leu Pro Arg Cys Tyr Leu805 810 815Leu Met Arg Gln Pro Gly Leu Asn Thr Pro Glu Phe Phe Leu Gly Gly820 825 830Gly Pro Gly Asp Ala Gln Gly Gln Asn Asp Gly Asn Thr Gly Asn Gln835 840 845Gly Lys His Glu85032523DNAArtificial SequenceMultiparameter optimized nucleic acid molecule 3atgggcccta gagccaagac aatctgcagc ctgtttttcc tgctgtgggt gctggccgag 60cccgccgaga atagcgactt ttacctgccc ggcgactatc tgctgggcgg cctgttcagc 120ctgcacgcca atatgaaggg gatcgtgcac ctgaattttc tgcaagtccc aatgtgtaaa 180gaatacgaag tgaaagtgat cggctacaat ctgatgcagg ctatgagatt tgccgtggag 240gaaatcaata acgactcctc cctgctgccc ggcgtgctcc tcggctacga gattgtggac 300gtctgttaca tcagcaacaa cgtgcagccc gtgctgtatt ttctggccca cgaggataat 360ctgctgccta tccaggagga ttactccaac tacatcagcc gcgtggtggc cgtgatcggc 420cccgacaaca gcgagagcgt gatgaccgtg gccaattttc tgagcctgtt cctgctgccc 480cagatcacat actccgccat ctccgacgag ctgagggaca aagtgagatt ccccgccctg 540ctgagaacca ccccttctgc cgatcaccat gtggaagcta tggtccagct catgctccat 600ttccgctgga attggattat cgtcctggtg tcctccgata cctacggcag agataacgga 660cagctgctgg gagagcgcgt ggccagacgg gatatctgta ttgcctttca ggaaaccctg 720cctaccctgc agcctaatca gaatatgacc agcgaggagc ggcagagact cgtgacaatc 780gtggataaac tccagcagtc caccgccaga gtggtggtgg tgttcagccc cgatctgaca 840ctgtatcatt tttttaacga agtgctgagg caaaatttca ccggggctgt gtggattgcc 900agcgaaagct gggccattga tcccgtgctg cacaatctga ccgaactggg gcacctgggc 960acatttctcg ggatcacaat ccagtccgtg cctatccctg gcttttccga gttcagggag 1020tggggacctc aggccggacc ccccccactg agcagaacct cccagtccta cacctgtaat 1080caggagtgtg ataattgtct gaatgccacc ctgagcttca ataccatcct gcggctgagc 1140ggcgagagag tggtgtactc cgtgtacagc gccgtgtacg ccgtggccca cgctctgcac 1200tccctgctgg gctgcgataa gtccacctgc acaaagcgcg tggtgtatcc ttggcagctc 1260ctggaggaaa tttggaaagt gaacttcacc ctgctggatc atcagatctt ctttgacccc 1320cagggcgatg tggcactgca cctggaaatc gtgcaatggc agtgggacag atcccagaac 1380ccctttcagt ccgtggccag ctactatcct ctccagaggc agctcaagaa tatccaggat 1440atcagctggc acacagtgaa taataccatc cccatgagca tgtgcagcaa gagatgccag 1500agcggccaga agaagaaacc cgtggggatt cacgtgtgct gttttgagtg tattgattgc 1560ctgcctggga ccttcctgaa tcacaccgag gacgagtacg agtgtcaggc ctgtcccaac 1620aatgagtgga gctaccagtc cgagaccagc tgctttaagc gccagctcgt gtttctggag 1680tggcacgagg cccctacaat tgccgtggca ctgctggctg ctttgggctt tctgtccaca 1740ctggctattc tggtgatctt ttggagacac tttcagaccc ccatcgtgag aagcgccgga 1800ggccctatgt gttttctcat gctcaccctg ctgctcgtcg cctatatggt ggtgcccgtg 1860tatgttggcc ctcccaaagt gagcacctgc ctgtgtagac aggccctgtt ccccctgtgt 1920ttcaccatct gcatcagctg catcgccgtg aggagctttc agatcgtgtg tgcttttaag 1980atggcctccc ggttccccag agcctactcc tactgggtga gataccaggg cccttacgtg 2040agcatggcct tcatcaccgt gctgaagatg gtgatcgtgg tcatcgggat gctggctacc 2100ggcctgagcc ctaccaccag aaccgacccc gacgatccta agattaccat cgtgagctgc 2160aaccccaatt acaggaactc cctgctgttt aatacctctc tggatctgct gctgagcgtg 2220gtgggcttct ctttcgccta tatggggaag gaactgccta ccaattacaa tgaagctaag 2280tttatcaccc tgtccatgac cttttacttc acaagcagcg tgagcctgtg cacctttatg 2340agcgcctact ccggcgtgct ggtgacaatc gtcgatctgc tggtgaccgt gctgaatctg 2400ctggccatct ctctgggcta ttttgggcct aagtgttaca tgattctgtt ttaccccgag 2460aggaacaccc ctgcctactt taattccatg attcagggct