Transgenic mouse for screening and for studies of the pharmacodynamics and pharmacokinetics of ligands acting on the oestrogen receptor and its intracellular receptors, and method for the preparation thereof

Abstract
The object of the invention is a method for the production of a non-human transgenic mammal by means of which it is possible to monitor in vivo and in all the tissues the state of activation of any intracellular receptor, utilising a reporter gene inducible by natural or synthetic molecules which modulate the activity of such receptor. The mammal is question is preferably a mouse.
Description


SPECIFICATION

[0001] Research carried out in the last ten years has shown that oestrogen deficiency is associated with an increased risk of the appearance of a large number of diseases of the nervous, immune, cardiovascular and bone systems. These studies thus suggest that the oestrogens, traditionally considered to be hormones responsible solely for the control of the reproductive functions, in fact have an important role in the maintenance of the homeostasis of many tissues in the mammal (for a review, see Nilsson and Gustafsson, 2000); pharmacological research is thus directed towards the obtaining of therapeutically active molecules which can be substituted for the hormone in order to exert their protective and beneficial activity solely in specific target organs.


[0002] Meanwhile, research at the molecular and cellular level has clarified the mechanism by which the hormone acts on the target cells (for a review, see Tsai and O'Malley, 1994). In particular, the hormone recognises and binds to intracellular proteins, called oestrogen receptors (so far, two types of receptor, designated by the first two letters of the Greek alphabet, are known). The hormone-receptor complex is then enabled for the recognition of specific sequences of DNA (called hormone response elements), for interaction with other protein factors (some of them ubiquitous and others tissue-specific) for the activation of the transcription of the target genes. This mechanism provides the molecular basis for the explanation of an observation made with synthetic ligands of the oestrogen receptors, namely that these can function as receptor agonists in some organs and antagonists in others.


[0003] At present, a considerable number of pharmaceutical multinationals are involved in the development of molecules for use in replacement therapies, which may be capable of acting as activators of oestrogen receptors in some organs and as antagonists in others. For example, in the case of the oestrogen receptor, molecules are currently being sought which activate this receptor in organs such as bone and brain, but not the uterus and mammary tissue where the trophic activity of the receptor could favour the onset of neoplasias.


[0004] The bottleneck in these studies comprises the identification of the molecules endowed with the desired agonistic/antagonistic activity. At present, the screening of such molecules is carried out in transformed cell lines suitably engineered to express an exogenous marker gene of hormonal activity.


[0005] The limits of this type of methodology are connected with the fact that this technique is performed on immortalised/neoplastic cells, which are thus modified with respect to the physiological target; further, it does not provide any information concerning the pharmacokinetics and the bioavailability of the compound with potential pharmacological activity. For these reasons, the ligands identified by screening on cells must in any case be subjected to tests on animals. Finally, not all the areas of the organism which are targets of these hormones are yet fully known. According to the present invention, it was decided to overcome these limitations by the generation of a mouse engineered so that it expresses the marker gene of hormonal activity in all its cells and tissues. Hence, the administration to such a mouse of any compound with oestrogenic activity would result in an accumulation of the gene product in those organs/cells where the compound acts as a receptor agonist. This mouse represents an ideal system for the screening of molecules acting on the oestrogen receptors in that:


[0006] 1. It allows the simultaneous visualisation of all the organs/cells in which the compound under test exerts a pharmacological activity (including cells whose responsivity to the hormone is unknown).


[0007] 2. It makes it possible to know the distribution of the compound (for example whether it penetrates the blood-brain barrier).


[0008] 3. It makes it possible also to carry out pharmacokinetic studies to clarify the catabolism of the ligand in question, and its absorption times as well as its possible persistence and activity in all parts of the mouse, allowing the prediction of possible side-effects. Such methods can be carried out in vivo by non-invasive imaging methodologies.


[0009] Another use of the animal model according to the invention concerns the ecotoxicology field. One of the major environmental contamination problems is connected with the activity of the so-called xenooestrogens. It has been proved that many synthetic substances widely used in agriculture (e.g. atrazine) and other compounds present in industrial wastes (e.g. dioxin) display active interference with the endocrine system in mammals causing strong repercussions on the reproductive system right up to sterility; the increase in such substances in the environment is now recognised as one of the causes of increased sterility in man (Sharpe and Skakkebaek, 1993). It will thus also be possible to use the animal as a biosensor for specific environmental pollutant substances (e.g. pesticides).


[0010] Finally, it will be possible to use the animal as a source of cells for in vitro culturing. Primary or immortalised cultures from different tissues can be obtained by standard cell culturing protocols. Such cultures can be used in the screening of compounds with hormonal action.


[0011] The invention here described concerns the generation of a transgenic animal. As is well-known, the transgenics are animals into whose chromosome apparatus an exogenous gene has been inserted and are capable of transmitting it to their progeny. Two basic procedures exist for the generation of transgenic animals which were initially developed in the mouse: 1. the microinjection of the fragment of DNA into the genome at the single-cell stage, in the male pronucleus of a fertilised oocyte and its reimplantation into pseudogravid females (Constantini and Lacy, 1981; Gordon and Ruddle, 1981); 2. the transfection of the DNA into embryonic stem cells and the injection of these into the blastocysts (Robertson et al., 1986; Gossler et al., 1986). Various other animal species have been engineered in this way: for example Hammer and coworkers (1985) have described the application of trangenesis in rabbits, in sheep and in pigs. The mouse is still an animal much utilised in transgenesis and is also the first transgenic animal to have been patented (EP 0169672B1).


[0012] The present invention relates to a method for the production of a non-human transgenic mammalian animal by means of which it is possible to monitor in vivo and in all the tissues the activation state of an intra-cellular receptor, characterized in that a reporter gene whose transcription is inducible by synthetic or natural molecules which modulate the activity of such a receptor is incorporated into the genome of the animal.


[0013] Therefore, in the following the said reporter gene will also be referred to as reporter transgene.


[0014] More specifically, the present invention provides a method for the production of a transgenic mammalian animal utilisable as an experimental model for simultaneously monitoring in vivo the pharmacological activity, the tissue-specificity, the pharmacokinetics and the pharmaco-dynamics dynamics of molecules with potential activity on intracellular receptors.


[0015] Generation of the Animal Model


[0016] The transgenic model which is the object of the present invention can be obtained by the integration into the somatic and germinal line of a gene called a reporter, provided with control regions that make its expression inducible by such hormones. The gene in question is not present in the genome of the mammal and codes for an enzyme easily quantifiable by an enzyme test. Any reporter gene is utilisable for the purposes of the present invention. A list of reporter genes currently available are [sic] indicated, together with some of their characteristics, in Table I given later. These are luciferase, green fluorescent protein (GFP), beta-galactosidase, beta-lactamase and chloramphenicol acetyltransferase (CAT).


[0017] Also mentioned are dopamine 2 receptor (D2R) and thymidine kinase (TK).


[0018] The possibility of hormone-dependently inducing the transcription of a reporter gene is a function of the presence of specific sequences to which the ligand-receptor complex binds. In particular according to the invention a series of constructs consisting of palindromic sequences responsive to oestrogens (ERE) shown in FIG. 2 of the attached drawings, and described later, was generated. They are:


[0019] ERE4X


[0020] ERE2X-33 bp linker-ERE2X, and


[0021] ERE2X, wherein


[0022] ERE2X or 4X=oestrogen-responsive element multimerised 2- or 4-fold, respectively and


[0023] 33 bp linker=non-responsive DNA linking sequence between the ERE.


[0024] Of these, the construct utilised for the generation of the oestrogen-responsive transgene according to the invention preferably has the following structure:


[0025] ERE2X.


[0026] Such sequences were placed upstream of a promoter which does not display tissue-specificity, such as the constitutive promoter of the gene for viral thymidine kinase, TKprom. The promoter of the final construct is thus constituted as follows:


[0027] ERE2X-TKprom-Reporter.


[0028] A constitutive promoter according to the invention can also be a minimal promoter consisting of TATA box and transcription initiator sequence.


[0029] In the tissues of the model animal presented, the administration of agonists causes an increase in the activity of the protein encoded by the reporter; this increase depends specifically on the presence of intracellular receptors activated by the ligand in the target cell and is proportional to the degree of hormonal stimulation. In other words, the expression of the transgene constitutes a signal of the existence of some activity of the specific hormone in some specific body area of the transgenic mouse. The model also allows the study of molecules with antagonistic activity towards oestrogen receptors. In this case, the evaluation of the antagonistic activity of the molecule in question will be effected in animals simultaneously treated with oestrogens to evaluate the blocking of the hormonal activity by the presumed antagonist.


[0030] The stable introduction of an ERE-TKprom-Reporter construct into the genome of fertilised oocytes or of embryonic stem cells makes it possible to obtain a first series of individuals (called founders). From these founders are obtained, via hereditary transmission, firstly the heterozygotic individuals and then the homozygotic individuals for the transgene which will be inherited in the Mendelian manner in case of insertion into a unique site. However, the expression of the transgene inserted in this manner into the animal is subject to the so-called positional effect, namely the influence of the chromatin surrounding the site of insertion into the genome. As a result, the transgene will be expressed or not expressed, in an incorrect manner and often with undesired tissue preferences. This phenomenon, called PVE (Position Variegation Effect), is typical of eukaryotic cells and has been amply characterised and studied; it was the principal cause of the failure of preceding attempts to generate the animal model which is the object of this patent. To circumvent this problem, according to the present invention suitable sequences called insulators were used, which, placed at the flanks of the transgene, have the ability to isolate it from the effect of the heterochromatin and from surrounding enhancer sequences (for a review, see Fang-Lin and Elgin, 1999). Some of these sequences are shown in Table II.


[0031] The final constructs, or plasmids, utilised for the transgenesis of the mouse which is the object of the present invention were constituted as follows (FIG. 3):


[0032] pMAR=MAR-ERE2X-TKpr-Luciferase-MAR


[0033] pHS4=HS4-ERE2X-TKpr-Luciferase-HS4.


[0034] The screening system developed according to the present invention can be extended to any ligand acting on intracellular receptors. In this case the DNA sequence responsive to the hormone is selected among those known and available as described in Table III below, such as PRE, GRE, RARE, TRE, VD3RE and PPRE.







[0035] The Figures cited above are described below:


[0036]
FIG. 1 is the schematic representation of the starting expression plasmid containing the luciferase reporter gene under the control of the constitutive promoter of viral thymidine kinase (TK).


[0037]
FIG. 2 is the schematic representation of the plasmids tested for inducibility by oestrogens, in which are present the ERE sequences, upstream of the TK promoter and with various arrangements: the ERE palindromes have been dimerised (1), or tetramerised with (3) or without (2) spacing sequences.


[0038]
FIG. 3 is the schematic representation of the fusion of the insulator sequences MAR and HS4 with the plasmid ERE2X-TK-luciferase.


[0039]
FIG. 4 is the schematic representation of the 0.77 Kb EcoRI/EcoRI probe used for the screening of the transgenic mice generated with the pMAR and pHS4 constructs.






PREPARATION OF THE CONSTRUCTS

[0040] The starting construct pTKluc described in FIG. 1 was obtained by subcloning the luciferase gene from pGL2basic (Promega) into the BamHI site of pBluescript (Stratagene); the −109 bp fragment of the TK promoter was isolated from the vector pBLCAT2 (Luckov and Schutz, 1987) and subcloned in the Hindill site of pBluescript. The construct obtained (pTKluc) was sequenced and the expression of luciferase was verified in MCF-7 human mammary carcinoma cells.


[0041] The oestrogen-responsive sequence (ERE), see Table III below, multimerised in the various arrangements described in FIG. 2, was subcloned in the Sail site of pTKluc; the oestrogen-responsivitiy of the constructs thus obtained was examined in MCF-7 cells and in human neuroblastoma cells SK-N-BE. The highest responsivity to oestradiol was obtained from the construct pERE2X-TKluc (FIG. 2) which was chosen for generation of the final vectors utilised in the transgenesis.


[0042] The insulators HS4 (Chung et al., 1993) and MAR (Stief et al., 1989) were subcloned into the flanking regions of pERE2XTKluc in the Kpnl and Notl sites generating the vectors pHS4 and pMAR (FIG. 3).


[0043] The probe used in the screening of the transgenic animals (FIG. 4) was obtained by digestion of the plasmid pERE2XTKluc with the restriction enzyme EcoRI and the 0.77 Kb fragment was separated by electrophoresis on agarose gel and purified by electroelution (Maniatis et al., 1982).


[0044] Production of Transgenic Mice


[0045] The DNA fragments containing the transgenes were obtained from the plasmids pMAR and pHS4 by digesting them with the restriction enzyme BsshII and thus releasing the inserts of 8.95 Kb and 7.85 Kb respectively (FIG. 3); the transgenes were purified by electrophoresis on agarose gel and subsequent electroelution procedure (Maniatis et al., 1982). About 400 copies of the insert thus purified were injected into the male pronucleus of a fertilised egg deriving from individuals of the B6D2F1 strain of mouse. The mice were obtained from the Charles River Laboratories. The injected eggs were reimplanted into pseudo-gravid females. In this way, about 100 individuals were obtained. At the age of four weeks, the DNA from the biopsy of the tails of these individuals was extracted (Maniatis et al., 1982). About 10 μg of DNA for each sample were immobilised with a Slot Blot device (Schleicher and Schuell) on nylon filters (HybondN, Amersham). The filters obtained were subjected to hybridisation experiments with the 0.7 Kb EcoRI/EcoRI DNA probe described in FIG. 4, labelled with 32P using a kit based on the multiprimer principle, according to the producer's instructions (Megaprime DNA labelling system, Amersham). The hybridisation procedure was effected essentially as previously described (Maniatis et al., 1982); in particular, the hybridisation temperature used was 60° C. and the washings were performed in a solution containing 0.1×SSC/0.1% SDS (3 washings of 10 minutes at ambient temperature and 2 washings of 30 minutes at 60° C.).


[0046] The screening of the founders, effected as previously described, made it possible to identify 17 mice with the transgene integrated: 10 pMAR and 7 pHS4. Only 12 founders succeeded in generating fertile progeny positive for the presence of the transgene.


[0047] The number of copies integrated in the different lines is variable from one to thirty.


[0048] The founders were crossed with non-injected animals from the same original strain (B6D2F1) and the new-born at the age of four weeks were analysed for the presence of the transgene as described for the founders, by means of the Slot Blot technique.


[0049] Through successive recrossing of each of the 12 fertile lines, heterozygotic individuals and in the end also homozygotic lines were obtained.


[0050] Analysis of the Transgenic Mice


[0051] To verify the expression of the transgene in the different tissues and its inducibility by oestrogens, 4 independent experiments were carried out on 8 female individuals of each line (4 heterozygotic and 4 homozygotic). For each experiment, two female individuals were ovariectomised. With the aim of reducing the oestrogens present in the bloodstream of the ovariectomised animals below the detection limit, there was a wait of two weeks before the induction experiment was carried out; the individuals were then subjected to subcutaneous (s.c.) injection of mineral oil with or without 50 μg/kg of dissolved 17-β oestradiol; 16 hours after the treatment, the individuals were sacrificed and the following tissues were dissected out of them: uterus, liver, spleen, lung, heart, kidney, hypothalamus and brain. The protein extracts were obtained by disintegration of the tissue with a homogenizer in the presence of a phosphate lysis buffer consisting of: 0.1 M KH2PO4, 0.1 M K2HPO4, 1 mM DTT, 4 mM EGTA and 4 mM EDTA at pH 7.8. The suspension was frozen and thawed three times, and centrifuged for 30 minutes at 13,000 g; the supernatant was collected and equal quantities of protein, measured by the colorimetric method of Bradford, were subjected to enzymatic testing to reveal the activity of the enzyme luciferase. The method for detection of the activity of the reporter was carried out with a kit by the procedure recommended by the producer (Sigma), mixing ca. 20 μg of protein extract with the luciferin substrate in the presence of 0.5 mM ATP and 30 mM DTT and measuring the fluorescence emitted using a luminometer (Lumat BL 9500, Berthold). With reference to FIGS. 5 to 7 of the attached drawings, individuals were subcutaneously injected (s.c.) with 50 μg/Kg 17β-estradiol (E2) or with vehicle (vegetable oil) and sacrificed at 24 hours. Luciferase enzymatic activity was evaluated and plotted in the graph. The arbitrary units are obtained considering the luciferase activity in the uterus of control individuals=100.


[0052]
FIG. 5 shows the expression of luciferase in line 2 of the transgenic mice treated with 50 μg/kg of 17-β oestradiol (s.c.) for 24 hours, FIG. 6 shows the expression of luciferase in line 61 of the transgenic mice treated with 50 μg/kg of 17-β oestradiol (s.c.) for 24 hours, and FIG. 7 shows the expression of luciferase in line 59 of the transgenic mice treated with 50 μg/kg of 17-β oestradiol (s.c.) for 24 hours.


[0053] Of the 10 lines analysed, one (line 2, transgene pMAR) displayed ubiquitous and inducible expression of the transgene (FIG. 5); one was found to be inducible predominantly in the brain and in the liver (line 61, transgene pHS4) (FIG. 6), and one is inducible in the brain and in the hypothalamus (line 59, transgene PMAR) (FIG. 7).


[0054] Test for the Agonistic and Antagonistic Action of a Compound and for the Activity of Xenooestrogens


[0055] The animal of this invention can be utilised to test the ability of a compound to behave as an agonist or as an antagonist in the various tissues. By subcutaneous injection of the substance in the presence or in the absence of 17-β oestradiol it is in fact possible to compare the distribution and the intensity of expression of the luciferase gene in the various tissues and to compare it with the control or with the injection of 17-β oestradiol alone. The effect of in vivo administration of two known ER antagonists was investigated in line 2 of transgenic mice. FIG. 8 (upper panel) shows blockade of 17-β oestradiol (E2) activation by 4- hydroxytamoxifen (T) and ICI 182,780 (ICI) and partial agonist activity of tamoxifen (T) in liver and brain. Bars represent the average±s.e.m of 5-7 mice. *(P<0.01 as compared to the control), °(P<0.01 as compared to the E2-treated); P were calculated with ANOVA followed by Scheffé test. Ligands were administered at the following doses: E2 50 μg/Kg, 4-hydroxytamoxifen and ICI 182,780 250 μg/Kg. Luciferase is expressed as fold induction obtained as the ratio between luciferase activity in ligands treated/control mice.


[0056] With reference to the upper panel of FIG. 8, s.c. administration of 250 μg/kg of 4-hydroxy-tamoxifen for 6 hours increased the level of luciferase in the liver 12-fold and 7-fold respectively, confirming in vivo the partial agonist activity of 4-hydroxytamoxifen in these tissues. The injection of 250 μg/kg of tamoxifen or ICI 182,780 one hour before the administration of 50 μg/kg of E2 inhibits the E2-dependent activation of luciferase expression as expected from the antagonistic effect of the compounds with respect to E2.


[0057] The animal of this invention can be utilised to verify the interference of environmental contaminants (pesticides, fertilisers, etc.) with the endocrine system, by administration of these by the transdermal or subcutaneous route or via the diet. The effects of a paradigmatic xenooestrogen, the organochlorine DDT, were investigated in our model. FIG. 8 (lower panel) shows the luciferase activity evaluated in the liver and brain (two known target organs for DDT toxicity) of individuals injected s.c. with 5000 μg/ml DDT and sacrificed at 3, 6, 16, 24, 48 or 72 hours. As shown, at 24 hours, maximal induction of luciferase activity was detected in both liver and brain compared to a control animal injected with vegetable oil (vehicle); this induction persisted until 72 hours.


[0058] Pharmacokinetics and Pharmacodynamics of E2 in the Transgenic Animals


[0059] The pharmacokinetics of any compound acting via intracellular receptors can be studied in the animal of the invention, if the reporter used codes for a protein with a fast turnover in mammalian cells (e.g. luciferase, which has a 3 hour half-life in mammalian cells). This feature is required in order to follow the variation in receptor activation with time. FIG. 9 (upper panel) shows a pharmacokinetic study of E2 in the transgenic animal generated; individuals were injected s.c. with 50 μg/kg of E2 and sacrificed after 3, 6 or 16 hours. The maximal luciferase accumulation was observed at 6 hours in bone and liver, which correlates with the expected peak of oestrogen receptor transcriptional activity. Luciferase activity is expressed as fold induction as defined in FIG. 8. Bars are representative of the average values of at least 5 individuals.


[0060] The pharmacodynamics of any compound acting via intracellular receptors can also be correctly predicted by the transgenic model of the invention. As an illustrative example, ovariectomised luciferase activity was measured in 26 different tissues from 2-month old female mice which had been ovariectomised 2 weeks prior to the experiment. Mice were treated for 16 hours with either vehicle or E2 subcutaneously (s.c.). FIG. 9 (lower panel) shows that hormonal treatment induced an increase in the enzyme content compared to controls in oestrogen target organs. This distribution is very consistent with the reported tissue distribution of oestrogen receptors.


[0061] Tissue Cultures


[0062] The tissues of the animal which is the object of this invention can further be utilised as sources of cells for in vitro culture by means of standard culturing techniques. These techniques make it possible to obtain primary cultures which can be utilised directly as nontransformed lines for the screening of substances with oestrogenic activity, or can be transformed in order to obtain lines whose cells continue to proliferate. As an example of such an application, bone marrow cells from the transgenic mouse generated were obtained and used in a pharmacological study; the results are summarised in FIG. 10. Two millions of bone marrow cells were suspended in phenol red-free RPMI 1640 with 10% stripped serum. Cells were treated with vehicle (C), with increasing concentrations of E2 (0,001; 0,01; 0,1; 1 and 10 nM), with 100 nM ICI 182,780 (ICI) with 4-hydroxytamoxifen (T) either alone or with 1 nM E2; progesterone (Prog) and dexametasone (Dex) were used at 10 nM final concentration. Bars represent the average±s.e.m. of 5 individual experiments each done in triplicate. *(P<0.01 as compared to the control), **(P<0.005 as compared to the control), °(P<0.05 as compared to the T-treated); P were calculated with ANOVA followed by Scheffe test. The cells were treated for 16 hours with increasing concentrations of E2 (0.01-10 nM) or with 100 nM of two ER antagonists: 4-hydroxytamoxifen (T) and ICI 182,780 (ICI) alone or in the presence of 1 nM E2. E2 induced a dose-dependent increase in luciferase accumulation blocked by the presence of ICI 182,780. ICI 182,780 by itself did not sort [sic] any effect. Conversely, 4-hydroxytamoxifen induced a significant increase in luciferase levels even though lower than E2 at the same concentration. On co-administration with E2, 4-hydroxytamoxifen induced higher luciferase accumulation, yet the level reached was still lower than with E2 alone. This is compatible with the partial agonist activity of 4-hydroxytamoxifen and with the fact that it is present in the solution at a concentration 100 times higher than that of E2. As a control, we also tested progesterone and dexamethasone (10 nM). Both ligands did not have any effect on the ER reporter. Taken together, these date confirm that primary cells can be explanted from the engineered mice; the transgene is controlled by ligands of ER with modalities recapitulating those reported for the natural target genes.


[0063] Deposits


[0064] The plasmids utilised for the transgenesis, described in FIG. 3, have been deposited at the ECACC (European Collection of Cell Cultures) Institute, Salisbury, Wiltshire SP4 OJG, UK and have been given the following access numbers: pMAR00041120 and pHS400041121.


[0065] The sequences of the plasmids have been deposited at the EMBL (European Molecular Biology Laboratory) Institute, Heidelberg, Germany, and have been given the following access numbers: pHS4=AJ277959 and pMAR=A277960.


[0066] Finally, the list of literature references cited is given.


[0067] Costantini F and Lacy E., (1981) Nature, 294: 92-94.


[0068] Chalfie M., Tu Y., Euskirchen G., Ward W. W. and Prasher D. C. (1994) Science, 263: 802-805.


[0069] Chung J. H., Whiteley M and Felsenfeld G. (1993) Cell, 74: 505-514.


[0070] De The H., Vivanco Ruiz M. D. M., Tiollais P., Stunnenberg H. and Dejean A. (1990) Nature, 343, 177-180.


[0071] de Wet J. R. et al. (1987) Mol. Cell. Biology, 7: 725.


[0072] Durand B., Saunders M., Leroy P., Leid M. and Chambon P. (1992) Cell, 71: 73-86.


[0073] Fang-Lin S. and Elgin S. C. R. (1999) Cell, 99: 459-462.


[0074] Fu Y., Wang Y and Evans S. M. (1998) Nature Biotechnology, 16: 253-257.


[0075] Gordon J. W. and Ruddle F. H. (1981) Science, 214: 1244-1246.


[0076] Gorman C. M., Moffat L. F. and Howard B. H. (1982) Mol. Cell. Biol., 2: 1044.


[0077] Gossler A. T., Doetschman R., Korn E. and Kemler R. (1986) Proc. Natl. Acad. Sci. USA, 83: 9065-9069.


[0078] Klein-Hitpass L., Schorpp M., Wagner U., Ryffel G. U., Kliewer S. A. and Umesono K. (1986) Cell, 46: 1053-61.


[0079] Noonan D. J., Heyman R. A. and Evans R. M. (1992) Nature, 358: 771-774.


[0080] Luckow B. and Schutz G. (1987) Nucleic Acids Res., 15: 5490.


[0081] Maniatis T., Fritsch E. F. and Sambrook J. (1982) Cold Spring Harbor Laboratory Press. Cold Spring Harbor, N.Y.


[0082] Miller J. H. (1972) Cold Spring Harbor Laboratory Press. Cold Spring Harbor, N.Y.


[0083] Nilsson S and Gustafsson J- A (2000) in press.


[0084] Noda M., Vogel R., Craig A. M., Prahl J., DeLuca H. F. and Denhardt D. T. (1990) Proc. NatI. Acad. Sci. USA, 87: 9995-9999.


[0085] Robertson E. A., Bradley M. and Evans M. (1986) Nature, 323: 445-448.


[0086] Sap J., de Magistris L., Scmitt J., Stunnenberg H. and Vennenstrom B. (1990) EMBO J., 9: 887-896.


[0087] Sharpe R. M. and Skakkebaek N. E. (1993) Lancet, 341: 1392-1395.


[0088] Stief A., Winter D., Stratling W. H. and Stippel A. E. (1989) Nature, 341: 343-345.


[0089] Stunnenberg H. G. (1993) Bioessays, 15: 309-15.


[0090] Tsai M- J. and O'Malley B. W. (1994) Ann. Rev. Biochem., 63: 451-486.


[0091] von der Ahe D., Janich S., Scheidereit C., Renkawitz R., Schutz G. and Beato M. (1985) Nature, 313: 706-709.


[0092] Zlokarnik G., Negulescu P. A., Knapp T. E., Mere L., Burres N., Feng L., Whitney M., Roemer K. and Tsien R. Y. (1998) Science, 279: 84-88.
1TABLE IEnzymaticDetection inReportertestvital cellsReferencesLuciferaseyesyesde Wet1987GreennoyesChalfie et al.,fluorescent1994proteinbetayesnoMillergalactosidase1982betayesyesZlokarnik et al.,lactamase1998chloramphenicolyesnoGorman et al.,acetyl-1982transferase


[0093]

2









TABLE II











Insulator
Gene of origin
References









HS4
human beta-
Chung et al.,




globin
1993



MAR
chicken
Stief et al.,




lysozyme
1989



ITR
adeno-
Fu et al.,




associated virus
1998











[0094]

3









TABLE III










Gene regulated by
Receptor that binds



Responsive
Sequence
the responsive
the responsive


sequence
recognised
element cited
sequence
References







ERE
ggtca(n)3tgacc
vitellogenin
oestrogen receptor
Klein-Hitpass L,






et al., 1986


GRE/PRE
agaaca(n)3tgttct
MMTV
glucocorticoid and
von der Ahe D. et al., 1985





progesterone





receptors


RARE
ggttca(n)5agttca
RAR-β2
retinoic acid receptor
De The et al, 1990



agttca(n)2aggtca
CRABPII



agttcanaggtca
CRABPII

Durand et al., 1992


TRE
gggtca(n)4aggtcc
growth hormone
thyroid hormone
Sap et al., 1990





receptor


VD3RE
gttca(n)3ggttca
osteopontin
vitamin D receptor
Noda et al, 1990


PPRE
aggtcanaggtca
acyl CoA oxidase
peroxysome
Kliewer et al, 1992





proliferation factor





receptor










Claims
  • 1. A method for the production of a non-human transgenic mammalian animal by means of which it is possible to monitor in vivo and in all the tissues the state of activation of an intracellular receptor, characterized in that a reporter gene, the transcription of which is inducible by natural or synthetic molecules which modulate the activity of such receptor, is incorporated into the genome of the said animal.
  • 2. A method according to claim 1, characterized in that the said reporter transgene is ligated to insulator sequences, sequences responsive to intracellular hormones, and constitutive promoters.
  • 3. A method according to claim 1, characterized in that the said reporter transgene is selected among: luciferase, green fluorescent protein (GFP), beta-galactosidase, beta-lactamase, chloramphenicol acetyltransferase (CAT), dopamine 2 receptor (D2R) and thymldine kinase (TK).
  • 4. A method according to claims 1 and 2, characterized in that the induction of the said transcription by the said molecules is governed by the insertion into the said transgene of one of the following sequences responsive to the said molecules which modulate the activity of such receptor: ERE, PRE, GRE, RERE, TRE, VD3RE or PPRE.
  • 5. A method according to claims 2 and 4, characterized in that one of the following constitutive promoters is ligated to the said sequences: viral thymidine kinase promoter (TK) or minimum promoter consisting of TATA box and transcription initiator sequence.
  • 6. Method according to claim 5, characterized in that the resulting construct is constituted as follows: ERE2X-TKprom-Reporter.
  • 7. A method according to claims 2 and 4, characterized in that the insulators MAR, HS4 or ITR are ligated to the said sequences by subcloning them into the regions flanking the said transgene, so giving rise to a plasmid.
  • 8. Method according to claim 7, characterized in that the said plasmids are constituted as follows: pMAR=MAR-ERE2X-TKpr-Luciferase-MAR pHA4=HS4-ERE2X-TKpr-Luciferase-HS4.
  • 9. A method according to claim 8, characterized in that sequences of the said plasmids defined by the EMBL access numbers pHS4=AJ277959 and pMAR=AJ277960 are used.
  • 10. A method according to claim 1, which provides for the incorporation of the said transgene into the genome at a stage not higher than the single embryonic cell.
  • 11. A method according to claim 1, which provides for the incorporation of the said transgene into the genome at a stage not higher than B embryonic cells.
  • 12. A method according to claim 1, characterized in that it provides for the injection of the said transgene into the male pronucleus of a fertilised egg of the said animal.
  • 13. A method for checking the agonistic and antagonistic action of ligands for intracellular receptors, characterized in that a transgenic animal generated with one of the methods stated in the foregoing claims is used.
  • 14. A method for studying the pharmacokinetics and the pharmacokinetics combined to the pharmacodynamics of any compound acting through intracellular receptors in order to evaluate the biotransformation or the dynamics of compound absorption, distribution and elimination within the tissues of the said animal, characterized in that a transgenic animal as generated by a method according to one or more of the foregoing claims is used.
  • 15. A method for performing external monitoring of compounds in living mammals by non-invasive imaging, characterized in that a transgenic animal as generated by a method according to one or more of the foregoing claims is used.
  • 16. A method for checking the action of pesticides, insecticides or in general of environmental contaminants, on the endocrine system, characterized in that a transgenic animal generated by one of the methods stated in the foregoing claims is utilised.
  • 17. Plasmids defined by the ECACC access number pMAR00041120 and pHS400041121.
  • 18. A sequence of plasmids defined by the EMBL access number pHS4=AJ277959 and pMAR=AJ277960.
  • 19. A chromosome of a transgenic animal generated by the method of the foregoing claims.
  • 20. A method for providing cells in culture deriving from a transgenic animal as defined in any one of the foregoing claims.
  • 21. A cell derived from the soma obtained from a transgenic animal as defined in any one of the foregoing claims.
  • 22. Non-human transgenic mammal with genome modified according to the method of any one of the foregoing claims.
  • 23. Transgenic mammal according to claim 22, characterized in that it is a mouse.
Priority Claims (1)
Number Date Country Kind
MI2000A001503 Jul 2000 IT
PCT Information
Filing Document Filing Date Country Kind
PCT/EP01/07622 7/2/2001 WO