acacaatgcg gcgcgactga 2520taa 252342582DNAArtificial SequenceMultiparameter optimized nucleic acid molecule 4atgctgggac ccgccgtgct gggcctgtcc ctgtgggcac tgctgcaccc cggcacaggc 60gcccctctgt gcctgagcca gcagctgaga atgaagggcg attatgtgct cggcggcctg 120tttcctctgg gcgaagccga agaagccggc ctgaggagca gaaccagacc tagctccccc 180gtgtgcacaa gattcagcag caacgggctg ctgtgggccc tcgctatgaa gatggccgtc 240gaggaaatca ataataagag cgacctgctg cctggcctga gactgggcta tgacctgttc 300gacacctgca gcgaacccgt ggtggctatg aaaccctccc tgatgtttct ggccaaagcc 360ggcagcagag atattgccgc ctattgtaat tacacccagt atcagcctag agtgctggcc 420gtgatcggcc ctcacagcag cgagctggcc atggtgacag ggaagttttt cagctttttt 480ctgatgcccc aagtgagcta tggcgcctcc atggaactgc tgtccgccag agagacattc 540cccagcttct tcaggacagt gccctccgat agagttcagc tgaccgccgc tgccgaactg 600ctgcaggaat ttggatggaa ttgggtcgca gctctgggct ccgatgatga gtacggcaga 660caggggctga gcattttcag cgccctggct gccgccagag gcatctgtat tgcccatgag 720ggactggtgc ccctgcctag agccgatgat agccggctgg gcaaagtcca ggatgtgctg 780caccaagtga atcagtcctc cgtgcaagtg gtcctgctct ttgccagcgt gcatgccgcc 840catgccctgt tcaattactc catcagcagc cgcctgagcc ctaaagtgtg ggttgcctcc 900gaggcttggc tgacaagcga tctggtgatg ggcctgcctg ggatggctca aatgggcacc 960gtgctgggct ttctgcagag aggcgctcag ctccacgaat ttcctcagta tgtgaaaacc 1020catctggccc tcgccacaga tcctgccttc tgcagcgccc tgggagaaag agaacagggc 1080ctggaggaag atgtggtcgg acagagatgc cctcagtgcg attgtattac cctgcagaac 1140gtgtctgccg gcctgaatca ccatcagacc ttcagcgtgt acgccgccgt gtactccgtg 1200gctcaggctc tgcacaatac cctgcagtgt aacgccagcg gctgccctgc ccaggatcct 1260gtgaaacctt ggcagctgct cgagaatatg tataatctga catttcacgt gggaggcctg 1320cccctgagat tcgatagctc cggcaatgtg gatatggaat atgatctgaa actgtgggtc 1380tggcaaggca gcgtgcctag actgcacgat gtgggccgct ttaacggctc tctgcggacc 1440gagaggctga agattagatg gcacaccagc gacaatcaga agcctgtgag ccggtgcagc 1500agacagtgtc aggagggaca agtgcggaga gtgaagggct tccacagctg ctgttacgac 1560tgcgtggatt gtgaggccgg ctcctacaga cagaaccccg atgatatcgc ctgtaccttc 1620tgcggacagg acgagtggag ccctgagaga agcaccagat gcttccggcg gaggagcaga 1680tttctggcct ggggagaacc cgctgtgctg cttctgctgt tgctgctctc tctggccctg 1740ggactggtgc tggccgcact gggcctgttt gtgcaccaca gagacagccc cctggtgcag 1800gccagcggcg gacctctggc ctgtttcggg ctcgtgtgtc tgggccttgt gtgtctgagc 1860gtgctgctct ttccaggcca gccttcccca gctagatgtc tggctcagca acccctgagc 1920cacctgcccc tgaccggctg tctgagcacc ctgtttctcc aggccgctga gatctttgtg 1980gagagcgagc tgcccctgtc ttgggccgac agactgagcg gctgtctgag aggcccctgg 2040gcttggctgg ttgtcctcct ggctatgctg gttgaagtgg ccctgtgtac ctggtatctg 2100gtcgcctttc cacccgaagt ggtgaccgat tggcatatgt tgcctaccga ggccctggtg 2160cactgtagaa ccaggagctg ggtgagcttt ggactggccc acgccacaaa cgccaccctg 2220gccttcctgt gtttcctggg gacctttctg gtgagaagcc agcccggcag atacaataga 2280gccaggggcc tgacattcgc catgctcgct tactttatta catgggtgag cttcgtgcct 2340ctgctggcca acgtgcaagt tgtgctgcgc cctgccgtgc aaatgggagc actgctgctg 2400tgcgtgctgg gcattctggc cgcttttcat ctgccccgct gctatctgct gatgagacag 2460cccggcctga atacccctga gttttttctg ggcggaggcc ctggcgacgc ccagggacag 2520aacgacggca acaccggcaa ccagggcaag cacgagtgat aagagaccgc ggccgctgat 2580ca 2582

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed