MXPA01003808A - G-protein coupled receptors - Google Patents

G-protein coupled receptors

Info

Publication number
MXPA01003808A
MXPA01003808A MXPA/A/2001/003808A MXPA01003808A MXPA01003808A MX PA01003808 A MXPA01003808 A MX PA01003808A MX PA01003808 A MXPA01003808 A MX PA01003808A MX PA01003808 A MXPA01003808 A MX PA01003808A
Authority
MX
Mexico
Prior art keywords
amino acid
nucleic acid
receptor
seq
cell
Prior art date
Application number
MXPA/A/2001/003808A
Other languages
Spanish (es)
Inventor
Maria Alexandra Glucksmann
Nadine S Weich
Original Assignee
Maria Alexandra Glucksmann
Millennium Pharmaceuticals Inc
Nadine S Weich
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Maria Alexandra Glucksmann, Millennium Pharmaceuticals Inc, Nadine S Weich filed Critical Maria Alexandra Glucksmann
Publication of MXPA01003808A publication Critical patent/MXPA01003808A/en

Links

Abstract

The present invention relates to newly identified receptors belonging to the superfamily of G-protein-coupled receptors. The invention also relates to polynucleotides encoding the receptors. The invention further relates to methods using the receptor polypeptides and polynucleotides as a target for diagnosis and treatment in receptor-mediated disorders. The invention further relates to drug-screening methods using the receptor polypeptides and polynucleotides to identify agonists and antagonists for diagnosis and treatment. The invention further encompasses agonists and antagonists based on the receptor polypeptides and polynucleotides. The invention further relates to procedures for producing the receptor polypeptides and polynucleotides.

Description

/ RECEPTORS COUPLED BY PROTEIN G Field of the Invention The present invention relates to newly identified receptors that belong to the superfamily of G-protein coupled receptors. The present invention also relates to polynucleotides that encode the receptors. The present invention further relates to methods that use the polypeptides and receptor polynucleotides as a target for diakosis and treatment in diseases related or transmitted by the receptor. The invention further relates to methods of drug selection that utilize the polypeptides and receptor polynucleotides to identify antagonists and antagonists for diagnosis and treatment. The invention further comprises agonists and antagonists based on the polypeptides and receptor polynucleotides. The invention further relates to methods for producing the polypeptides and receptor polynucleotides.
Background of the Invention G protein-coupled receptors • G protein-coupled receptors (GPCRs) 5 constitute a major class of proteins responsible for the transduction of a signal within a cell. GPCRs have three structural fields: an extracellular field with amino terminal, a transmembrane field containing seven segments of transmembrane, three extracellular spirals and three intracellular spirals, and an intracellular field with carboxy terminal. At the time of the binding of a ligand to an extracellular portion of a GPCR, a signal is transduced into the cell which results in a change in a biological or physiological property of the cell. The GPCRs together with G proteins and performers (intracellular enzymes and channels modulated by G proteins), are the components of a signaling system Modular that connects the condition of the intracellular second messengers with the extracellular inputs. GPCR genes and gene products are potentially disease-causing agents (Spiegel et al. (1993) J. Clin. Invest. 92: pages 1119 to 1125; McKusick and associates (1993) J. Med. Genet. 30: pages 1 to 26). Specific defects in the rhodopsin gene and the vasopressin V2 receptor gene have been shown to cause various forms of retinitis pigmentosum (Nathans and associates (1992) Annu. Rev. Genet. 26: pages 403 to 424), and nephrogenic insipidus diabetes (Holtzman and associates (1993) Hum. Mol. Genet 2: pages 1201 to 1204). These receptors are of critical importance for both the central nervous system and peripheral physiological processes. Evolution analyzes suggest that the ancestor of these proteins was originally developed in combination with complex plans of the body and nervous systems. The superfamily of the GPCR protein can be divided into five families: Family I, receptors typified by rhodopsin and the β2 adrenergic receptor and currently represented by more than 200 unique members (Dohlman and associates (1991) Annu Rev. Biochem. 653 to 688); Family II, the parathyroid hormone / calcitonin / secretin receptor family (Juppner and associates (1991) Science 254: pages 1024 to 1026; Lin and associates (1991) Science 254: pages 1022 to 1024; Family III, the metabotropic glutamate receptor family (Nakanishi (1992) Science 258 597: page 603); Family IV, the cAMP receptor family, important in the chemotaxis and development of D, discoideum (Klein and associates (1988) Science • 241: pages 1467 to 1472); and Family V, fungal mating pheromone 5 receptors such as STE2 (Kurjan (1992) Annu., Rev. Biochem. 61: pages 1097 to 1129). There is also a small amount of other proteins which have seven segments • 10 putative hydrophobic ones and it seems that they are not related to the GPCRs protein; these have not been shown to be coupled to the G proteins. The drosophila expresses a specific protein of the photoreceptor, here is a pair of less than seven (boss), a protein of seven transmembrane seqments which has been extensively studied and shows no evidence of being a GPCR (Hart and associates (1993) Proc. Nati. Acad. Sci. USA 90: pages 5047 to 5051). It is also believed that the spiral curled gene (fz) in the Drosophi is a protein with seven transmembrane segments. Like the leader, fz has not shown to be coupled to the G proteins (Vinson et al. (1989) Nature 338: pages 263 to 264). The G proteins represent a family of heterotrimeric proteins composed of subunits a, β and β, which binds to the guanidine nucleotides. These proteins are generally linked to cell surface receptors, for example, receptors that contain seven transmembrane segments. After binding of the binder to the GPCR protein, a conformational change was transmitted to the G protein, which causes the a subunit to be exchanged, a link with the GDP molecule for a GTP molecule and separated from the β subunits. The bound GTP form of the a subunit generally functions as a modulator portion of the executor, leading to the production of a second messenger, such as cAMP (for example, by activation of the adenyl cyclase), and diacylglycerol or inositol phosphates. In humans, there are more than 20 different types of subunits a. These subunits are associated with a smaller set of β and β subunits. Examples of G proteins in mammals include Gi, Go, Gq, Gs and Gt. G proteins are extensively described in the publication by Lodish and associates Molecular Cell Biology, (Scientific American Books Inc., New York, N. Y., 1995), the content of which is incorporated herein by reference. The GPCRs, G proteins and implementers linked by G protein and the second messenger systems were reviewed in The Fact Book of the Protein G-linked Receptors, Watson and associate editors, Academic Press (1994). Purinoreceptors Purines and especially adenosine and adenine nucleotides have a wide range of pharmacological effects transmitted through cell surface receptors. For a general review, you should consult Adenosine and Adenine Nucleotides (Adenosine and Adenine Nucleotides) in The G-Protein Linked Receptor Facts Book, Watson et al. (Eds.) Academic Press 1994, pages 19 to 31. Some effects of ATP include regulation of soft muscle activity, stimulation of soft intestinal muscle relaxation and contraction of the bladder, stimulation of platelet activation by the ADP when it is released from the vascular endotely, and excitatory effects of the central nervous system. Some effects of adenosine include vasodilation, bronchoconstriction, immunosuppression, inhibition of platelet aggregation, cardiac depression, stimulation of nociceptive afferents, inhibition of neurotransmitter release, pre and postsynaptic depressive action, reduction of motor activity, depressive breathing , the induction of sleep, the release of anxiety, and the inhibition of the release of factors, such as hormones. There are different receptors for adenosine and adenine nucleotides. The clinical actions of such analogues such as methylxanthines, for example, theophylline and caffeine, are considered to affect their effects by antagonizing adenosine receptors. Adenosine has a low affinity for adenine nucleotide receptors, whereas adenine nucleotides have a low affinity for adenosine receptors. There are four accepted subtypes of adenosine receptors designated i, A2A, A2B, and A3. In addition, an A4 receptor based on its brand has been proposed for phenylaminoadenosine 2 (Cornfield et al. (1992) Mol.Pharmacol 42: pages 552 to 561). P2X receptors are cation channels regulated by ATP (see Neuropharmacology 36 (1997)). The proposed topology for P2X receivers are two transmembrane regions, a large extracellular spiral, and an intracellular N and C term. It has been proposed that many cloned receptors designated P2Y are members of the family of G-protein coupled receptors. UDP, UTP, ADP, and ATP have been identified as agonists. To date, P2 ?? - 7 have been characterized although it has been proposed that P2Y7 may be a B4 leukotriene receptor (Yokomizo et al. (1997) Nature 387: pages 620 to 624). However, it is widely accepted that P2Y 1,2,4, and 6 are members of the family coupled by G protein of P2 ?. At least three P2 purinoreceptors of the hematopoietic cell line HEL have been identified by intracellular calcium mobilization and by photoaffinity labeling (Akbar et al. (1996) J.
Biochem. 271: pages 18363 to 18567). The adenosine receptor i was designated in view of its ability to inhibit adenyl cyclase. The receptors are distributed in many peripheral tissues such as the heart, adipose tissue, kidney, stomach and pancreas. It has also been discovered in peripheral nerves, for example intestines and vas deferens. They are also present at high levels in the central nervous system, including the cerebral cortex, the hippocampus, cerebellum, thalamus and striatum, as well as in several cell lines. Agonists and antagonists can be found on page 22 of The G-Protein Linked Receptor Facts Book cited above, and incorporated herein by reference. It has been reported that these receptors inhibit adenyl cyclase and voltage-gated calcium channels and activate potassium channels through a G protein sensitive to pertussis toxin that is suggested to be of the Gi / Grj class. The receptors i have also been reported as inducers of the activation of phospholipase C and how they potentiate the capacity of other receptors to activate this step. Adenosine A2A receptors have been discovered in the brain, such as the striatum, tubercle of olfaction and nucleus accumbenus. In the periphery, A2 receptors transmit vasodilation, immunosuppression, inhibition of platelet aggregation and gluconeogenesis. Agonists and antagonists have been found in The G-Protein Linked Receptor Facts Book cited above on page 25, incorporated herein by reference. This receptor transmits the activation of adenyl cyclase through Gs. The A2B receptor has been shown to be present in the human brain and in the intestine and urinary bladder of the rat. The agonists and antagonists explain on page 27 of The G-Protein Linked Receptor Facts Book cited above, incorporated herein by reference. This receptor transmits the cAMP stimulus through the Gs. The A3 adenosine receptor is expressed in the testes, lung, kidney, heart, central nervous system, including the cerebral cortex, striatum, and the olfactory bulb. An explanation of the agonists and antagonists can be found on page 28 of the Accepted Book of Facts of the G-Protein Linked Receptor Facts Book cited above, incorporated herein by reference. The receptor transmits the inhibition of adenyl cyclase through a G protein that is sensitive to the pertussis toxin that is suggested to be of the Gi / Grj class. The P2 purinoreceptor? shows a similar affinity for ATP and ADP as a lower affinity for AMP. The receptor has been discovered in the soft muscle, for example, taeni caeci and in the vascular tissue where it induces vasodilation through the endothelium-dependent release of nitric oxide. It has also been shown in avian erythrocytes. Agonists and antagonists are explained on page 30 of the Accepted Book of Facts of the G-Protein Linked Receptor Facts Book cited above, incorporated herein by reference. The function of the receptor through the activation of the phosphoinositide metabolism to • through a G protein that is not sensitive to the toxin the whooping cough, which is suggested to be of the class G? / G0. Accordingly, GPCRs, and especially purinoreceptors are major targets for the action and development of the drug. Therefore, it is valuable for the field of pharmaceutical development with • 10 the object of identifying and characterizing previously unknown GPCRs. The present invention anticipates the condition of the material by providing a previously unidentified human GPCR having homology with the purinoreceptors. Summary of the Invention It is an object of the present invention to identify new GPCRs. It is a further object of the present invention, Provide novel GPCR polypeptides that are useful as reagents or targets in receptor assays applicable to the treatment and diakosis of diseases transmitted by GPCR. It is yet another object of the present invention to provide the polynucleotides corresponding to the New GPCR receptor polypeptides which are useful as targets and reagents in receptor assays applicable to the treatment and diagnosis of diseases transmitted by GPCR and useful for producing polypeptides. new receptors by recombinant methods. A specific object of the present invention is to identify compounds that act as agonists and antagonists and modulate the expression of new receptors. It is a further specific object of the present invention to provide compounds that modulate the expression of receptors for the treatment and diagnosis of diseases related to GPCR. Therefore, the present invention is based on the identification of GPCRs, designated as receptors 2838, 14618, and 15334. The present invention provides polypeptides isolated from the '2838 receptor that include a polypeptide having an amino acid sequence shown in SEQ ID NO: 1. The present invention provides polypeptides isolated from receptor 14618 that include a polypeptide having an amino acid sequence shown in SEQ ID NO: 3. The present invention provides polypeptides isolated from receptor 15334 that include a polypeptide having the amino acid sequence shown in SEQ ID NO: 5. The present invention also provides nucleic acid molecules isolated from the 2838 receptor having the sequence shown in SEQ ID NO: 2. The present invention also provides nucleic acid molecules isolated from receptor 14618 having the sequence shown in SEQ ID NO: 4. The present invention also provides nucleic acid molecules isolated from receptor 15334 having the sequence shown in SEQ. ID NO: 6. The present invention also provides variant polypeptides having an amino acid sequence that is substantially homologous to the amino acid sequence shown in SEQ ID NO: 1. The present invention also provides variant polypeptides having a sequence of amino acid that is substantially homologous to the amino acid sequence shown in SEQ ID NO: 3.
The present invention also provides variant polypeptides having an amino acid sequence that is substantially homologous to the amino acid sequence shown in SEQ ID NO: 5. The present invention also provides variant nucleic acid sequences that are substantially homologous to the sequence of nucleotides shown in SEQ ID N0: 2. The present invention also provides variant nucleic acid sequences that are substantially homologous to the nucleotide sequence shown in SEQ ID NO:. The present invention also provides variant nucleic acid sequences that are substantially homologous to the nucleotide sequence shown in SEQ ID NO: 6. The present invention also provides fragments of the polypeptide shown in SEQ ID NO: 1 and nucleotides shown in SEQ ID NO: 2, as well as substantially homologous fragments of the polypeptide or nucleic acid. The present invention also provides fragments of the polypeptide shown in SEQ ID NO: 3 and the nucleotide sequence shown in SEQ ID NO: 4, as well as substantially homologous fragments of the polypeptide or nucleic acid. The present invention also provides fragments of the polypeptide shown in SEQ ID NO: 5 and the nucleotide sequence shown in SEQ ID NO: 6, as well as substantially homologous fragments of the polypeptide or nucleic acid. The present invention also provides nucleic acid constructs comprising the 10 nucleic acid molecules described above. In a preferred embodiment, the nucleic acid molecules of the present invention are operably linked to a regulatory sequence. The present invention also provides, vectors and host cells for the expression of the nucleic acid receptor molecules and polypeptides and particularly recombinant vectors and cells • Guest. The present invention also provides methods for the preparation of vectors and host cells and methods for using them to produce the nucleic acid receptor molecules and polypeptides. The present invention also provides antibodies or antigen binding fragments thereof that selectively bind to polypeptides and receptor fragments. The present invention also provides screening methods for compounds that modulate the expression or activity of the polypeptides or receptor nucleic acid (RNA or DNA). The present invention also provides a process for modulating the expression or activity of the receptor polypeptide or nucleic acid, especially using the selected compounds. The modulation can be used to treat conditions related to activity or aberrant expression of the polypeptides or receptor nucleic acids. The present invention also provides assays for determining the presence or absence and level of the nucleic acid molecules or receptor polypeptides in a biological sample, including disease diakosis. The present invention also provides assays for determining the presence of a mutation in the nucleic acid receptor molecules or polypeptides including diagnosis of the disease. Still in a further embodiment, the present invention provides computer readable means which contain the nucleotide and / or amino acid sequences of the nucleic acids and polypeptides of the present invention, respectively.
Brief Description of the Drawings Figure 1 illustrates a comparison of the 2838 receptor against the Prosite database of protein standards, which specifically shows a high score against the rhodopsin superfamily of ^ 10 seven transmembrane segments. The underlined area illustrates a GPCR signature and specifically the position of an arginine residue, conserved in GPCRs. The most commonly conserved sequence is a triplet of aspartate, arginine, tyrosine (DRY). The DRY is involved in the transduction of the signal. Arginine is not variant. Aspartate is placed conservatively in several GPCRs. In the current case, it has • found the arginine in the DRF sequence, which coincides with the position of the DRY or the arginine not variant in the GPCRs of the rhodopsin receptor superfamily. Figure 2 shows an analysis of the amino acid sequence 2838: the spin and spiral regions, the hydrophilicity; unfriendly regions; flexible regions; antigenic index; and trace of surface probability. Figure 3 shows a trace of hydrophobicity • receptor 2838. The amino acids correspond to 1 to 5 319 and show the seven transmembrane segments. Figure 4 shows an analysis of the open reading frame of 2838 for amino acids corresponding to specific functional sites. A glycosylation site is found in amino acids • 10 5 to 8 which correspond to the extracellular field of amino terminal. A second glycosylation site is found in amino acids 171 to 174 which correspond to the second extracellular spiral. The site of kinase protein phosphorylation C is found in amino acids 134 to 136 which are in the second intracellular spiral. One second The phosphorylation site of the kinase C protein is found in amino acids 178 to 180 which is the second extracellular spiral. A site of kinase II casein phosphorylation is found in amino acids 6 to 9 which are in the intracellular field of the carboxy terminus. A second phosphorylation site of casein kinase II is found at amino acids 95 through 98, which are in the first extracellular spiral. A N-myristoylation site is found in amino acids 34 to 39 which are in the first transmembrane segment. A second N-myristoylation site is found in • amino acids 107 to 112 which are in the 5th third transmembrane segment. A third site N-myristoylation is found in amino acids 140 to 145 which are in the fourth transmembrane segment. An amidation site is found at amino acids 209 to 212 which extends to along the fifth transmembrane segment and the third intracellular spiral. In addition, the corresponding amino acids at the position of the GPCR signature and containing the non-variant arqinin are found in the DRF sequence at amino acids 118 to 120. Figure 5 shows a comparison of the 14618 receptor against the Prosite database of protein patterns, specifically showing a high score against segment seven of transmembrane of the rhodopsin superfamily. The underlined data show a GPCR signature and specifically the position of the arginine residue, conserved in the GPCRs. The most commonly conserved sequence is a triplet aspartate, arginine, tyrosine (DRY). DRY is involved in the transduction of the signal. Arginine is not variant. Aspartate is placed conservatively in several GPCRs. In the present case, arginine is found in the DRF sequence which coincides with the DRY or non-variant arginine position • in the GPCRs of the superfamily of .5 rhodopsin receptors. Figure 6 shows an analysis of amino acid sequence 14618: a spin and spin aß region; hydrophilicity; unfriendly regions; flexible regions; antigenic index; and strokes of surface probability. Figure 7 shows a hydrophobicidal trace of the 14618 receptor. Amino acids correspond from 1 to 337 and show the seven transmembrane segments. Figure 8 shows an analysis of a framework of open reading 14618 for amino acids corresponding to specific functional sites. A (B glycosylation site is found in amino acids 6 to 9 which correspond to the extracellular field of amino terminal. glycosylation is found in amino acids 169 a 172 which correspond to the second extracellular spiral. A third glycosylation site is found in amino acids 180 to 183 which also correspond to the second spiral extracellular. A fourth glycosylation site is found in amino acids 224 to 227 which correspond to the third intracellular spiral. A fifth glycosylation site is found in amino acids 262 to 265 which correspond to the third extracellular spiral. A kinase phosphorylation site of the cAMP-dependent protein and cGMP is found at amino acids 304 to 307, 310 to 313 and 323 to 326 all of them in the intracellular field of the carboxy terminus. A phosphorylation site of the kinase C protein is found in amino acids 136 to 138 which correspond to the second intracellular spiral. The second and third phosphorylation sites of kinase C protein are found in amino acids 220 to 222 and 227 to 229 corresponding to both the third intracellular spiral. A fourth phosphorylation site of the kinase C protein is found at amino acids 308 to 310, corresponding to the intracellular field of the carboxy terminus. The kinase II casein phosphorylation sites are found in amino acids 13 to 16 and 17 to 20, both in the extracellular field of the amino terminal. A third casein phosphorylation site of kinase II is found at amino acids 326 to 329 corresponding to the intracellular field of the carboxy terminus. A C-terminal signal of microbodies is found in amino acids 335 to 338, corresponding to the intracellular field of the • carboxy terminal. In addition, the corresponding amino acids at the position to the GPCR signature and containing non-variant arginine, are found in the FRC sequence at amino acids 120 to 122. Figure 9 shows a comparison of the 15334 receptor against the Prosite database. patterns of • 10 protein, showing specifically, a high score against the seven transmembrane segments of the rhodopsin superfamily. The underlined area shows a GPCR signature, and specifically the positions of an arginine residue, conserved in GPCRs. The most commonly conserved sequence is a triplet aspartate, arginine, tyrosine (DRY). DRY is involved in the transduction of the signal.
• Arginine is not a variant. Aspartate is placed conservatively in several GPCRs. If Currently, arginine is found in the DRY sequence, which coincides with the DRY or non-variant arginine position in the GPCRs of the rhodopsin receptor superfamily. Figure 10 shows a sequence analysis amino acids of the 15334: the aß spiral turn regions; hydrophilicity; unfriendly regions; flexible regions, antigenic index; and a trace of surface probability. Figure 11 shows a hydrophobicity trace of receptor 15334. The amino acids show seven transmembrane segments. Figure 12 shows an analysis of the open reading frame of receiver 15334, for amino acids corresponding to specific functional sites. The glycosylation sites are found in amino acids from 4 to 7 and 9 to 12, which are in the extracellular field of the amino terminal. A glycosylation site is also found in amino acids 251 to 254, which are in the sixth transmembrane segment. An additional glycosylation site is found at amino acids 323 to 326, which are in the carboxy terminal field.
• A protein kinase phosphorylation site dependent on cAMP and cGMP is found in amino acids 229 to 232, which are in the third intracellular spiral. The protein kinase C phosphorylation sites are from amino acids 21 to 23, which correspond to the field of the amino terminal, 211 to 213, 226 to 228 and 232 to 234, which correspond to the third intracellular spiral, and 307 to 309 and 332 to 334 correspond to the intracellular field of the carboxy terminal. A kinase II casein phosphorylation site, flB is found at amino acids 178 through 181 which are in the second extracellular spiral and from 342 to 345 which is in the intracellular field of the carboxy terminal. The N-myristoylation sites are found in amino acids 36 to 41, which are in the extracellular field of the amino terminal, from 258 to ^ 10 263, which extends along the sixth transmembrane segment, and the third extracellular spiral, of the 324 to 329 which correspond to the intracellular field of the carboxy terminal. In addition, a signature GPCR and DRY are found in amino acids 118 to 120 which are in the second intracellular spiral. Figure 13 shows the expression of the receptor ^ * fc 14618 in several human normal tissues. Figure 14 shows the expression of the 14618 receptor in various tissues and in cells of the hematopoietic system. Figure 15 shows the expression of the 2838 receptor in various cells and tissues including cells of the hematopoietic system. Figure 16 shows the expression of the receptor 153343 in normal human tissues.
Figure 17 shows the expression of receptor 15334 in various tissues and cells of the hematopoietic system. Figure 18 shows the expression of receptor 15334 in several cells / hematopoietic cell lines, including erythroid and megakaryocyte cells. Figure 19 shows the expression of receptor 15334 in several cells or hematopoietic cell lines, including erythroid and megakaryocyte cells.
Detailed Description of the Invention. Functional trajectory / receptor signal The receptor proteins 2838, 14618, and 15334 are GPCRs that participate in signaling trajectories. As used in the present invention, a "signaling path" refers to the modulation (eg, stimulation or inhibition) of a cellular activity / function in binding of a ligand to GPCRs (proteins 2838, 14618, or 15334). Examples of such functions include mobilization of intracellular molecules that participate in a signal transduction pathway, for example, phosphatidylinositol 4,5-bisphosphate (PIP2), inositol 1, 5-triphosphate (IP3) and adenylate cyclase; polarization of the plasma membrane; production or secretion of molecules; alteration in the structure of a cellular component; cell proliferation, for example, DNA synthesis; cellular migration; Cell differentiation; and about cellular experience. As the 2838 receptor protein is expressed in the thymus, lymph node, spleen, testes, colon and peripheral blood lymphocytes including but not limited to activated 1 T helper cells, activated 2 helper cells T, CD3 (both CD4 and CD8) , activated B cells, and granulocytes, cells that participate in the signaling path of receptor protein 2838 including, but not limited to, these tissues and cells. As the receptor protein 14618 is expressed in the chest or muscle of the skeleton, thyroid, lymph node, spleen and peripheral blood lymphocytes including, but not limited to CD34 + cells, resting B cells, and megakaryocytes, the cells involved in the signaling pathway of receptor protein 14618 including, but not limited to, cells derived from these tissues and cells. As the receptor protein 15334 is expressed in the colon, pancreas, tonsils, lymph node, spleen, thymus, adrenal gland, heart, and peripheral blood cells including but not limited to those illustrated in Figure 17, megakaryocytes, and erythroblas coughs, the cells that participate in the signaling path of the receptor protein 15334 including, but not limited to, cells derived from these tissues and cells. The response transmitted by the receptor protein depends on the type of cell. For example, in some cells, the binding of a binder to the receptor protein can stimulate an activity such as the release of compounds, the regulation of a channel, cell adhesion, migration, differentiation, etc., through phosphatidylinositol or the cyclic metabolism of AMP and rotation while in other cells, binding of the binder will produce a different result. Regardless of the cellular activity / response modulated by the receptor protein, it is universal that the protein is a GPCR and interacts with the G proteins to produce one or more secondary signals, in a variety of intracellular signal transduction pathways, for example, to through phosphatidylinositol or the metabolism of cyclic AMP and rotation, in a cell.
As used in the present description, "phosphatidylinositol turnover and metabolism" refers to the molecules comprised in the metabolism rotation of phosphatidylinositol 4,5-bisphosphate (PIP2) as well as the activities of these molecules. PIP is a phospholipid found in the cytosolic flake of the plasma membrane. The binding of the ligand to the receptor, activates in some cells, the phospholipase C enzyme of the plasma membrane, which in turn can hydrolyze PIP2 to produce 1,2-diacylglycerol (DAG) and inositol 1,4,5-triphosphate ( IP3). Once formed, IP3 can diffuse to the surface of the endoplasmic reticulum where it can bind to an IP3 receptor, for example, a calcium channel protein containing an IP3 binding site. The IP3 bond can induce the opening of the channel, allowing calcium ions to be released into the cytoplasm. IP3 can also be phosphorylated by a specific kinase to form inositol 1, 3, 4, 5-tetrafosfato (IP4), a molecule which can cause the entry of calcium into the cytoplasm of the extracellular medium. IP3 and IP4 can subsequently be hydrolyzed very rapidly to inactive inositol products 1,4-biphosphate (IP2) and inositol 1,3-triphosphate, respectively. These inactive products can be recycled by the cell to synthesize PIP2. The other second messenger produced by the hydrolysis of PIP2, is flB say 1, 2-diacylglycerol (DAG), remains in the cell membrane, where it can serve to activate kinase C of the enzyme protein. Protein kinase C is generally soluble in the cytoplasm of the cell, but at an increase in intracellular calcium concentration, this enzyme ^ 10 can move to the plasma membrane where it can be activated by the DAG. Activation of protein kinase C in different cells results in several cellular responses such as phosphorylation of glycogen synthase, or phosphorylation of several transcription factors, for example, NF-kB. The term "phosphatidylinositol activity", as used in the present invention, refers to an activity of PIP2 or one of its metabolites. 20 Another signaling path in which the receiver can participate is the cAMP rotation path. As used in the present invention, "rotation and AMP cyclic metabolism" refers to molecules comprised in the rotation and metabolism of AMP cyclic (cAMP) as well as the activities of these molecules. Cyclic AMP is a second messenger produced in response to the stimulus induced by the binder of certain receptors coupled by βpG protein. In the cAMP signaling path, linkage 5 from a binder to a GPCR can lead to the activation of the cyclase adenyl of the enzyme, which catalyzes the synthesis of cAMP. Recently synthesized cAMP can activate the protein kinase dependent on cAMP. This activated kinase ^ 10 can phosphorylate a voltage-regulated potassium channel protein, or an associated protein and lead to the inability of the potassium channel to open during an action potential. The lack of ability of the calcium channel to open results, as a result, in a decrease in the flow of potassium to the outside, which normally repolarizes the membrane of a neuron, leading to prolonged depolarization of the membrane.
Polypeptides The present invention is based on the discovery of the G-protein coupled receptors. Specifically, a sequence tag expressed in (EST) was selected based on the homology with the sequences of the G-protein coupled receptors. This EST was used to design primers based on the sequences it contains, and used to identify a 2838 cDNA from a B cell cDNA library. A cDNA 14618 from a library of cDNA of the liver and spleen and a 15334 cDNA from a spleen cDNA library. Sequences were cloned positive, and overlapping fragments were assembled. The analysis of the assembled sequences revealed that the three cloned cDNA molecules encode the G protein coupled receptors. The present invention therefore refers to New GPCRs having a deduced amino acid sequence illustrated as SEQ ID NO: 1. The present invention therefore also relates to new GPCRs having a deduced amino acid sequence illustrated as SEQ ID NO: 3. Therefore, the present invention also relates to a novel GPCR having a deduced amino acid sequence illustrated in SEQ ID NO: 5. The Sequence List shows the sequence of nucleotide 2838 (SEQ ID NO: 2) and the deduced sequence of amino acid 2838 (SEQ ID NO: 1). It is predicted that amino acids from 1 to approximately 24, constitute an extracellular field of amino terminal, the amino acids from approximately 25 to 292, constitute a region that extends along the transmembrane field and amino acids from approximately 293 to 319 they constitute an intracellular field of the carboxy terminal. The transmembrane field contains seven transmembrane segments, three extracellular coils and three intracellular coils. The transmembrane segments are from about amino acid 25 to f 10 amino acid 49, from about amino acid 56 to about amino acid 79, from about amino acid 100 to about amino acid 117, from about amino acid 138 to about 15 amino acid 159, from about amino acid 187 to about amino acid 210, from about amino acid 224 to about amino acid 248, and from about amino acid 268 to about amino acid 292. Within the region extending along the entire transmembrane field, there are three intracellular and three extracellular coils. The three intracellular coils are from about amino acid 50 to about 25 amino acid 55, from about amino acid 118 to about amino acid 137, and from about amino acid 211 to about amino acid 223. The three extracellular coils are B from about amino acid 80 to about 5 amino acid 99, from about amino acid 160 to about amino acid 186, and from about amino acid 249 to about amino acid 267. The Sequence List shows the sequence of the nucleotide 14618 (SEQ ID NO: 4) and the deduced amino acid sequence 14618 (SEQ ID NO: 3). It is predicted that amino acids from 1 to about 28 constitute an extracellular field of amino terminal amino acids from about 29 to 297 constitute the region that extend along the transmembrane field, and the amino acids from about 298 to 337 constitute an intracellular carboxy terminal field. The transmembrane field contains seven transmembrane segments, three extracellular coils and three intracellular coils. The transmembrane segments are from about amino acid 29 to about amino acid 49, from about amino acid 60 to about amino acid 84, from about amino acid 103 to about amino acid 127, from about amino acid 142 to about amino acid 161, from B about amino acid 194 to approximately amino acid 217, from about amino acid 231 to about amino acid 247, and from about amino acid 276 to about amino acid 297. Within region 10 that extends throughout the entire transmembrane field, there are three intracellular coils and three extracellular The three intracellular coils are from about amino acid 50 to about amino acid 59, from approximately amino acid 128 to about amino acid 141, and from about amino acid 218 to about amino acid 230. The three extracellular coils are from about amino acid 85 to about amino acid 102, from about amino acid 162 to about amino acid 193, and from about amino acid 248 to about amino acid 275.
The Sequence List shows the nucleotide sequence 15334 (SEQ ID NO: 6) and the deduced amino acid sequence 15334 (SEQ ID NO: 5). It has been predicted, that amino acids 1 to 25 constitute an extracellular field of amino terminal, the amino acids of approximately 26 to 299 constitute the region that extends along the length of the transmembrane field and the amino acids of approximately 300 to 372 constitute the intracellular field of carboxy terminal. The transmembrane field contains seven transmembrane seqments, three extracellular coils and three intracellular coils. The transmembrane seqments are from about amino acid 26 to about amino acid 48, from about amino acid 56 to about amino acid 77, from about amino acid 99 to about amino acid 115, from about amino acid 140 to about amino acid 157, from about approximately amino acid 188 to about amino acid 209, from about amino acid 235 to about amino acid 259, and from about amino acid 277 to about amino acid 299. Within the region extending throughout the entire transmembrane field, there are three intracellular spirals and three extracellular spirals. The three intracellular coils are from about amino acid 49 to about amino acid 55, from about amino acid 116 to about amino acid 139, and from about amino acid 210 to about amino acid 234. The three extracellular coils are found from about the amino acid 78 to about amino acid 98, from about amino acid 158 to about amino acid 187, from about amino acid 260 to about amino acid 276. The terms "2838 receptor polypeptide" or "2838 receptor protein" refer to a polypeptide of SEQ ID NO: 1. The terms "polypeptide of receptor 14618" or "receptor protein 14618" refer to the polypeptide of SEQ ID NO: 3. The terms "receptor 15334 polypeptide" or "receptor protein 15334" refer to polypeptide of SEQ ID NO: 5. The term "protein re- ptora "or" receptor polypeptide ", however, additionally includes the numerous variants of the peptides 2838, 14618, or 15334 described herein, as well as the derivatives derived from the total length 2838, 14618, or 15334 polypeptides and variants. Therefore, the present invention provides the isolated or purified 2838, 14618 and 15334 receptor polypeptides and variants or fragments thereof. Polypeptide 2838 is a protein of residue 319 that exhibits three major structural fields. The extracellular field of the amino terminal is identified to be within the residues of 1 to about 24 in SEQ ID NO: 1. The transmembrane field is identified to be within the residues of about 25 to about 292 in SEQ ID NO: 1. The intracellular field of the carboxy terminus is identified as being within the residues from about 293 to 319 in SEQ ID NO: 1. The transmembrane field contains seven segments that extend along the membrane. Transmembrane segments have been discovered from about amino acid 25 to about amino acid 49, from about amino acid 56 to about amino acid 79, from about amino acid 100 to about amino acid 117, from about amino acid 138 to about IB amino acid 159 , from about 5 amino acid 187 to about amino acid 210, from about amino acid 224 to about amino acid 248 and from about amino acid 268 to about approximately amino acid 292. Within the region extending along the entire transmembrane field, there are three intracellular and three extracellular coils. The three intracellular spirals were found from about amino acid 50 to about amino acid 55, from about amino acid 118 to about amino acid 137, and from • approximately amino acid 211 to approximately amino acid 223. The three spirals extracellulars were found from about amino acid 80 to about amino acid 99, from about amino acid 160 to about amino acid 186, and from about amino acid 249 to about approximately amino acid 267. The transmembrane field includes a signal transduction signature GPCR, DRF, at residues 118 to 120. The sequence includes an arginine at residue 119 and a non-variant amino acid in GPCRs. Based on a BLAST search, the highest homology with the purinoreceptors was shown. Polypeptide 14618 is a protein of residue 337 that exhibits three major structural fields. The extracellular field of the amino terminal is identified to be within residues 1 to about 28 in SEQ ID NO: 3 The transmembrane field is identified as being within the residues of about 29 to about 297 in SEQ ID NO: 3. The intracellular field of the carboxy terminal is identified as being within the residues of about 298 to 337 in SEQ ID NO: 3. The transmembrane field contains seven segments extending as far as possible. length of the membrane. Transmembrane segments have been found approximately from amino acid 29 to approximately amino acid 49, from about amino acid 84 to about amino acid 60, from about amino acid 103 to about amino acid 127, from about amino acid 142 to about amino acid 161, from approximately amino acid 194 to about amino acid 217, from about amino acid 231 to about amino acid 247 and from about amino acid 276 to about amino acid 297. Within the region extending through the entire transmembrane field, there are three cell and three extracellular spirals. The three intracellular coils are from about amino acid 50 to about amino acid 59, from about amino acid 128 to about amino acid 141 and from about amino acid 218 to about amino acid 230. The three extracellular coils were found from about amino acid 85 to about amino acid 102, from about amino acid 162 to about amino acid 193 and from about amino acid 248 to about amino acid 275. The transmembrane field includes a signal transduction signature GPCR, FRC in residues 121 through 123. The sequence includes an arginine at residue 122, a non-variable amino acid in GPCRs. Based on the BLAST search, the highest homology with the purinoceptors was shown. Polypeptide 15334 is a protein of residue 372 that exhibits three major structural fields. The extracellular field of the amino terminal is identified within residues 1 to about 25 of SEQ ID NO: 5. The transmembrane field is identified within the residues of about 26 to about 299 in SEQ ID NO: 5. The field intracellular carboxy terminal is identified within the residues of about 300 to 372 in SEQ ID NO: 5. The transmembrane field contains seven segments that extend along the membrane. The transmembrane segments are from about amino acid 26 to about amino acid 48, from about amino acid 56 to about amino acid 77, from about amino acid 99 to about amino acid 115, from about amino acid 140 to about amino acid 157, from about amino acid 188 to about amino acid 209, from about amino acid 235 up to about amino acid 259 and from about amino acid 277 up ^ approximately amino acid 299. Within the region which extends along the entire transmembrane field, there are three intracellular and three extracellular coils. The three intracellular coils are from about amino acid 49 to about amino acid 55, ^ 10 from about amino acid 116 to about amino acid 139, and from about amino acid 210 to about amino acid 234. The three extracellular coils were found from about amino acid 78 to about amino acid 98, from about amino acid 158 to about amino acid 187, and from • about amino acid 260 to about amino acid 276. The transmembrane field includes a signal transduction signature GPCR, DRY, at residues 118 to 120. The sequence includes an arginine at residue 119, a non-variant amino acid in the GPCRs. Based on the BLAST search, the homology more showed up with the purinoceptors.
As used in the present disclosure, a polypeptide is said to be "isolated" or "purified" when it is substantially free of cellular material is flp when it is isolated from recombinant and non-combinant cells, or free from chemical precursors or other chemicals, when chemically synthesized, however, a polypeptide may be linked to another polypeptide with which it is not normally associated in a cell and can still be considered "isolated" or "purified". The receptor polypeptides can be purified to homogeneity. However, it is understood that preparations in which the polypeptide is not purified to homogeneity are useful and considers that they contain an isolated form of the polypeptide. The critical characteristic in that the preparation allows the desired function of the polypeptide, even in the presence of considerable amounts of other components. Therefore, the The present invention comprises several purities of purity. In one embodiment, the term "substantially free of cellular material" includes preparations of the receptor polypeptide having less than about 30% (dry weight) other proteins (for example contaminating protein), and less than about 20% of other proteins, less than about 10% of other proteins or less than about 5% of other proteins. When the receptor polypeptide is produced in a recombinant, can also be substantially free of the culture medium, for example the culture medium represents less than about 20%, less than about 10% or less than about 5% of the volume of the preparation of ^ P 10 protein. A receptor polypeptide is also considered isolated, when it is part of a membrane preparation or is purified and then reconstituted with the vesicles or liposomes of the membrane. The term "substantially free of chemical precursors or other chemicals" includes preparations of receptor polypeptide, which is separated from • chemical precursors or other chemicals that are included in their synthesis. In one modality, the The term "substantially free of chemical precursors or other chemicals" includes polypeptide preparations having less than about 30% (in dry weight) of chemical precursors or other chemicals, less than about 20% chemical precursors or other chemicals, less than about 10% chemical precursors or other chemicals, or less than about 5% chemical precursors or other chemicals. ß In one embodiment, the receptor polypeptide comprises the amino acid sequence shown in SEQ ID NO: l. However, the present invention also comprises variants of the sequence. The variants include a substantially homologous protein encoded by the same genetic locus in a ^ P 10 organism, for example, an allelic variant. The receiver is traced to chromosome 2, close to WI-7921. The variants also comprise proteins derived from other genetic loci in an organism, but which have a substantial homology with the receptor protein 2838 of SEQ ID NO: 1. The variants also include proteins substantially homologous to the 2838 receptor protein, but derived from another organism, for example, an ortholog. The variants also include proteins that are substantially homologous to the 2838 receptor protein that are produced by chemical synthesis. The variants also include proteins that are substantially homologous to the 2838 receptor protein that are produced by recombinant methods. Without However, it should be understood that the variants exclude any amino acid sequences disclosed before the present invention. In another embodiment, the receptor polypeptide comprises the amino acid sequence shown in SEQ ID NO: 3. However, the present invention also comprises variants of the sequence. The variants include allelic variants. The variants also comprise proteins derived from another genetic loci in an organism, but which have a substantial homology with the receptor protein 14618 of SEQ ID NO: 3. The variants also include substantially homologous orthologs. Variants also include proteins that are substantially homologous to the 14618 receptor protein that are produced by chemical synthesis. The variants also include proteins that are substantially homologous to the 14618 receptor protein, which are produced by recombinant methods. It should be understood, however, that the variants exclude any amino acid sequences that have been disclosed before the present invention. In another embodiment, the receptor polypeptide comprises the amino acid sequence shown in SEQ J D NO: 5. However, the present invention also encompasses variants of the sequence. The variants include allelic variants. The receptor is traced to chromosome 12 in close proximity to SHGC-30262. The variants also comprise proteins derived from another genetic loci in an organism, but 5 having a substantial homology to the receptor protein 15334 of SEQ ID NO: 5. Variants also include substantially homologous orthologs. The variants also include proteins that are substantially homologous to the receptor protein 15334, which are produced by chemical synthesis. Variants also include proteins that are substantially homologous to the 15334 receptor protein, in that they are produced by recombinant methods. However, it must be understood that the variants exclude any amino acid sequences disclosed before the present invention. As used in the present invention, two • proteins (or a region of proteins) are substantially homologous to the 2838 protein when The amino acid sequences are at least about 40 to 45%, 45 to 50%, 50 to 55%, 55 to 60%, generally at least about 70 to 75%, more generally at least about 80 to 85% and even more generally of at least about 90 to 95% or more homologs. A substantially homologous amino acid sequence, according to the present invention, will be encoded by a nucleic acid sequence that hybridizes to the nucleic acid sequence, or a portion thereof of the sequence shown in SEQ ID NO: 2. severe conditions as will be described in more detail below. As used in the present invention, two proteins (or a region of the proteins) are ^ P 10 substantially homologous to the 14618 protein, when the amino acid sequences are at least about 35 to 40%, 40 to 45%, from 45 to 50%, from 55 to 60%, generally from at least approximately 70 to 75%, more generally from At least about 80 to 85%, and even more generally at least about 90 to 95% or more homologous. A sequence of amino acids • Substantially homologous, according to the present invention, will be encoded by an acid sequence Nucleic acid that hybridizes to the nucleic acid sequence, or a portion thereof, of the sequence shown in SEQ ID NO:. Under severe conditions as will be explained in more detail later. As used in the present invention, two proteins (or a region of the proteins) are substantially homologous to the 15334 protein when the amino acid sequences are at least about 40 to 45%, 45 to 50%, 50 to 55%, 55 to 60 %, generally of at least about 70 to 75%, more generally of at least about 80 to 85% and even more generally of at least about 90 to 95% or more homologs. A substantially homologous amino acid sequence, according to the present invention, will be encoded by a nucleic acid sequence that hybridizes to the nucleic acid sequence or a portion thereof, of the sequence shown in SEQ ID NO: 6 under conditions severe as will be described in more detail later. In order to determine the percentage of homology of two amino acid sequences, or of two nucleic acids, the sequences are aligned for purposes of optimal comparison (for example, spaces can be introduced in the sequence of a protein or a nucleic acid for the optimal alignment with another protein or nucleic acid). The residues or nucleotides are then compared at the corresponding amino acid or nucleotide positions. When a position in a sequence is occupied, the same amino acid residue or nucleotide as in the corresponding position in the other sequence, then the molecules are homologous in that position. As used in the present invention, the "homology" of amino acids or nucleic acid is equivalent to the "identity" of the nucleic acid or amino acids. The percentage of homology between the two sequences in a function of the number of identical positions shared by the sequences (for example the percentage of homology is equal to the number of identical positions / total number of positions 100 times). The invention also encompasses polypeptides which have a lower degree of identity, but which are sufficiently similar to carry out one or more of the same functions performed by polypeptides 2838, 14618 or 15334. The similarity is determined by the substitution conserved of amino acids. Said substitutions are those that substitute a certain amino acid in a polypeptide for another amino acid of similar characteristics. Conservative substitutions are likely to be phenotypically silent. Generally, replacements are seen as conservative substitutions, one for the other, between the aliphatic amino acids Ala, Val, Leu e lie; the exchange of the hydroxyl residues Ser and Tr, the exchange of the acid residues Asp and Glu, the substitution between the amide residues Asn and Gln, the exchange of the basic residues Lis and Arg and the replacements between the aromatic residues Fe, Tir. The guidance regarding which amino acid changes are likely to be phenotypically silent is found in Bowie and Associates (1990 Science 547: pages 1306 to 1310).
TABLE 1. Conservative Substitutions of Amino Acids Aromatic Phenylalanine Tryptophan Tyrosine Hydrophobic Leucine Isoleucine Polar Valine Glutamine • 10 Basic Asparagine Arginine Lysine Histidine Acid Aspartic Acid 15 Glutamic Acid Small Alanine Serine • Threonine Methionine 20 Glycine.
Both identity and similarity can be easily calculated (Computer Biology Molecular [Computational Molecular Biology, Lesk, A.M., ed., Oxford University Press, New York, 1998]); Biocomputing: Computer and Genome Projects, (Biocomputing: Informatics and Genome Projects, Smith, D.W., ed., Academic Press, New York, 1993); Analysis Computerized Sequence Data, Part 1, (Computer Analysis of Sequence Datam Part 1, Griffin, A.M., and Griffin, H.G., eds., Humana Press, New Jersey, 1994); Analysis of the Sequence in Molecular Biology, (Sequence Analysis in Molecular Biology, von Heinje, ^ P 10 G., Academic Press, 1987); and Preparation of Analysis of Sequence, (Sequence Analysis Primer, Gribskov, M. And Devereux, J., eds., M Stockton Press, New York, 1991). A preferred non-limiting example of said mathematical alquorithm is described by Karlin and Associates 15 (1993) in Proc. Nati Acad. Sci. USA 90: pages from 5873 to 5877. This algorithm is incorporated, in the NBLAST in the NBLAST and XBLAST programs (version 2.0) • as described by Altschul and Associates (1997) Nucleic Acids Resistance. 25: pages 3389 to 3402. 20 When the BLAST and Gapped BLAST programs are used, the defaul parameters of the respective programs (for example NBLAST) can be used. See http: / www. ncbi .nlm.nih.gov. In one embodiment, the parameters for the comparison of sequences can be established in a score equal to 100, word length = 12, and can be varied (for example W = 5 or W = 20). In a preferred embodiment, the percent identity between two amino acid sequences is determined using the algorithm of Needleman and Associates (1970) (J. Mol. Biol. 48: pages 444-453) which has been incorporated into the GAP program. in the GCG software package (which can be obtained at http: / ww.gcg.com), using either the BLOSUM ^ P 10 62 matrix or a PAM250 matrix, and an aperture weight of 16, 14, 12, 10 , 8, 6 or 4 and a length weight of 1, 2, 3, 4, 5, or 6. Still in another preferred embodiment, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (Devereux and Associates (1984) Nucleic Acid Resistance 12 (1): page 387) (available at http: / www. Gcg. Com), using a • NWS gapdna matrix. CMP and an opening weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6. Another preferred non-limiting example of the mathematical algorithm used for the comparison of the sequences is the algorithm of Myers and Miller, CABIOS (1989). Said algorithm is incorporated in the ALING program (version 2.0) which is part of the CGC sequence alignment software package. When the ALING program is used for the comparison of amino acid sequences, the weight residue table PAM120 can be used, a penalty of 12 of the length of the opening, and an opening penalty of 4. Additional algorithms for sequence analysis are known in the art and include the ADVANCE and ADAM as described by Torellis and Associates (1994) Comput. Appl.
Biosci. 10: pages from 3 to 5; and FASTA described in Pearson and Associates (1988) PNAS 85: pages 2444 to 2448. A variant polypeptide may differ in amino acid sequence by one or more substitutions, cancellations, insertions, investments, mergers and truncations or combination of any of these. The variant polypeptides can be • completely functional, or lacking function in one or more activities. Therefore, in the current case, The variations may affect the function such as, for example, one or more of the regions corresponding to the binder bond, membrane association, G protein binding and signal transduction. The fully functional variants generally contain only a non-critical conservative variation or a non-critical variation of residues or a non-critical region. The functional variants may also contain the substitution of similar amino acids which do not result in a change or result in a negligible change in function. AlternativelySuch substitutions can affect the function in a positive or negative way to some degree. Non-functional variants generally contain one or more substitutions, withdrawals, insertions, inversions, truncations or substitutions, insertions, investments or non-conservative withdrawals in a critical waste or in a critical region. As indicated, the variants may occur naturally or may be originated by recombinant means or chemical synthesis to produce useful and novel features for the receptor polypeptide. This includes avoiding the immunogenicity of the pharmaceutical formulations by preventing protein aggregation. Useful variations also include alteration of binder bond characteristics. For example, one embodiment comprises a variation in the binding site that results in a link but not a release, or a slower binding of the binder. A further useful variation in the same sites may result in a greater affinity for the binder. Useful variations also include changes that provide affinity for another binder. Another useful variation includes one that allows the link but prevents activation by the binder. Another useful variation includes the variation in the G protein signal / binding transduction field of the transmembrane which provides a reduced or increased linkage by the appropriate G protein or by the linkage by a different G protein with which it is generally associated. receiver. Another useful variation provides a fusion protein in which one or more fields or subregions are operatively fused to one or more fields or sub-regions of another G protein-coupled receptor. The amino acids that are essential for the function to be identified by methods known in the art, such as site-directed mutagenesis or alanine scanning mutagenesis (Cunningham and Associates (1989) Science 244: pages 1081 to 1085). This last procedure introduces simple mutations of alanine in each residue of the molecule. The resulting mutant molecules are then subjected to biological testing such as binding to the receptor or in vi tro, or proliferative activity in vi tro. Sites that are critical for binding the receptor to the binder can also be determined by structural analysis such as crystallization, nuclear magnetic resonance, or photoaffinity labeling (Smith and Associates (1992) J. Mol. Biol. 224: pages 899 to 904; de Vos y Asociados (1992) Science 255: pages 306 to 312). 10 Substantial homology may also be for the entire nucleic acid or amino acid sequence, or for fragments of these sequences. The invention therefore also includes, the polypeptide fragments of the receptor protein 2838, fragments that can be derived from the amino acid sequence shown in SEQ ID NO: 1. However, the invention also comprises fragments of the variants of the receptor protein 2838 as described in the present invention. Therefore, the present invention also includes polypeptide fragments of receptor protein 14618. Fragments can be derived from the amino acid sequence illustrated in SEQ ID NO: 3. However, the present invention comprises also fragments of the variants of the receptor protein 14618 as described in the present invention. Therefore, the present invention includes fragments of polypeptides of the receptor protein 15334. Fragments that can be derived from the amino acid sequence shown in SEQ ID NO: 5. However, the present invention also comprises fragments of the variant of the 15334 receptor protein as described herein • 10 invention. The fragments to which the present invention pertains, however, should not be construed as comprising fragments that may have been described prior to the present invention. 15 The fragments may retain one or more of the biological activities of the protein, for example, the ability to bind to a G protein or binder, • as well as fragments that can be used as immunogens to generate receptor antibodies. 20 The biologically active fragments of the 2838 receptor (peptides which are, for example, of a length of 8, 12, 15, 20, 30, 35, 36, 37, 38, 39, 40, 50, 100 or more amino acids) they can comprise a field or pattern, for example, an extracellular field or spiral or intracellular, one or more transmembrane segments, or part thereof, a binding site for the G protein, a GPCR signature, qylosylation sites, phosphorylation sites of protein kinase C and flp kinase II, casein N-myristoylation and 5 amidation sites. These fields or patterns can be identified through computerized routine homologation search procedures. Possible fragments include, but are not limited to: 1) soluble peptides comprising the P ^ complete extracellular field of the amino terminus of amino acid 1 to about amino acid 24 of SEQ ID NO: 1, or parts thereof; 2) peptides comprising the complete intracellular field of the carboxy terminal of about amino acid 293 to amino acid 319 of SEQ ID NO: 1, or part thereof; 3) polypeptides comprising the reqion extending along the entire transmembrane field from about amino acid 25 to about amino acid 292, or part of the same; 4) any of the specific transmembrane segments or portions thereof, from about amino acid 25 to about amino acid 49, from about amino acid 56 to about amino acid 79, from about amino acid 100 to about amino acid 117, from about amino acid 138 to about amino acid 159, from • approximately amino acid 187 up approximately amino acid 210, from about amino acid 224 to about amino acid 248, and from about amino acid 268 to about amino acid 292; 5) any of ^ 10 the three intracellular coils or the three extracellular coils, or part thereof, from about amino acid 50 to about amino acid 55, from about amino acid 118 to about 15 amino acid 137, from about the amino acid 211 to about amino acid 223, from about amino acid 80 to about amino acid 99, from about amino acid 160 to about amino acid 186, and from about amino acid 249 to about amino acid 267. The fragments further include combinations of the above fragments, such as an amino terminal field, combined with one or more transmembrane segments and the extra or intracellular present coils or one or more transmembrane seqments, and the intra or extracellular spirals present, plus the carboxyl terminal field. In this way, any of the above fragments can be combined. Other fragments include the mature protein from about amino acid 6 to 319. Other fragments contain various functional sites described herein, such as phosphorylation sites, glycosylation sites and myristoylation sites and a sequence containing the GPCR signature sequence. for example in the fragments they may extend in one or both directions from the functional site to comprise amino acids 5, 10, 15, 20, 30, 40, 50, or up to 100. In addition, fragments may include subfragments of specific fields mentioned previously, whose subfragments retain the function of the field from which they were derived. The fragments also include amino acid sequences greater than 7 amino acids from amino acid 1 to approximately amino acid 264. Fragments also include antigenic fragments and specifically those that have been shown to have an anti-genic or high index in Figure 2.
Accordingly, possible fragments include fragments that define liqueur binding sites, fragments that define a glycosylation site, flB fragments that define a membrane association, fragments that define phosphorylation sites, fragments that define interaction with G protein and signal transduction, and fragments that define myristoylation sites. This is why it is intended to include the separate fragments that make the ^ P 10 relevant function or allows the relevant function. In a preferred embodiment, the fragment contains the binding site of the binder. Biologically active fragments of the 14618 receptor (polypeptides which for example are 9, 12, 15, 20, 30, 35, 36, 37, 38, 39, 40, 50, 100 or more amino acids in length) can comprise a field or pattern, for example an extracellular field or spiral • or intracellular, one or more transmembrane segments or part thereof, a G-protein binding site, or a GPCR signature, glycosylation sites, dependent on cAMP and cGMP, kinase C protein phosphorylation sites and casein kinase II. Possible fragments include, but are not limited to: 1) soluble peptides comprising the complete extracellular field of the amino terminus near from amino acid 1 to about amino acid 28 of SEQ ID NO: 3, or part thereof; 2) peptides comprising the complete intracellular P field of the carboxy terminus from amino acid 298 to amino acid 337 of SEQ ID NO: 3 or part thereof; 3) peptides comprising the region extending along the entire transmembrane field from about amino acid 29 to about amino acid 297, or • 10 parts of them; 4) any of the specific transmembrane segments or part thereof, from about amino acid 29 to about amino acid 49, from about amino acid 60 to about amino acid 84, from about amino acid 103 to about amino acid 127, from about amino acid 142 to about amino acid 161, from about amino acid 194 to approximately amino acid 217, from about amino acid 231 to about amino acid 247, and from about amino acid 276 to about amino acid 297; 5) any of the three intracellular coils or the three extracellular coils or part thereof; from about amino acid 50 to amino acid 59, from about amino acid 128 to about fj) amino acid 141, from about amino acid 218 to about amino acid 230, from about amino acid 85 to about amino acid 102, from about amino acid 162 to approximately amino acid 193, and since approximately amino acid 248 to approximately amino acid 275. The fragments further include combinations of the above fragments, such as an amino terminal field combined with one or more transmembrane segments and the extra or intracellular spirals present or one or more transmembrane segments, and the intra or extracellular spirals present plus the carboxy terminal field. In this way, any of the above fragments can be combined. Other fragments can include the mature protein from about amino acid 6 to 337. Other fragments contain the different functional sites described herein, such as phosphorylation sites, glycosylation sites, and a sequence containing the signature sequence GPCR. Fragments, for example, may extend in one or both directions from the functional site to comprise 5, 10, 15, 20, 30, 40, 50, or up to 100 amino acids. In addition, fragments can include subfragments of the specific fields mentioned above, whose subfragments retain the function of the field from which they have been derived. The fragments also include amino acid sequences greater than 8 amino acids from amino acid 1 to approximately amino acid 244. The fragments also include antigenic fragments and specifically those that have been shown to have a high antigenic index in Figure 2. Accordingly, the possible fragments include fragments that define a binding sites to the ligand, fragments that define a glycosylation site, fragments that define the association of the membrane, fragments that define phosphorylation sites, fragments that define the interaction with G proteins and signal transduction, and fragments that define myristoylation sites. By means of this a separate fragment is attempted which provides the identification of the relevant function or allows the relevant function. In a preferred embodiment, the fragment contains the binding site to the binder.
Biologically active fragments (peptides which are, for example, of a length of 8, 12, 15, 20, 30, 35, 36, 37, 38, 39, 40, 50, 100 or more amino acid) can comprise a field or pattern, for example an extracellular or intracellular field or spiral, one or more transmembrane segments, or part thereof, binding site for the G protein, or signature GPCR, glycosylation sites, Ca p, cGMP, phosphorylation sites of kinase C, protein and kinase II casein, and N-myristoylation sites. Possible fragments include but are not limited to: 1) soluble peptides comprising the complete extracellular field of the amino terminus from about amino acid 1 to about amino acid 23 of SEQ ID NO: 5, or part thereof; 2) polypeptides comprising the complete intracellular field of the carboxy terminus from about amino acid 300 to amino acid 372 of SEQ ID NO: 5, or part thereof; 3) peptides comprising the reqion extending along the entire transmembrane field from about amino acid 26 to about amino acid 299 or part thereof; 4) any of the specific transmembrane segments or parts thereof, from about amino acid 26 to about amino acid 48, from about amino acid fl 56 to about amino acid 77, from about 5 amino acid 99 to about amino acid 115 , from about amino acid 140 to about amino acid 157, from about amino acid 188 to about amino acid 209, from approximately amino acid 235 to about amino acid 259, and from about amino acid 277 to about amino acid 299; 5) any of the three intracellular spirals or the three spirals extracellular or part thereof. From amino acid 49 to amino acid 55, from about amino acid 78 to about • amino acid 98, from about amino acid 116 to about amino acid 139, from approximately amino acid 158 to about amino acid 187, from about amino acid 210 to about amino acid 234, and from about amino acid 260 to about The fragments further include combinations of the above fragments, such as an amino terminal field combined with one or more transmembrane segments and the extra B or extracellular present B or one or more 5 transmembrane segments, and the coils present. intra or extracellular plus the carboxy terminal field. Therefore, any of the above fragments can be combined. Other fragments include the mature protein from amino acid 6 • 10 through 372. Other fragments contain the different functional sites described herein, such as phosphorylation sites, qyllosylation sites and myristoylation sites and a sequence containing the GPCR signature sequence. For example, the fragments may extend in one or both directions from the functional site to comprise 5, 10, 15, 20, 30, 40, 50 or up to 100 amino acids. In addition fragments may include subfragments of the specific fields mentioned above, whose subfragments retain the function of the field from which they are derived. The fragments also include amino acid sequences greater than 7 amino acids.
The fragments also include antigenic fragments and specifically those that show a high antigenic index in Figure 2. • These regions can be identified by 5 well-known methods, which comprise the computerized homology analysis. Accordingly, possible fragments include fragments that define a binding site to the binder, fragments that define a glycosylation site, ^ P 10 fragments that define a membrane association, fragments that define phosphorylation sites, fragments that define the interaction with the G protein and signal transduction, and fragments that define myristoylation sites. Thus, a separate fragment is attempted which provides that the relevant function is identified or allows the relevant function. In a preferred embodiment, the fragment contains the binding site to the binder. The present invention also provides fragments of the 2838 receptor with immunogenic properties. These contain the carrier portion of the epitope of the receptor protein 2838 and the variants. These epitope-bearing peptides are useful for increasing antibodies that specifically bind to a receptor polypeptide or region or fragment. These polypeptides may contain at least 8, 12, at least 14, or between at least about 15 to about 30 • amino acids . The present invention also provides fragments of the 14618 receptor with immunogenic properties. These contain a carrier portion of the epitope of the 14618 receptor protein and variants. These peptides carrying the epitope are useful for raising antibodies that specifically bind to a receptor polypeptide or a region or fragment. These peptides may contain at least 9, 12, at least 14 or between at least about 15 to about 30 amino acids. The present invention also provides 15334 receptor fragments with properties • Immunogenic. They contain a carrier portion of the epitope of the receptor protein 15334 and variants. These epitope-bearing peptides are useful for increasing antibodies that specifically bind to a receptor polypeptide or region or fragment. These peptides may contain at least 8, 12, at least 14 or between at least about 15 to about 30 amino acids. Non-limiting examples of the antigenic flP polypeptides that can be used to generate Antibodies include peptides derived from the extracellular field of the amino terminus or any of the extracellular coils. Regions that have a high index of antigenicity are illustrated in the Figures 2, 6, and 10. ^ P 10 The epitope-bearing receptor and the polypeptides can be produced by conventional means (Houghten (1985) Proc. Nati. Acad. Sci. USA 82: pages 5131 to 5135). The simultaneous synthesis of multiple polypeptides is described in the patent North American No. 4,631,211. The fragments can be separated (not fused to other amino acids or polypeptides) or they can be • inside a larger polypeptide. In addition, several fragments can be understood within a single larger polypeptide. In one embodiment, a fragment designed for expression in a host may have heterologous pre and pro polypeptide regions fused to the amino acid terminus of the receptor fragment and an additional region fused to the carboxyl terminus of the fragment.
Therefore, the present invention provides chimeric or fusion proteins. These comprise a receptor protein operably linked to a heterologous flB protein having a sequence of 5 amino acids that is not substantially homologous to the receptor protein. "operably linked" indicates that the receptor protein and the heterologous protein are fused to the structure. The heterologous protein can be fused to the N terminus or to the C-terminus of the receptor protein. In one embodiment, the fusion protein does not affect the function of the receptor itself. For example, the fusion protein can be a fusion protein by GST in which the receptor sequences are fused at the C terminus of the GST sequences. Other types of fusion proteins include, but are not limited to, enzymatic fusion proteins, eg, beta-galactosidase fusions, GAL two-hybrid yeast fusions, poly-His fusions and fusions Ig. Said fusion proteins, particularly poly-His fusions, can facilitate the purification of the recombinant receptor protein. In certain host cells (e.g., mammalian host cells), the expression and / or secretion of a protein may be increased using a heterologous signal sequence. Therefore, in another embodiment, the fusion protein contains a heterologous signal sequence at its N. VP terminus. EP-A-0 464 533 describes proteins of fusion comprising several portions of constant immunoglobulin regions. Fc is useful in therapy and diagnosis and therefore results, for example, in improved pharmacokinetic properties (EP-A 0232 262). In the discovery of medicines by • For example, human proteins have been fused with Fc portions for the purpose of high throughput screening assays to identify antagonists. Bennett and Associates (1995) J. Mol. Pick up 8: pages 52 to 58 and Johanson and Associates (1995) J. Biol. Chem. 270, 16: pages 9459 to 9471. Thus, the present invention also comprises soluble fusion proteins containing a receptor polypeptide and several portions of the constant region of heavy or light immunoglobulin chains of different subclasses (IgG, IgM, IgA, IgE). Preferred as immunoglobulin is the constant part of the heavy chain of human IgG, particularly IgG1, where the fusion takes place in a binding region. For some uses it is desirable to remove the Fc after that the fusion protein has been used for its intended purpose, for example, when the fusion protein is to be used as an antigen for immunizations. In a particular embodiment, the flB Fc part can be removed in a simple way by a washing sequence, which is also incorporated and can be washed with factor Xa. A chimeric or fusion protein can be produced by standard recombinant DNA techniques. For example, DNA fragments encoders of different protein sequences are ligated together with the structure according to conventional techniques. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated synthesizers. of DNA. Alteratively, PCR amplification of the gene fragments can be performed using anchor primers, which elevate the complementary projections between two consecutive gene fragments, which can subsequently be recosted and re-amplify them to generate a chimeric gene sequence (see Ausubel and Associates (1992), Current Protocols in Molecular Biology [see Ausubel et al., (1992) Current Protocols in Molecular Biology]). In addition, they can be obtained commercially Many expression vectors already encode a fusion portion (eg, a GST protein). A nucleic acid encoding the receptor protein can be cloned into one of said expression vectors such that the fusion portion is linked in the structure to the receptor protein. Another form of fusion protein is one that directly affects the functions of the receptor. Accordingly, a receptor polypeptide is comprised by the present invention, in which one or more of the P ^ or receptor fields (or part thereof) have been replaced by homologous fields (or part thereof) of another coupled receptor. by G protein or another type of receptor. Accordingly, several swaps are possible. The extracellular field of the terminal Amino or a sub-region thereof (for example that of the binding to the binder) can be replaced by the field or sub-region of another B-receptor-binding protein. • binder Alternatively, the entire transmembrane field can be replaced, or any of the 7 segments or spirals or part thereof, for example, the G protein binding / signal transduction. Finally, the intracellular carboxy terminal field or a subregion can be replaced. Therefore, chimeric receptors can be formed in which one or more of the native fields or sub-regions have been replaced. The isolated 2838 receptor protein can be purified from cells that are naturally expressed, such as lymph node, thymus, vessel, testes, colon and peripheral blood lymphocytes or from those cells in which it is expressed importantly as illustrated in Figure 15, such as auxiliary cells • 10 activated T (1 and 2) hypoxic Hep 3B cells, CD3 cells (both CD4 and CD8) activated B cells, Jurkat cells, among others purified from cells that have been altered to express it (recombinant) or synthesized using methods of synthesis of known proteins. The isolated 14618 receptor protein can be purified from cells expressing it from • natural way, such as breast cells, skeletal muscle, lymph node, vessel and peripheral blood lymphocytes, as well as those tissues in which the gene is expressed in a significant manner as illustrated in Figures 13 and 14 including, but not limited to CD34 + cells and megakaryocytes, purified from cells that have been altered to express it (recombinant) or synthesized using known protein synthesis methods. The 15334 receptor protein isolated can be purified ßp from cells that express it in a natural way such as cells of the colon, placenta, pancreas, tonsils, lymph node, vessel, peripheral blood cells, thymus, adrenal glands and heart , as well as those tissues shown in Figures 16 through 19, including K562 cells, • 10 erythroblasts, and megakaryocytes, purified from cells that have been altered to express them (recombinant) or synthesized using known protein synthesis methods. In one embodiment, the protein is produced by recombinant DNA techniques. For example, the nucleic acid molecule encoding the receptor polypeptide is cloned into an expression vector, the • Expression vector is introduced into a host cell and the protein expressed in the host cell.
The protein can then be isolated from the cell by an appropriate purification scheme using standard protein purification techniques. Frequently polypeptides contain amino acids different from the 20 amino acids that we refer to generally as an amino acid that occurs naturally 20. In addition, many amino acids, including terminal amino acids, W? they can be modified by natural processes, such as processing and other post-translational modifications, or by chemical modification techniques well known in the art. The common modifications that occur naturally in polypeptides are described in the texts ^ 10 basic, detailed monographs and research literature and are well known to those experts in the field. Accordingly, the polypeptides also comprise derivatives or analogs in which the 15 substituted amino acid residues are not one encoded by the genetic code, in which a substituent group is included, in which the mature polypeptide is fused with another compound, such as a compound for increasing the half-life of the polypeptide (eg, polyethylene glycol) or to which additional amino acids are fused to the mature polypeptide, such as a major or secretory sequence or a sequence for the purification of the mature polypeptide or a pro-protein sequence .
Known modifications include, but are not limited to, acetylation, acylation, ADP-ribosylation, amidation, covalent bonding of flavin, covalent bonding of a heme moiety, covalent bonding of a nucleotide or nucleotide derivative, covalent bonding of a lipid or derivative lipid, covalent linkage of phosphotidylnositol, cross-linking, cyclization, disulfide bond formation, dimethylation, covalent crosslink formation, cystine formation, pyroglutamate formation, formylation, carboxylation range, glycosylation, GPl anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, addition by RNA of transfer of amino acids to proteins such as arginilation and ubiquitination. Such modifications are well known to those skilled in the art and have been described in greater detail in the scientific literature. Several particularly common modifications, glycosylation, lipid bonding, sulfation, gamma carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation, for example, are described in the more basic texts such as Protein-Structure and Molecular Properties (Proteins-St Ructure and Molecular Properties, 2nd Flp Edition, TE Creighton, H. Freeman and Company, 5 New York (1993) Many detailed journals are available on this subject, such as, for example, Covalent Modification After Protein Translation by F Wold, (Posttranslational Covalent Modification of Proteins, B.C. Jhonson, Ed. , Academic Press, New York 1-12 (1993); Seifter and Associates (1990) Meth. Enzymol. 182: pages 626 to 646) and Rattan and Associates (1992) Ann. New York Acad. Sci. 663: pages 48 to 62). It is also well known that polypeptides do not are always completely linear. For example, polypeptides can be branched as a result of ubiquitination, and can also be circular with or without branching, usually as a result of post-translational events, including events of • natural processing and events developed by human manipulation which do not occur naturally. Circular, branched or circular branched polypeptides can be synthesized by natural processes that are not translational and by synthetic methods.
Modifications can occur anywhere in a polypeptide, including the peptide structure, the amino acid side chains, and the amino and carboxyl terminal flp. The blocking of the group amino or carboxyl in a polypeptide, or both, by a covalent modification is common in synthetic polypeptides and those that occur naturally. For example, the amino terminal residue of the polypeptides made in E. col i, before ^ P 10 proteolytic processing will almost invariably be N-formylmethionine. The modifications can be a function of how the protein was made. For recombinant polypeptides, for example, modifications will be determined by the post-translational modification capacity of the host cell and the modification signals in ? k the polypeptide amino acid sequence. Accordingly, when glycocylation is desired, a polypeptide must be expressed in a host of glycocylation, generally a eukaryotic cell. Insect cells frequently perform the same post-translocation glycocylations as mammalian cells and, for this reason, insect cell expression systems have have been developed to efficiently express mammalian proteins that have native glycocylation patterns. Similar considerations are applicable to other modifications. ^ P The same type of modification may be present in the same or to a variable degree at several sites in a given polypeptide. Also, a particular polypeptide may contain more than one type of modification. • 10 Polypeptide Uses The receptor polypeptides are useful to produce antibodies specific for proteins, regions or fragments of the 2838, 14618 and 15334 receptor. Regions that have a qualification of The high index of antigenicity is illustrated in Figures 2, 6, and 10. Polypeptides, variants and receptor fragments, (including those which may have been described prior to the present invention) are useful for biological assays related to GPCRs. Said assays comprise any of the known functions or activities or properties of the GPCRs useful for the diagnosis and treatment of diseases related to the GPCR.
The receptor polypeptides are also useful in drug selection assays, in cell-based systems, or cell-free. The cell-based systems can be native, for example, cells that normally express the receptor protein, as a biopsy or expanded in a cell culture. However, in one of the embodiments, the cell-based assays comprise recombinant host cells that express the receptor protein. In determining the ability of the test compound to interact with the polypeptide, it may also comprise the ability of the test compound to preferentially bind to the polypeptide compared to the binder capacity, or a biologically active portion thereof, to bind to the polypeptide. The polypeptides can be used to identify compounds that modulate receptor activity. These compounds, for example, they can increase or decrease the affinity or linkage index to a known liqant, compete with the binder for the link to the receptor, or displace the ligand linked to the receptor, proteins 2838, 14618 and 15334 and the appropriate variants and fragments they can be used in high-throughput selections to test candidate compounds in their ability to bind to the receptor. These compounds can be further screened against a functional receptor to determine the effect of the compound on receptor activity. The compounds can be identified as active (agonist) or inactive (antagonists) of the receptor to a desired degree. Modulation methods can be performed in vi tro (for example, by culturing the cell with the agent) or alternatively, in vi ve (for example, by administering the agent to a subject). Examples include purine analogues such as those explained above. The receptor polypeptides can be used to select a compound for its ability to stimulate or inhibit the interaction between the receptor protein and a target molecule that normally interacts with the receptor protein. The target can be a ligand or a component of the signal path with which the receptor protein normally interacts (eg, a G protein or other interactor involved in the cAMP, or a phosphatidylinositol rotation and / or the activation of the cyclase of adenylate or phospholipase C). The assay includes the steps of combining the receptor protein with a candidate compound, under conditions that allow the receptor protein or a fragment thereof to interact with the target molecule, and to detect the formation of a complex between the protein and the protein. objective or to detect the biochemical consequence of the interaction with the receptor protein and the target, so that any of the associated effects of signal transduction of said P10 phosphorylation by G protein, cyclic AMP or phosphatidylinositol rotation, and said activation of adenylate cytosol or phospholipase C. The determination of the ability of the protein to bind to a target molecule can also be to be performed using a technology such as Real Time Bimolecular Interaction Analysis (BIA). Sjolander, S. and Urbaniczky, C. (1991) Anal Chem. • 63: pages 2338 to 2345 and Szabo and associates (1995) Curr. Opin. Struct. Biol. 5: pages 699 to 705. As is used in the present invention, "BIA" is a technology for studying biospecific interactions in real time, without marking any of the interactors (for example, BIAcore ™). Changes in the optical resonance of the plasmon of the The phenomenon surface (SPR) can be used as an indication of real-time reactions between biological molecules. The test compounds of the present invention,? can be obtained using any of the Many methods in combination with combination library methods known in the art, including: biological libraries, parallel solid phase libraries or solution phase can be spatially directed; synthetic library methods that require deconvolution; the "one-account-one" library method; Synthetic library methods that use the affinity chromatography selection. The biological library method is limited to polypeptide libraries, While the other four methods are applicable to polypeptides, non-peptide oligomers, or libraries in small molecules of compounds (Lam, K: S: (1997) Anticancer Druq Des.12: page 145). Examples of methods for the synthesis of molecular libraries can be found in the subject, for example, in the publication De W1T and associates (1993) Proc. Nati Acad. Sci. USA 90: page 6909; Erb and associates (1994) Proc. Nati Acad. Sci. USA 91: page 11422; Zuckermann and associates (1994).
J. Med. Chem. 37: page 2678; Cho and associates (1993) Sciencie 261: page 1303; Carell and associates (1994) Angew. Chem. Int. Ed. Engl. 33: page 2059; Carell and associates (1994) Angew. Chem. Int. Ed. Engl. 33: page 2061; and Gallop and associates (1994) J: Med. Chem. 5 37: page 1233. The libraries of compounds can be present in solution (for example Houghten (1992) Biotechniques 13: pages 412 to 421), or in beads (Lam (1991) Nature 354: pages 82 to 84), chips (Fodor (1993) Nature 364: pages 555 to 556), ^ 10 bacteria (North American Patent) Number 5,223,409 granted to Ladner), spores (North American Patent 09 granted to Ladner) plasmids (Culi and associated (1992) Proc. Nati Acad. Sci. USA 89: pages 1865, 1869), or in a phage (Scott and Smith (1990) Science 249: pages 386 to 390); (Devlin (1990) Science 249: pages 404 to 406); (Cwirla and associates (1990) Proc. Nati, Acad. Sci. 97: pages 6378 to 6382); (Felici • (1991) J: Mol. Biol. 222: pages 301 to 310); (Ladner mentioned above). Candidate compounds include, for example, 1) purine analogues, 2) peptides such as soluble peptides including tail and Ig fusion peptides and members of random peptide libraries (see for example the publication by Lam et al. (1991)). Nature 354: pages 82 to 84; Houghten and associates (1991) Nature 354: pages 84 to 86), and molecular chemistry derived combination libraries made of amino acids of D- and / or L- configuration; 3) phosphopeptides (for example members of libraries of targeted phosphopeptides, randomly and partially degenerate (see for example the Publication of Songyang and associates (1993) Cell 72: pages 767 to 778); 4) antibodies (eg polyclonal, monoclonal, humanized, anti-idiotypic, chimeric B 10, and single chain antibodies as well as Fab, F (ab ') 2, fragments of the Fab expression library, and epitope binding fragments of antibodies); and 5) small organic and inorganic molecules (e.g., molecules obtained from libraries of natural and combination products). A candidate compound is a soluble total length receptor or fragment that competes for the liquefier bond. Other candidate compounds include mutant receptors or appropriate fragments that contain mutations that affect the receptor function and therefore compete for the binder. Accordingly, a fragment that competes for the liq- erant, for example with a higher affinity, or a fragment that binds to the liq- ant but does not allow for release, is encompassed by the present invention. The present invention provides other fi nal spots to identify the compounds that modulate the receptor activity (stimulates or inhibits). The assays generally comprise an assay for events in the signal transduction path that indicates receptor activity. Therefore, the expression of genes that are required up or down in ^ ff 10 response to receptor protein dependent signal cascade, can also be tested. In one embodiment, the regulatory region of said genes can be operably linked to a marker that is readily detectable, such as luciferaza.
Alternatively, one could also measure the phosphorylation of the receptor protein, or the target of the receptor protein. The linkage and / or activation compounds can also be selected, using chimeric receptor proteins in which the extracellular field of the amino terminus or parts thereof, the entire membrane field or sub-regions, such as any of the seven segments after membrane and any of the intracellular or extracellular coils and the intracellular field of the carboxy terminus, or parts thereof, can be replaced by heterologous fields or subregions. For example, a binding region for protein G can be i? used so that it acts with a G protein different, and then this is recognized by the native receiver. Accordingly, a different set of signal transduction components is available as an endpoint assay for activation. Alternatively, you can replace ^ P 10 complete transmembrane portion (s) (such as transmembrane segments or intracellular or extracellular spirals) by complete transmembrane portions or subregions specific to a host cell that is different from the host cell from which the extracellular terminal field is derived amino acid and / or the binding region by protein G. This allows the assays to be carried out in • cells other than the specific host cells from which the receptor was derived. Alternatively, the extracellular field of the amino terminal (and / or other binding regions of binders) could be replaced by a field (and / or other linking regions) that bind to different binders, thereby providing an assay for the 25 test of compounds that interact with the heterologous extracellular field of the amino terminal (or regions) but still cause signal transduction. Finally, the activation can be detected by means of a reporter gene containing an easily detectable coding region, operably linked to a regulatory transcription sequence that is part of the path of native signal transduction. 2. Polypeptides receivers are also useful in competition binding assays in methods designed to discover compounds that interact with the receptor. In this way, a compound is exposed to a receptor polypeptide under conditions that allow the compound to bind or interact otherwise with the polypeptide. Soluble receptor polypeptides are also added to the mixture. If the test compound interacts with the soluble receptor polypeptide, the amount of the complex formed, or the activity from the target, decreases. receiver. This type of assay is particularly useful in cases in which it is anticipated that the compounds interact with specific regions of the receptor. Therefore, the soluble polypeptide that competes with the target region of the receptor is designed to contain polypeptide sequences corresponding to the region of interest. To form cell-free drug selection assays, it is desirable to immobilize either the receptor protein or fragment, or its target molecule to facilitate the separation of complexes from non-complexed forms of one or both of the proteins, as well as as to accommodate the automation of the trial. Techniques for protein immobilization in matrices can be used in drug selection trials. In one embodiment, a fusion protein can be provided which is added to a field that allows the protein to be bound to the matrix. For example, the transferase-S-glutathione fusion proteins (2838, 14618, and 15334 can be adsorbed on the glutathione sepharose beads (Sigma Chemical, St, Louis, MO) or the glutathione-derived microtiter plates, which they are then combined with cell lysates (eg S-labeled) and the candidate compound and the mixture is incubated under conduction conditions for the formation of a complex (eg under physiological conditions for salt and pH). incubation, the beads are washed to remove any unbound label, and the matrix is immobilized and radiolabelling is determined directly, or in the supernatant after the complexes are dissociated.Alternatively, the compounds can be dissociated from the matrix, separated by SDS-PAGE, and the level of receptor binding protein found in the fraction of the count quantified from the gel, using electrophoretic techniques is For example, any polypeptide or its target molecule can be immobilized using the conjugation of biotin and streptavidin using techniques well known in the art. Alternatively, antibodies reactive with the protein but that do not interfere with the binding of the protein to its target molecule can be derived to the plate vessels, and the protein trapped in the vessels by conjugation of antibodies. The preparations of a receptor-binding protein and a candidate compound are incubated in the receptor deposits presenting the protein and the amount of complex trapped in the reservoir can be quantified. The methods for the detection of said complexes, in addition to those described above for complexes immobilized by GST, include the immunodetection of complexes using antibodies reactive with the receptor protein target molecule, or which are reactive with the receptor protein and "Q compete with the target molecule, as well as the 5 assays linked by enzyme, which depend on the detection of an enzymatic activity associated with the target molecule The modulators of the receptor activity of the protein identified according to these tests of drug selection can be used to treat to a subject with a disease transmitted by the trajectory of the recipient, treating the cells expressing the 2838 protein such as the lymph node, thymus, vessel, testes, colon, and lymphocytes peripheral blood cells including, but not limited to, T (1 and 2), CD3 + (CD4 and CD8) helper cells, ^ P B cells and qranulocytes. The activity of the receptor protein modulators identified according to these drug selection trials can be used to treat a subject with a disease transmitted by the path of the recipient, by treating the cells expressing the 14618 protein, such as in the breast, skeletal muscle, vessel, and peripheral blood lymphocytes as well as CD34 + cells and megakaryocytes. Modulators of the activity of the receptor protein identified according to these drug selection assays can be used to treat a subject with a disease transmitted by the path of the receptor, treating the cells expressing the 15334 protein such as in the node of the lymph, tonsils, pancreas, colon, vessel, peripheral blood cells, thymus, adrenal glands and heart as well as megakaryocytes and erythroblasts. These methods of treatment include the steps of administering modulators of the activity of the protein in a pharmaceutical composition as described in the present invention, to a subject in need of such treatment. Figure 3 shows the expression of the 14618 receptor in a variety of normal human tissues. The gene is highly expressed in the chest and skeletal muscle. Accordingly, expression of the gene in the diseases comprising these tissues is particularly important. Significant expression also occurs in the thyroid, placenta, fetal kidney, fetal heart, and lymph node. Accordingly, expression of the gene is important for diseases comprising these tissues as well. In addition, as illustrated in Fig. 13, the lowest levels of expression are seen in a variety of other tissues. Therefore, gene expression is also important in diseases that • understand these tissues. 5 The 14618 receptor is also expressed in several hematopoietic cells with and without activation. The gene is highly expressed in cells of the bone marrow CD34 +. Accordingly, gene expression is important in a variety of blood cell progenitors. The expression of this gene is therefore important for diseases that comprise deficiencies in any of the major types of blood cells, for example, neutropenia, thrombocytopenia and anemia. The gene is also expressed highly in the meqacariocytes of the mobilized peripheral blood cells (mobilized with G-jßb CSF). Accordingly, the expression of this gene is important for diseases comprising the function of platelets such as thrombocytopenia. The Insignificant expression is also seen in leukocytes of mobilized peripheral blood cells, CD34 cells "of mobilized bone marrow, or CD 434 cells" of spinal cells. Therefore, the expression of The gene is important for the function of these cells and therefore important for diseases that comprise immune or inflammation functions. In addition, gene expression occurs in activated peripheral blood mononuclear cells. Activated B cells express the gene differentially. Accordingly, expression of the gene is important for diseases comprising immune functions and / or inflammation. The 2838 receptor is expressed in the cells and tissues illustrated in Figure 15. High levels of expression are shown at the lymph node and the thymus. Accordingly, expression of the gene is especially important for diseases comprising these tissues. Extremely high expression has been found in CD8 cells and activated B cells. High expression also occurs in activated T helper cells (1 and 2) CD4 cells and Jurkat cells (a T cell line). Expression is differential in activated B cells and activated T helper cells. The expression increases to the activation in both of these cell types. By request, the expression of the gene is important for diseases that include immunological functions and inflammation. The gene is also expressed in an important way in granulocytes. Accordingly, expression of the gene is important for diseases comprising these cells. Figure 16 shows the expression of receptor 15334 in normal human tissues. The gene is highly expressed in the lymph node, tonsils and pancreas. Accordingly, expression of the gene is particularly important for diseases comprising these tissues. The expression of the gene is also high in the colon, testes, placenta, fetal heart and vessel. Accordingly, expression of the gene is also important for diseases comprising these tissues. In addition, the figure illustrates low levels of expression in several of the other tissues. Accordingly, expression of the gene may be important for diseases comprising these tissues. The expression of the 15334 receptor has also been studied in several hematopoietic cells (Figures 17 to 19). An extremely high expression occurs in megakaryocytes and major erythroblasts. Therefore, gene expression is important for erythrocyte differentiation and megakaryocyte differentiation and therefore is important for the treatment of anemia and thrombocytopenia. In addition, gene expression is significantly increased in resting B cells compared to activated B cells. Therefore, expression of the gene is important for the immunological function of B cells. In addition, lower levels of expression were found in several other cells of the hematopoietic lineage. See Figure 17. Expression of this gene in hematopoietic cells in a restricted manner by lineage indicates that expression is important for the regulation of the development of lineage cells, erythrocyte / red blood cell or megakaryocyte / platelet blood cells.
Diseases comprising the vessel include, but are not limited to, splenomegaly, which include non-specific splenitis, congestive splenomegaly, and splenic infarcts; neoplasms, congenital anomalies and rupture. Diseases associated with splenomegaly include infections, such as nonspecific splenitis, infectious mononucleosis, tuberculosis, typhoid fever, brucellosis, cytomegalovirus, syphilis, malaria, isoplasmosis, toxoplasmosis, kala-azar, trypanosomiasis, schistosomiasis, leishmaniasis, and echinococcosis; congestive diseases related to partial hypertension, such as liver cirrhosis, portal or splenic vein thrombosis, and heart failure; lymphohaematogenic diseases, such as Hodgkin's disease, lymphocytes / non-Hodgkin's leukemia, multiple myeloma, myeloproliferative diseases, hemolytic anemias, and rhizostopenic purpura; immunological-inflammatory conditions such as rheumatoid arthritis and chronic systemic lupus; storage diseases such as Gaucher's disease, Niemann-Pick disease, and mucopolysaccharidosis; and other conditions, such as amyloidosis, neoplasms and primary cystitis, and secondary neoplasm. Diseases that comprise the lung include but are not limited to congenital anomalies; atelectasis; disease of vascular origin, such as pulmonary congestion and edema, including pulmonary hemodynamic edema and edema caused by microvascular injury, respiratory disease syndrome in adults (diffuse alveolar damage), pulmonary embolism, hemorrhage and infarction, and pulmonary hypertension and vascular sclerosis, chronic obstructive pulmonary disease such as emphysema, chronic bronchitis, bronchial asthma and bronchiectasis; diffuse interstitial diseases (infiltrative or restrictive) such as pneumoconioses, sarcoidosis, idiopathic pulmonary fibrosis, desquamative intercisial pneumonitis, hypersensitivity pneumonitis, pulmonary eosonifilia (pulmonary infiltration with eosonifilia), pneumonia organized by Bronchi li ti s obl i teran s, hemorrhage syndromes diffuse pulmonary disease, including Goodpasture syndrome, idiopathic pulmonary hemosiderosis and other hemorrhagic syndromes, pulmonary involvement in collagen vascular diseases, pulmonary alveolar proteinosis, complications of therapies such as drug-induced lung disease, radiation-induced lung disease, and lung transplant; tumors such as bronchogenic carcinoma, including syndromes for neoplastic, bronchioloalveolar carcinoma, neuroendocrine tumors such as bronchial carcinoid, miscellaneous tumors, and metastatic tumors; pathologies of the pleura including inflammatory effusions of the pleura, non-inflammatory effusions of the pleura, pneumothorax, tumors of the pleura including solitary fibrous tumors (fibroma of the pleura) and malignant mesothelioma. Diseases comprising the colon include but are not limited to congenital anomalies, such as atresia and stenosis, Meckel's diverticula, Hischsprung's disease / congenital megacolon-aganglionic, enterocolitis, such as diarrhea and dysentery, infectious enterocolitis, including viral gastroenteritis, enterocolitis bacterial, necrotizing enterocolitis, colitis associated with antibiotics (pseudomembranous colitis), collagen or lymphositic colitis, miscellaneous intestinal inflammatory diseases, including parasites and protozoa, acquired immunodeficiency syndrome, transplants, drug-induced intestinal injury, radiation enterocolitis, colitis neutropenic (typhlitis) and fun colitis; idiopathic inflammatory bowel diseases such as Crohn's disease, ulserative colitis, colon tumors, such as non-neoplastic polyps, edenomas, familial syndromes, colorectal carcinogenesis, colorectal carcinoma and carcinoid tumors. Diseases that include the liver, include but are not limited to liver injury; jaundice and cholestasis, such as formation of bilirubin and bile, liver failure and cirrhosis, such as cirrhosis, portal hypertension, asceticis including toxic deviations and splenomegaly portosis, infectious diseases such as viral hepatitis, including infection from hepatitis A to E and infection for other hepatitis viruses, clinopathological syndromes such as transporter status, asymptomatic infection, acute viral hepatitis, chronic viral hepatitis, and fulminant hepatitis; autoimmune hepatitis, liver disease induced by drugs and toxins, such as alcoholic liver disease, errors in the birth of metabolism, and pediatric liver disease, such as hemochromatosis, Wilson's disease, f-antitrypsin deficiency, and neonatal hepatitis, intra hepatic biliary tract disease such as secondary biliary cirrhosis, primary biliary cirrhosis, primary sclerosing cholangitis and biliary tree abnormalities, circulatory diseases such as impaired blood flow within the liver including, hepatic artery involvement, and obstruction of portal vein and thrombosis, impaired blood flow through the liver including passive congestion and centrilobular necrosis and hepatis peliosis, obstruction of hepatic vein outflow including hepatic vein thrombosis (Budd-Chiari syndrome) and veno-occlusive disease, Liver disease associated with pregnancy, such as preclampsia and eclampsia, acute fatty liver of pregnancy, intrehepatic cholestasis of pregnancy, liver complications of organ or bone marrow transplants, such as drug toxicity after a bone marrow transplant, host-versus-graft disease and liver rejection , and nonimmunological damage to liver grafts, tumors and tumorous conditions, such as nodular hyperplasias, adenomas, and malignancies including primary carcinoma of the liver and tumors of metastasis. Diseases that comprise the uterus and endometrium include, but are not limited to endometrial histology in menstrual cycles; functional endometrial diseases, such as anovulatory cycle, inadequate luteal phase, oral contraceptives, and induced endometrial changes, and menopausal and post-menopausal changes, inflammations such as chronic endometritis, adenomyosis, endometriosis, endometrial polyps; endometrial hyperplasia; malignant tumors such as carcinoma of the endometrium; mixed Mullerian and mesenchymal tumors, such as mixed malignant malignant tumors; myometrial tumors, including leyomyosarcomas and endometrial stromal tumors. Diseases comprising the brain include, but are not limited to diseases comprising the neurons, and diseases comprising the qlia, such as astrocytes, oligodendrocytes, ependymal cells, and microglia; cerebral edema, increased intracranial pressure and herniation, and hydrocephalus, diseases due to deformation and development, such as neural tube defects, frontal brain anomalies, posterior fossa anomalies, and syringomyelia and hydromelia; perinatal injury to the brain; cerebrovascular diseases such as those related to hypoxia, ischemia and infarction, including hypotension, hypoperfusion, global cerebral ischemia due to low flow states and focal cerebral ischemia, infarcts from the obstruction of the local blood supply, intracranial hemorrhage, including hemorrhage intracerebral (intraparenchymal), subaranoid hemorrhages and berry aneurysms of rupture and vascular deformations, hypertensive cerebrovascular disease, including lacunar infarcts, haemorrhages by cuts, and hypertensive encephalopathy, infections such as meninqitis aquda, including acute pyogenic (bacterial) meninitis and acute aseptic meningitis ( viral), acute focal suppurative infections, including abscesses of the brain, subdural empyema, extradural abscess, chronic bacterial meningoencephalitis including tuberculosis and mycobacteriosis, neurosyphilis and neuroborreliosis (Lyme disease), viral meningo encephalitis, including viral encephalitis carried by arthropods (Arbo), Herpes simplex virus Type 1, Herpes simplex virus type 2, virus of varicella-zoster, (Herpes zoster), cytomegalovirus, poliomyelitis, rabies, and human immunodeficiency virus 1, including meningoencephalitis HIV-1 (subacute encephalitis), vacuolar myelopathy, myopathy associated with AIDS, peripheral neuropathy, and STDA in children, progressive multifocal legoencephalopathy, subacute sclerosing panencephalitis, meningoencephalysis fungal, and other infectious diseases of the nervous system, transmissible spongiform encephalopathies (prion disease); dimorylation diseases, including multiple sclerosis, variant of multiple sclerosis, acute disseminated encephalomyelitis and acute necrotizing hemorrhagic encephalomyelitis and other diseases with dimellination, degenerative diseases such as degenerative diseases that affect the cerebral cortex including Alzheimer's disease and Pick's disease , degenerative diseases of basal gangleos and brainstem, including Parkinson's disease, idiopathic Parkinson's disease (paralytic agitants), progressive supranuclear palsy, corticobasal degeneration, multiple system atrophy, including striatonigral degeneration, Shy-Drager syndrome, and olivopontocerebellar atrophy and Huntington's disease, spinocerebellar degenerations, including spinocerebellar ataxias, including Friedreich ataxia and ataxia-talenglectasia, degenerative diseases affecting motor neurons including sclerosis lateral amyotrophic (motor neurone disease) bulboespinal atrophy (Kennedy syndrome) and spinal muscular atrophy, errors in the birth of metabolism such as leuocodist rofias, including Krabbe's disease, metachromatic leukodystrophy, adrenoleukodystrophy, pelizaeus disease Merzbacher, Canavan disease, mitochondrial encephalomyopathies, including Leigh's disease and other mitochondrial encephalomyopathies, toxic and acquired metabolic diseases including deficiencies in the vitamin such as thiamin (vitamin Bl), deficiency and deficiency of vitamin B12, neurological sequelae of metabolic disturbances including hypoglycemia, hypergiukaemia and hepatic encephalopathy, toxic diseases including carbon monoxide, methanol, ethanol and radiation including combined methotrexate and radiation-induced injury; tumors such as gliomas, including astrocytoma, including fibrillar astrocytoma (diffuse) and glioblastoma multiforme, phylocytic astrocytoma, pleomorphic xanthoastrositoma, and brainstem glioma, oligodendroglioma, and ependinoma and related paraventricular mass lesions, neuron tumors, differentiated neoplasms deficiently including, medulloblastoma, other parenchymal tumors, including primary brain lymphoma, germinal cell tumors and pineal parenchymal tumors, meningiomas, metastatic tumors, paraneoplastic syndromes, peripheral nerve lining tumors, including schwannoma, neurofibroma, and malignant lining tumors peripheral nervous (malignant schwannoma) and neurocutaneous syndromes (phakomatous) including neurofibromatosis, including neurofibromatosis type 1 (NF1) and neurofibromatosis type 2 (NF2), tuberous sclerosis, and von Hippel-Lindau disease. Diseases comprising the T cell include, but are not limited to, hypersensitivity carried by the cells such as delayed-type hypersensitivity, and cytotoxicity carried by the T cell, rejection to transplants, autoimmune diseases such as chronic systemic lupus, Sjögren's syndrome, systemic sclerosis, inflammatory myopathies, mixed connective tissue diseases, and polyarteritis nodosa and other vasculitis; immunological deficiency syndromes including, but not limited to, primary immunodeficiencies, such as thymic hypoplasia, severe combined immunodeficiency diseases, and AIDS, leukopenia, reactive (inflammatory) proliferations of white cells, including but not limited to leukosytosis, acute nonspecific lymphadenitis and chronic non-specific lymphadenitis, neoplastic white cell proliferations, including but not limited to lymphoid neoplasms such as T-cell precursor neoplasms, such as acute lymphoblastic leukemia / lymphoma, peripheral T cell neoplasms, and natural killer cells that include peripheral T-cell lymphoma, adult T-cell leukemia / lymphoma, mycosis fungoides, and Sézary syndrome and Hodgkin's disease.
Skin diseases include but are not limited to pigmentation diseases and melanocytes, including but not limited to vitiligo, freckles, melasma, lentigo, nevocellular nevus, nevi dysplastic, malignant melanoma, benign epithelial tumors, including but not limited to seborrheic keratoses, acanthosis nigricans, fibroepithelial polyps, epithelial cysts, keratoacanthoma, and adnexal tumors (appendices), premalignant and malignant epidermal tumors, including but not limited to, actinic keratosis, squamous cell carcinoma, basal cell carcinoma, and carcinoma of the Merkel cell, skin tumors including but not limited to benign fibrous histiositoma, estedermatofibrosarcoma protuberans xanthomas, and vascular dermal tumors, tumors of cellular immigrants to the skin, including but not limited to histiositosis X, mycosis fungoides (cutaneous lymphoma of the T cell) and mastositosis; maturation diseases of the epidermis, including but not limited to ichthyosis; Acute inflammatory dermatoses, including but not limited to, urticaria, acute eczematous dermatitis, erythema multiforme, chronic inflammatory dermatoses, including but not limited to psoriasis, lichen planus, and chronic lupus, blistering diseases (bullous) (including but not limited to penifigus, bullous pemphigoid, dermatitis herpetiformis, and non-inflammatory blistering diseases: epidermolysis bullosa and porphyria; diseases of epidermal appendages, including but not limited to acne vulgaris; panniculitis, including but not limited to erythema nodosum and erythema induratum, and infections and infestation such as warts, moluscum contagiosum, impetigo, superficial fungal infections, and bites, pustules of anthropods and infestations.In normal bone marrow, the myelositic series (polymorphonuclear cells) make up approximately 60% of the cellular elements , and the erythrocytic series, from 20 to 30% Lymphocytes, monocytes, reticular cells, plasma cells and megakaryocytes together constitute 10 to 20% Lymphocytes constitute 5 to 15% of normal adult marrow. bone, the cell types are mixed so that the red blood cell precursors (er) i troblasts), macrophages (monoblasts) platelets (meqacariocytes), polymorphonuclear leukocytes (myeloblasts) and lymphocytes (lymphoblasts) may be visible in a microscopic field. In addition, there is a cell stalk for different cell lines, as well as a precursor cell stalk for the involved progenitor cells of the different lineages. The different cell types and stages of each would be known to those skilled in the art and are found, for example, on page 42 (Figures 2 to 8) of Immunology, Immunopathology and Immunity, Fifth Edition, Sell and Simon Associates and Schuster (1996), incorporated herein by reference for their teachings of the cell types found in the bone marrow. Accordingly, the present invention is directed to diseases that are originated from these cells. These diseases include, but are not limited to, the following diseases comprising hematopoietic stem cells; committed lymphoid progenitor cells, lymphoid cells including B and T cells, compromised myeloid progenitors, including monocytes, granulocytes and megakaryocytes; and committed erythroid progenitors. These include but are not limited to including lymphoid leukemias B-T-lymphoid leukemia, undifferentiated leukemias, erythroleukemia, megakaryoblastic leukemia, monocytic; [the leukemias are included with and without differentiation]; chronic and acute lymphoblastic leukemia, chronic and acute lymphocytic leukemia, acute and chronic myelogenous leukemia, lymphoma, myelodiplastic syndrome, chronic and acute myeloid leukemia, myelomonocytic leukemia, chronic and acute myeloblastic leukemia, chronic and acute myelogenous leukemia, chronic and acute promyelocytic leukemia , chronic and acute myelocytic leukemia, haematological diseases of the monocyte-macrophage lineage, such as chronic juvenile myelogenous leukemia, secondary AML, antecedent haematological disease; refractory anemia, aplastic anemia, reactive cutaneous angioendotheliomatosis; fibrosis diseases comprising the altered expression of dendritic cells, diseases including systemic sclerosis, E-M syndrome, toxic epidemic oil syndrome, eusinophilic fasciitis, localized forms of scleroma, keloid and fibrosis colonopathy; histiocinoma fibrosa maliqno angiomatoide, carcinoma, which include a primary carcinoma of squamous cells of the head and neck sarcoma, including kaposi sarcoma, fibroadenoma and phyllodes tumors, including fibroadenoma mammary; stromal tumors; phyllodes tumors; including histiocytoma; erythroblastosis; neurofibromatosis; diseases of the vascular endothelium; demyelinating agents, particularly in anterior lesions, qliosis, vasogenic edema, vascular disease, Alzheimer's disease and Parkinson's disease; T cell lymphomas and B cell lymphomas Diseases comprising the heart, include but are not limited to cardiac failures, including but not limited to, cardiac hypertrophy, failure of the left side of the heart, failure of the right side of the heart, ischemic heart disease, including but not limited to angina. of chest, myocardial infarction, chronic heart ischemic disease, and sudden cardiac death; hypertensive heart disease, including but not limited to hypertensive systemic heart disease (left side) and pulmonary hypertensive heart disease (right side); valvular heart diseases, including but not limited to valvular degeneration caused by calcification such as calcified aortic stenosis, calcification of a dicucosal aortic valve congenitally, and mitral annular calcification and myxomatous degeneration of the mitral valve (prolapse of the mitral valve), rheumatic fever, and rheumatic heart disease, infective endocarditis, and non-infected vegetations such as non-bacterial thrombotic endocarditis and chronic systemic lupus endocarditis (Libman-sacks disease), carcinoid heart disease and complications by artificial valves, myocardial disease including but not limited to dilated cardiomyopathy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, and myocarditis; diseases Pericardial QP, including but not limited to pericardial effusion and hemopericardium and pericarditis, including acute pericarditis and healed pericarditis, and rheumatoid heart disease, neoplastic heart disease, including but not limited to primary cardiac tumors, such as myxoma, ^ P 10 lipoma, papillary fibroelastoma, rhabdomyoma, and sarcoma, and cardiac effects of non-cardiac neoplasms; congenital heart disease, including but not limited to cyanosis, deviations from left to right posterior, such as septal defects atrial, septal defects, patent ductus arteriosus, and atrioventricular septal defects, ^ "t early cyanosis of right to left deviation, such as tetralogy of fallot, transposition of large arteries, truncated arteries, atresia tricuspid and total anomalous pulmonary venous connection, obstructive congenital anomalies such as coarctation of the aorta, stenosis and pulmonary atresia, and stenosis and aortic atresia, and diseases involving cardiac transplants.
Diseases that include blood vessels include, but are not limited to, responses of vascular cell walls to lesions, such as endothelial dysfunction, and endothelial activation and intimal thickening; vascular diseases including, but not limited to congenital anomalies, such as arterovenous fistula, arteriosclerosis and hypertensive vascular disease such as hypertension, vasculatives, inflammatory diseases, such as giant cell arteritis (temporal) takayasu arteritis, polyarteritis nodosa (classical) syndrome Kawasaki (mucocutaneous lymph node syndrome), microscopic polyangiitis (microscopic polyarteritis, hypersensitivity or leukocytoclastic anglitis), Wengener's granulomatosis, thromboanglitis obliterans (Buerger's disease, vasculitis associated with other diseases and infectious arteritis, Raynaud's disease, aneurysms and dissection) such as abdominal aortic aneurysms, syphilitic aneurysms (leutic) and aortic dissection (dissection hematoma), vein and lymphatic diseases, such as varicose veins, thrombophlebitis and phlebothrombosis, upper vena cava obstruction (superior vena cava syndrome) obstruction of the inferior vena cava (inferior vena cava syndrome, and lymphagitis, and lymphedema, tumors, including benign tumors and tumor-like conditions such as hemangioma, lymphangioma, glomus tumor, (glomangioma) vascular ectasias and bacillary angiomatosis, and tumors of intermediate grade (line of the limit of evil ignore the low degree) such as Kaposi's sarcoma and hemangioendothelioma, malignant tumors, such as angiosarcoma and hemangiopericytoma, and pathology of therapeutic interventions of vascular diseases, such as balloon angioplasty and related techniques and vascular replacements such as pacemaker graft surgery of the coronary artery. Diseases comprising red blood cells include, but are not limited to anemias, such as hemolytic anemias, including hereditary spherocytosis, life-long haemolytic disease to defects of the above red blood cells: glucose-6-phosphate dehydrogenase deficiency, HOZ cell, thalassemia syndromes, nocturnal paroxysmal hemoglobinuria, immunohemolytic anemia, and hemolytic anemia resulting from trauma to red blood cells; and anemia of diminished erythropoiesis, including megaloblastic anemias such as anemia due to vitamin B12 deficiency; pernicious anemia, and anemia of foleate deficiency, iron deficiency anemia, anemia due to chronic disease, plastic anemia, pure red cell aplasia, and other forms of bone marrow failure. Diseases comprising the thymus include developmental diseases, such as DiGeorge syndrome with hypoplasia and thymic aplasia; thymic cysts; thymic hypoplasia which includes the appearance of lymphoid follicles within the thymus, creating thymic follicular hyperplasia; and thymomas, including germ cell tumors, lymphomas, Hodgkin's disease and carcinoids. Thymomas may include encapsulated or benign thymoma and malignant thymoma Type 1 (invasive thymoma) or Type II, thymic thymic carcinoma. Diseases comprising B cells include, but are not limited to, B cell precursor neoplasms such as leukemia / lymphoblastic lymphoma. Peripheral B cell neoplasms include, but are not limited to, chronic lymphocytic leukemia / small lifocitic lymphoma, follicular lymphoma, diffuse large B-cell lymphoma, Burkitt's lymphoma, plasma cell neoplasms, multiple myeloma, and related, lymphoplasmacytic lymphoma (Waldenstrom's macroglobulinemia), mantle cell lymphoma, marginal zone lymphoma (MALToma), and hairy cell leukemia. Diseases comprising the kidney include but are not limited to congenital abnormalities including, but not limited to, cystic diseases of the kidneys, including but not limited to cystic kidney dysplasia, autosomal dominant polycystic kidney disease (adults), kidney disease autosomal recessive polycystic (childhood) cystic diseases of the renal medulla, which include but are not limited to medullary sponge disease and nephronophthisis / urea medullary cystic disease complex, acquired cystic disease (associated with dialysis) such as simple cysts , qlomerular diseases that include glomerular lesion pathologies including but not limited to immune complex deposition in itself including, but not limited to, anti-GBM nephritis, Heymann's nephritis and antibodies against planted antigens, circulating immune complex nephritis, antibodies for glomerular cells, i cell-mediated immunity in glomerulonephritis, activation of the alternative path of complement, epithelial cell injury and pathologies comprising glomerular lesion transmissions, including cellular and soluble transmitters, acute glomerulonephritis, such as acute proliferative glomerulonephritis (post-streptococcal, postinfectious), including but not limited to post-streptococcal glomerulonephritis and acute non-streptococcal glomerulonephritis, rapidly progressive (growing) glomerulonephritis, nephrotic syndrome, membranous glomerulonephritis (membranous nephropathy), minimal change disease (lipoid nephrosis), segmental focal glomerulosclerosis, membranoproliferative glomerulonephritis, IgA nephropathy (Berger's disease), necrotizing glomerulonephritis and proliferative focal (focal glomerulonephritis), hereditary nephritis, including but not limited to Alport syndrome and thin membrane disease (in benign familial hematuria) chronic glomerulonephritis, glomerular lesions associated with systemic diseases, including but not limited to systemic chronic lupus, Henoch purpura -Schonl ein, bacterial endocarditis, diabetic glomerulosclerosis, amyloidosis, fibrillar and immunotactoid glomerulonephritis, and other systemic diseases; diseases affecting the tubules and interstitium, including acute tubular necrosis and tubulointerstitial nephritis, including but not limited to pyelonephritis and urinary tract infection, acute pyelonephritis, chronic pyelonephritis and reflux nephropathy, tubulointerstitial nephritis induced by drugs and toxins, including but not limited to not limited to acute interstitial nephritis induced by drugs, analgesic abuse nephropathy, nephropathy associated with non-steroidal anti-inflammatory drugs, and other tubulointerstitial diseases including, but not limited to, urate nephropathy, hypercalcemia and nephrocalcinosis, and multiple myeloma, blood vessels, including benign nephrosclerosis, malignant hypertension and accelerated nephrosclerosis, renal artery stenosis, thrombotic microangiopathies, including but not limited to classical hemolytic-uremic syndrome (childhood), thrombocytopenic purpura haemolytic-uraemic / thrombotic syndrome in adults, idiopathic HUS / TTP, and other basilar diseases including, but not limited to, renal disease due to atherosclerotic ischemia, arteroembolic renal disease, nephropathy due to sickle cell disease, diffuse cortical necrosis, and renal infarction, obstruction of the urinary system (obstructive uropathy); urolithiasis (kidney stone stones); and kidney tumors including, but not limited to, tumors • benign such as papillary adenoma, renal fibroma or haematoma (renomedullary interstitial cell tumor), angiomyolipoma, and oncocytoma and malignancies including renal cell carcinoma (hypernephroma, kidney adenocarcinoma), which includes urothelial carcinomas of the pelvis renal. 10 Breast diseases include, but are not limited to, developmental disorders; inflammations including, but not limited to, acute mastitis, periductal mastitis, periductal mastitis (recurrent subareolar abscesses, squamous metaplasia of the lactiferous ducts), duct ectasia of the mammary duct, fat necrosis, granulomatous mastitis, and pathologies associated with silicone implants in the • chest, fibrocystic changes; proliferative breast diseases including but not limited to a, epithelial hyperplasia, sclerosing adenosis, and small duct papillomas, tumors including but not limited to, stromal tumors such as fibroadenoma, phyllodes tumors and sarcomas, epithelial tumors such as papilloma of the large duct; carcinoma of the breast including carcinoma in the site (non-invasive) that includes carcinoma of the ducto in si tu, (including Pa ge t 's disease) and lobular carcinoma in the site, and invasive carcinoma (infiltrante) including but not limited to a, invasive carcinoma of the duct, without special type, invasive lobular carcinoma, medullary carcinoma, colloid carcinoma (mucus), tubular carcinoma, invasive papillary carcinoma, and miscellaneous malignancies. Breast diseases in men include, but are not limited to, gynecomastia and carcinoma. Diseases comprising the testes and epididymis include but are not limited to congenital anomalies such as cryptorchidism, recessive changes such as atrophy, inflammations such as nonspecific epididymitis and orchitis, granulomatous (autoimmune) orchitis, and specific inflammations including but not limited to gonorrhea mumps, parotiditis, tuberculosis and syphilis, vascular disorders including tension, testicular tumors including germ cell tumors including but not limited to, seminoma, spermatocytic seminoma, embryonal carcinoma, yol k sa c tumor, choriocarcinoma, teratoma and mixed tumors, Stromal tumors of the sexual-gonadal cord, including but not limited to, tumors of the cells of the eydí g (interstitial), tumors of the cell • of sertol i (androblastoma) and testicular lymphoma, and 5 miscellaneous lesions of the sole trait. Diseases comprising the prostate include but are not limited to, inflammations, benign enlargement, eg, nodular hyperplasia (hypertrophy or prostatic hyperplasia) benign, and tumors such as carcinoma. Diseases that comprise the thyroid include, but are not limited to, hyperthyroidism; hypothyroidism including but not limited to cretinism and myxedema; thyroiditis including but not limited to, shimo thyroiditis, subacute thyroiditis (granulomatous), subacute lymphocytic thyroiditis (painless); Graves disease and Goiter • multinodular and diffuse, including but not limited to, non-toxic, diffuse goitre (simple) and multinodular goitre, thyroid neoplasms including, but not limited to, adenomas and other benign tumors and carcinomas which include but are not limited to, papillary carcinoma, follicular carcinoma, medullary carcinoma, and anaplastic carcinoma; and anomalies congenital Diseases that comprise the skeletal muscle include tumors such as rhabdomyosarcoma. Diseases comprising the pancreas include those of the exocrine pancreas such as congenital anomalies including, but not limited to, ectopic pancreas, pancreatitis including but not limited to, pancreatitis, cysts, including but not limited to pseudocysts, tumors including but not limited to, cystic tumors and carcinoma of the pancreas; and diseases of the endocrine pancreas such as diabetes mellitus; Islet cell tumors including but not limited to insulinomas, qastrinomas and other rare island cell tumors. Diseases that comprise the small intestine include malabsorption syndromes such as, cyclic orifices, tropical holes (postinfectious orifices) Wippl e disease, disaccharidase (lactose) deficiency, abetalipoproteinemia, and small bowel tumors that include adenomas and adenocarcinomas. Diseases related to the reduced number of platelets, thrombocytopenia, include idiopathic thrombocytopenic purpura including acute idiopathic thrombocytopenic purpura, drug-induced thrombocytopenia, thrombocytopenia associated with HIV, and thrombotic microangiopathies, thrombotic thrombocytopenic purpura, and hemolytic-uremic syndrome. Diseases comprising the T cell precursor neoplasms include lymphoma / lymphoblastic leukemia of the precursor T. Diseases comprising the peripheral T cell and the neoplasms of the natural deadly cell include, chronic T-cell lymphocytic leukemia, qranular lymphocytic leukemia large, mycosis fungoides and Sésa ry syndrome, peripheral T-cell lymphoma, angioimmunoblastic T-cell lymphoma not specified, angiocentric lymphoma (T cell / NK cell lymphoma), intestinal T-cell lymphoma, lymphoma / leukemia the T cell in adults and large anaplastic cell lymphoma. Diseases comprising the ovaries include, for example, polycystic ovarian disease, the syndrome of S te in-l a ven tha 1, pseudomyxoma of the peritoneum and stromal hypertecosis.; ovarian tumors such as coelomic epithelial tumors, serous tumors, mucosal tumors, endometeroid tumors, cellular clear adenocarcinoma, cystadenofibroma, Brenner's tumor, epithelial surface tumors, qerminal cell tumors such as mature (benign) teratomas, teratomas monodérmicos, malignant immature teratomas, dysgerminoma, endodermal sinus tumor, choriocarcinoma, sexual cord-stomal tumors such as, granulosa-teak cell tumors, fibromas-thecoma, androblastomas, deadly cell tumors, gonadoblastoma; tumors by metastases such as Krukenberg tumors. Bone-forming cells include osteoprogenitor cells, osteoblasts, and osteocytes. Bone diseases are complex because they can have an impact on the skeleton during any of its stages of development. Therefore, the diseases can have variable manifestations and can comprise one or multiple or all the bones of the body. Such diseases include, congenital deformities, achondroplasia and tanatoforic dwarfism, diseases associated with the abnormal matrix such as collagen diseases of type 1, osteoporosis, Paget's disease, rickets, osteomalacia, high-rotation osteodystrophy, low rotation of the aplastic disease, teonecrosis, pyogenic osteomyelitis, tuberculous ostiomyelitis, osteoma, osteoid osteoma, osteoblastoma, osteosarcoma, osteochondroma, chondromas, chondroblastoma, chondromyxoid fibroma, chondrosarcoma, fibrous cortical defects, fibrous dysia, fibrosarcoma, malignant fibrous histocytoma, Ewing's sarcoma, primitive neuroectodermal tumor, giant cell tumor and tumors by metastasis. The receptor polypeptides are therefore useful for the treatment of diseases associated with receptors that are characterized by an aberrant expression or activity of the receptor protein, especially as described above. In one embodiment, the method comprises the administration of an agent (eg, an agent identified by a screening assay described herein) or a combination of agents that regulates (e.g., upregulates or downregulates) the expression or activity of the protein. In another embodiment, the method comprises administering a protein as therapy to compensate for reduced or aberrant expression or activity of the protein. The stimulation of the activity of the protein is desirable in situations in which the protein is abnormally sub-regulated and / or in which the increased activity of the protein is likely to have a beneficial effect. In a similar way, inhibition of protein activity is desirable in situations in which the protein is abnormally sub-regulated and / or in which the decreased activity of the protein is likely to have a beneficial effect. In one example of such a situation, a subject has a disease characterized by aberrant development or cellular differentiation. In another example of such a situation, the subject has a proliferative disease (for example cancer) or a disease characterized by an aberrant hematopoietic response. In another example of such a situation, it is desirable to achieve tissue regeneration in a subject (e.g., where the subject has suffered a brain or spinal cord injury, and it is desirable to regenerate the tissue of the neurons in a regulated manner) . In yet another aspect of the present invention, the proteins of the invention can be used as "decoy proteins" in a two-hybrid assay, or a three-hybrid assay (see for example U.S. Patent No. 5,283,317; the Zervos publication and Associates (1993) Cell 72: pages 223-232, Madura and Associates (1993) J. Biol. Chem. 268: pages 12046-12054, Bartel and Associates (1993) Biotechni ques 14: page s 920 -924; Iwabuchi and Associates (1 993) Oncogene 8: page s 1 693-1 696; and the document 94/10300 granted to Bren t), to identify other proteins (captured proteins) which bind to or interact with the proteins of the present invention and regulate their activity. The 2838 receptor polypeptides are also useful for producing a target for diagnosis of a disease or predisposition to disease transmitted by the receptor protein, especially in hematopoietic cells such as helper cells -T (1 and 2) B cells and cells. granulocytes The 14618 receptor polypeptides are also useful for producing a target for the diagnosis of a disease or predisposition to a disease transmitted by the receptor protein, especially in the breast, skeletal, skeletal, muscle and lymph node, as well as the progenitor cells. CD34 + and cells generated from these progenitors, which include erythroid, lymphoid and megakaryocytic lineages. The 15334 receptor polypeptides are also useful for producing a target for the diagnosis of a disease or predisposition to disease transmitted by the receptor protein, especially in the pancreas, spleen, tonsils and lymph nodes, as well as in the cells of the lineages of megakaryocytes and erythroblasts. Accordingly, methods are provided for detecting the presence, or levels of the receptor protein in a cell, tissue or organism. The method comprises contacting a biological sample with a compound capable of interacting with the receptor protein so that the interaction can be detected. One agent for detecting the receptor protein is an antibody capable of selective binding to the receptor protein. A biological sample includes tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. The receptor protein also produces a target for the diagnosis of an active disease, or predisposition to disease, in a patient who has a variant receptor protein. In this way, the receptor protein can be isolated from a biological sample, analyzed for the presence of a genetic mutation that results in an aberrant receptor protein. This includes substitution, deletion, insertion, or rearrangement of amino acids (as a result of aberrant division events), and inadequate modifications subsequent to translation. Analytical methods include altered electropoietic mobility, tryptic digestion of the altered peptide, altered receptor activity in cell-based or cell-free assays, alteration in the binder, or antibody binding pattern, altered isoelectric point, of direct amino acid sequences, and any other of the known test techniques that are useful for detecting mutations in a protein. In vi tro techniques for the detection of the receptor protein include enzyme-linked immunosorbent assays (ELISAs), Western markers, immuno precipitation and immuno fluorescence. Alternatively, the protein can be detected in a subject by introducing a labeled antireceptor antibody into the subject. For example, the antibody can be labeled with a radioactive label whose presence and location in a subject can be detected by standard imaging techniques. Particularly useful are methods that detect the allelic variant of a receptor protein expressed in a subject and methods that detect the fragments of a receptor protein in a sample. The receptor polypeptides are also useful in • pharmacogenomic analysis. Pharmacogenomics deals with clinically important hereditary variations in response to medications due to an altered disposition to medications and abnormal action in affected individuals. See for example the publication of Eichelbaum (1997) Clin. Exp. • 10 Pharmacol. Phistol. 23 (10-11): pages 983 -985 (1996), and Linder (1997) Clin. Chem 43 (2): pages 254-266. The clinical results of these variations result in severe toxicity to therapeutic drugs in certain individuals, or failure drug therapy in certain individuals as a result of individual variation in metabolism. Therefore, the genotype of the individual can determine the way in which therapeutic compounds act in the body or how that the body metabolizes the compounds. In addition, the activity of the enzymes that metabolize the drug affect both the intensity and duration of the drug's action. Therefore, the pharmacoenomics of the individual allows the selection of effective compounds and effective doses of said compounds for prophylactic and therapeutic treatments based on the individual genotype. The discovery of genetic polymorphism in some enzymes that metabolize drugs has explained why patients do not obtain the expected effects of the drug, show an exaggerated effect of a drug, or experience serious toxicity by the standard doses of the drug. Polymorphisms can be expressed in the genotype of the extensive metabolist and the genotype of the deficient metaboliser. Accordingly, genetic polymorphism can lead to allelic protein variants of the receptor protein in which one or more of the receptor functions in a population is different from those in another population. Therefore the polypeptides allow an objective to ensure a genetic predisposition that can affect the treatment modality. Thus, in a binder-based treatment, the polymorphism can cause extracellular fields of the amino terminal and / or other binding regions of the binder that are more or less active at the binder bond and receptor activation. Therefore, a dose of binder would necessarily be modified to maximize the therapeutic effect within a given population that contains a polymorphism. As an alternative to the elaboration of the qenotypes, specific polymorphic polypeptides could be defined. The receptor polypeptides are also useful for monitoring therapeutic effects during clinical trials and other treatments. Therefore, the therapeutic effectiveness of an agent that is designed to increase or decrease gene expression, protein levels or receptor activity can be monitored during the course of treatment using the receptor polypeptides as an endpoint target. For example, supervision may be in the following manner: (I) obtaining a sample prior to the administration of a subject before administering the therapeutic agent; (ii) detect the level of expression or activity of a specified protein in the sample prior to treatment; (iii) obtaining one or more samples subsequent to the administration of the subject; (iv) detect the level of expression or activity of the protein in the post-administration samples; (v) comparing the level of expression or activity of the protein in the sample prior to administration with the protein of the sample or samples subsequent to administration; and (vi) increasing or decreasing the administration of the agent to the subject in accordance with the above results. The receptor polypeptides are also useful for the treatment of a disease associated with the receptor. Accordingly, the methods of treatment include the use of the soluble receptor or fragments of the receptor protein that compete with the binding of the binder. These receptors or fragments may have a higher affinity for the binder so as to provide effective competition. Antibodies The present invention also provides, antibodies that selectively bind to receptor proteins 2838, 14618 and 15334 as well as variants and fragments. An antibody is considered to bind selectively, even if it also binds to other proteins that are not substantially homologous with the receptor protein. These other proteins share the homology with a fragment or field of the receptor protein. Conservation in specific regions causes antibodies that bind to both proteins under the homologous sequence. In this case, it should be understood that the antibody that binds to the receptor protein is still selective. To generate antibodies, an isolated receptor polypeptide was used in the form of an immunogen to generate antibodies using standard techniques for the preparation of polyclonal and monoclonal antibodies. It can be used either full length proteins, or antigenic fragments of the peptide. Regions having a high index of antigenicity are illustrated in Figures 2, 6, and 10. Antibodies are preferably prepared from those regions of these regions, or from fragments separated in these regions. However, antibodies can be prepared from any region of the peptide as described in the present invention. A preferred fragment produces an antibody that decreases or completely prevents binding of the binder. Antibodies can be developed against the entire receptor or portions of the receptor, for example, the intracellular field of the carboxy terminus, the extracellular field of the amino terminus, the entire transmembrane field or specific segments, and any of the intra- or extracellular coils, or any portion of the above. Antibodies can also be developed against specific functional sites, such as the binding site of the binder, the site of coupling by the G protein, or sites that are • phosphorylates, glycosylates and myristoylates. An antigenic 2838 fragment will generally comprise at least 8 contiguous amino acid residues. The antigenic peptide may, however, comprise a contiguous sequence of at least 12, 14 amino acid residues, at least 15 residues of • 10 amino acids, and at least 20 amino acid residues or at least 30 amino acid residues. In one embodiment, the fragments correspond to regions that are located on the surface of the protein, for example, comprising any fragments that may have been described before the present invention. An anti-acidic molecule 14618 will generally comprise at least 9 contiguous amino acid residues. The antigenic peptide can comprise, without However, a contiguous sequence of at least 12, 14 amino acid residues, at least 15 amino acid residues, and at least 20 amino acid residues or at least 30 amino acid residues. In a preferred embodiment, the fragments correspond to regions that are located on the surface of the protein for example, hydrophilic regions. However, it should not be construed that these fragments comprise any fragments which have been disclosed prior to the present invention. An antigenic 15334 fragment will generally comprise at least 8 contiguous amino acid residues. The antigenic peptide may, however, comprise a sequence of at least 12, 14 contiguous amino acid residues, at least 15 contiguous amino acid residues, at least 20 contiguous amino acid residues or at least 30 contiguous amino acid residues. In one embodiment, the fragments correspond to regions that are located on the surface of the protein, for example, hydrophilic regions. However, it should not be considered that these fragments comprise any fragments which have been disclosed before the present invention. The antibodies can be polyclonal or monoclonal. An intact antibody or fragment thereof (for example Fab or F (ab ') 2) can be used. Detection can be facilitated by coupling (e.g., physically linking) the antibody to a detectable substance. Examples of the substances that can be detected include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase or acetylcholinesterase; examples of the complex prosthetic groups include streptavidin / biotin and avidin / biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, fluorescein dichlorotriazinylamine, dansyl chloride or phycoerythrin; an example of luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive materials include I125, I131, S35 or H3. An appropriate immunogenic preparation can be derived from native peptides, expressed recombinantly, proteins or chemically synthesized.
Uses of the Antibody The antibodies may not be used to isolate a receptor protein by standard techniques, such as affinity chromatography or immunoprecipitation. The antibodies can facilitate the purification of the native receptor protein from the cells, and the recombinantly produced receptor protein expressed in the host cells. Antibodies are useful for detecting the presence of the receptor protein in cells or tissues to determine the pattern of receptor expression between various tissues in an organism and with the course of normal development. Antibodies can be used to detect the receptor protein in si t u, in vi t ro, or in a cellular lysate or supernatant in order to evaluate the abundance and expression pattern. Antibodies can be used to assess abnormal tissue distribution, or abnormal expression during development. The detection of antibodies from the circulating fragments of the receptor protein, of total length, can be used to identify the rotation of the receptor. In addition, the antibodies can be used to evaluate the expression of the receptor in disease conditions, such as active stages of the disease or in an individual who has a predisposition towards the disease related to the receptor function. When a disease is caused by an inappropriate tissue distribution, expression of the development or level of expression of the receptor protein, the antibody can be prepared against the normal receptor protein. If a disease is characterized by a specific mutation in the receptor protein, antibodies specific for this mutant can be used to make an analysis of the presence of the specific mutant receptor protein. The antibodies can also be used to evaluate the normal and aberrant subcellular localization of the cells of the different tissues of an organism. The antibodies can be developed against the total receptor or portions of the receptor, for example, portions of the extracellular field of the amino terminal or extracellular coils. The uses of the diagnosis can be applied, not only in the genetic tests but in the supervision of a treatment modality. Accordingly, when a treatment has finally been achieved to correct the level of expression of the receptor or the presence of aberrant receptors and aberrant tissue distribution or developmental expression, antibodies directed against the receptor, or the fragments relevant to the recipient, can be used. monitor the therapeutic efficacy. Accordingly, antibodies can be used diagnostically to monitor protein levels in the tissue as part of a clinical trial procedure, for example, to determine the efficacy of a given treatment regimen. In addition, antibodies are useful in pharmacogenomic analysis. Therefore, antibodies prepared against polymorphic receptor proteins can be used in order to identify individuals that require modified modalities of treatment. The antibodies are also useful as diagnostic tools as an immunological marker for an aberrant receptor protein analyzed by electrophoretic mobility, isoelectric point, tryptic digestion of the peptide and other clinical assays known to those skilled in the art. Antibodies are also useful for tissue typing. Therefore, in cases where a specific receptor protein has been correlated with expression in a specific tissue, antibodies that are specific for this receptor protein can be used in order to identify the type of tissue. The antibodies are also useful in forensic identification. Accordingly, in cases where an individual with a specific genetic polymorphism that results in a specific polymorphic protein has been linked, an antibody specific for the polymorphic protein can be used as an aid for identification. Antibodies are also useful for inhibiting receptor function, for example, blocking binding of the binder. These uses can also be applied in a therapeutic context in which the treatment comprises the inhibition function of the receptor. For example, an antibody can be used to block the binding of the binder. The antibodies can be prepared against specific fragments containing sites required for the function or against the intact receptor associated with a cell. The fully human antibodies, are particularly desirable for the therapeutic treatment of human patients. For an overview of this technology, to produce human antibodies, consult Lonberg and Huszar's publication (1 995, In. Rev. Imm unol. 1 3: page s 65 -93). For a detailed explanation of this technology to produce human antibodies and human monoclonal antibodies and protocols for the production of said antibodies, see US Pat. No. 5,625,126; U.S. Patent No. 5,633,425; U.S. Patent 5,569,825; U.S. Patent 5,661,016; and U.S. Patent 5,545,806. The present invention also comprises, equipment for using antibodies to detect the presence of a receptor protein in a biological sample. The kit can comprise antibodies such as a labeled or labeled antibody, and a compound or agent for detecting the receptor protein in a biological sample; means for determining the amount of receptor protein in the sample; and means for comparing the amount of the receptor protein in the standard common sample; the compound or agent can be packaged in a suitable container; and the kit may further comprise instructions for using it to detect the receptor protein.
Polynucleotides The nucleotide sequence in SEQ ID NO: 2 was obtained by making the sequence of a full-length human cDNA. The nucleotide sequence of SEQ ID NO: 4 was obtained by elaborating the sequence of a full-length human cDNA. The nucleotide sequence of SEQ ID NO: 6 was obtained by elaborating the sequence of a full-length human cDNA. The specifically described cDNAs comprise the coding region of the 5 'and 3' untranslated sequences (SEQ ID NO: 2, SEQ TD NO: 4 and SEQ ID NO: 6). The human 2838 receptor cDNA is approximately 1617 nucleotides long and encodes a full length protein having a length of approximately 319 amino acid residues. The nucleic acid is expressed in the spleen, testes, colon, and peripheral blood lymphocytes as well as in those tissues illustrated in FIG. 15. The structural analysis of the amino acid sequence of SEQ ID NO: 1 is provided in the filter. 3, a hydropathy graph. The figure shows the putative structure of the seven transmembrane segments, the extracellular field of the amino terminal and the extracellular field of the carboxy terminal.
• The human 14618 receptor cDNA has a length of about 1358 nucleotides and encodes a full length protein having a length of about 337 amino acid residues. The nucleic acid is expressed in the spleen, peripheral blood lymphocytes, breast, skeletal muscle and other tissues shown in Figure 13, as well as several hematopoietic cells illustrated in Figure 14 especially megakaryocytes and CD34 + cells. The structural analysis of the amino acid sequence of SEQ ID NO: 3 is provided in the figure 7, a graph of hydropathy. The figure shows the putative structure of the 7 transmembrane segments, of the extracellular field of the terminal • amino and the intracellular field of the carboxy terminal. The human 153340 receptor cDNA has a length of about 2559 nucleotides and encodes a full length protein having a length of about 372 amino acid residues. The nucleic acid is expressed in the lymph, tonsils, pancreas, colon, spleen, peripheral blood cells, thymus, adrenal and heart glands as well as the cells of the megakaryocytic and erythroid lineages as illustrated in Figures 17 through 19. The structural analysis of the amino acid sequence of SEQ ID NO: 5 is provided in Figure 11, a graph of hydropathy. The figure illustrates the putative structure of the seven transmembrane segments, the extracellular field of the amino terminal and the intracellular field of the carboxy terminal. As used in the present invention, the term (transmembrane segment) refers to amino structural patterns which include a structural pattern of amino acids which includes a hydrophobic helix extending along the plasma membrane. The complete transmembrane field of protein 2838 extends from about amino acid 25 to about amino acid 292. The complete transmembrane field of protein 14618 extends from about amino acid 29 to about amino acid 297. The complete transmembrane field of protein 15334 it extends from about amino acid 26 to about amino acid 299. The seven segments extend along the membrane, and there are three intracellular coils and three extracellular coils in that field.
The present invention provides isolated polynucleotides encoding a 2838 receptor protein. The term "2838 polynucleotide" or "2838 nucleic acid" refers to the sequence illustrated in SEQ ID NO: 2. The present invention provides isolated polynucleotides encoding the receptor protein. 14618. The term "polynucleotide 14618" or "nucleic acid 14618" refers to the sequence shown in SEQ ID NO: 4. The present invention provides isolated polynucleotides that encode a 15334 receptor protein. The term "polynucleotide 15334" or " nucleic acid 15334"refers to the sequence shown in SEQ ID NO: 6. The term" receptor polynucleotide "or" receptor nucleic acid "further includes variants and fragments of polynucleotides 2838, 14618 and 15334.
An "isolated" receptor nucleic acid is one that is separated from another nucleic acid present in the natural source of the receptor nucleic acid. Preferably, an "isolated" nucleic acid is free of sequences which naturally blanch the nucleic acid (eg, the sequences located at the 5 'and 3' ends of the nucleic acid) in the genomic DNA of the organism from which it was derived the nucleic acid. However, there may be some flanking nucleotide sequences, such as up to about 5KB. The important point is that the nucleic acid is isolated from the sequences that flank it so that it can be subjected to the specific manipulations described in the present invention such as recombinant expression, preparations of test substances and preparers, and other specific uses for the nucleic acid receptor sequences. In addition, an "isolated" nucleic acid molecule, such as a cDNA or RNA molecule, may be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized. However, the nucleic acid molecule can be fused to other coding or regulatory sequences and can still be considered isolated. For example, recombinant DNA molecules contained in a vector are considered isolated. Additional examples of isolated DNA molecules include recombinant DNA molecules maintained in heterologous host cells or DNA molecules purified (partially or substantially) in the solution. Isolated RNA molecules include RNA transcripts in vi or in vi tro of the isolated DNA molecules of the present invention.
• Nucleic acid molecules isolated according to the present invention, further include said synthetically produced molecules. In some cases, the isolated material will form part of a composition (for example, a crude extract containing another substance), a system • 10 cushion, or reactive mixture. In other circumstances, the material can be purified to essential homogeneity, for example, as determined by the PAGE method or column chromatography such as HPLC. Preferably a nucleic acid The isolate comprises at least about 50, 80 or 90% (on a molar basis) of all the macromolecular species present. The receptor polynucleotides can encode the mature protein plus amino terminal amino acids or Carboxyl, or amino acids internal to the mature polypeptide, (when the mature form has more than one polypeptide chain). Such sequences may play a role in the processing of a protein from a precursor to the mature form, facilitating the Translating the protein, prolonging or shortening the average life of the protein or facilitating the manipulation of a protein for assay or production, among other things. As is generally the case in si t u, additional amino acids can be processed away from the mature protein by cellular enzymes. The receptor polynucleotides include, but are not limited to, the sequence encoding the mature polypeptide alone, the sequence encoding the mature polypeptide and the encoders of additional sequences, such as a major or secretory sequence (e.g., a pre-protein or pro-sequence). -protein), the sequence encoding the mature polypeptide, with or without additional coding sequences, plus additional sequences that are not coding, for example, introns and 5 'and 3' non-coding sequences so that transcribed but non-translated sequences can play a role in transcription, mRNA processing (including division and polyadenylation signals) of ribosome binding, and mRNA stability. In addition, the polynucleotide can be fused with a marker coding sequence, for example, a peptide that facilitates purification. The receptor polynucleotides may be in the form of an RNA, such as mRNA, or in the form of DNA, including the cDNA and genomic DNA obtained by cloning or produced by synthetic chemical techniques, or by the combination thereof. The nucleic acid, especially the DNA, can be of double strands or single strands. The single stranded nucleic acid can be the coding thread (sense yarn) or the non-coding yarn (antisense yarn). A receptor nucleic acid comprises the nucleotide sequence illustrated in SEQ ID NO: 2, corresponding to human 2838 cDNA. A receptor nucleic acid comprises the nucleotide sequence shown in SEQ ID NO: 4, corresponding to human cDNA 14618. A receptor nucleic acid comprises the nucleotide sequence shown in SEQ ID NO: 6, corresponding to human cDNA 15334. In one embodiment, the receptor nucleic acid comprises only the coding reqion. The present invention further provides variant receptor polynucleotides, and fragments thereof, which differ from the nucleotide sequence illustrated in SEQ ID NO: 2, due to the degeneracy of the genetic code and therefore encode the same protein that was encoded by the nucleotide sequence shown in SEQ ID N0: 2. The present invention also provides variant receptor polynucleotides and fragments thereof that differ from the nucleotide sequence shown in SEQ ID NO: 4, due to the degeneracy of the genetic code and therefore encode the same protein that is encoded by the sequence of nucleotides illustrated in SEQ ID NO: 4. The present invention further provides, variant receptor polynucleotides and fragments thereof, which differ from the nucleotide sequence shown in SEQ ID NO: 6, due to the degeneracy of the genetic code and therefore it encodes the same protein that is encoded by the nucleotide sequence shown in SEQ ID NO: 6. The present invention also provides nucleic acid receptor molecules encoding variant polypeptides as described in the present invention. Said polynucleotides can occur naturally, such as the allelic variants (same position) homologs (different position) and orthologs (different organism) or can be constructed by recombinant DNA methods or chemical synthesis.
Said variants that occur in a way that is not natural can be done by mutagenesis techniques, including those techniques applied to • the polynucleotides, cells or organisms. Therefore, as explained above, the variants may contain substitutions, deletions, inversions and insertions of nucleotides. Variation can occur in either or both of the coding regions and not • 10 encoders. Variations can produce both conservative and non-conservative amino acid substitutions. Generally, the variants have a substantial identity with a nucleic acid molecule selected from the group consisting of SEQ ID NO: S: 2, 4 and 6 and their complements. The orthologous, homologous and allelic variants can be identified using methods well known in the art. Variants 2838 comprise a nucleotide sequence encoding a receptor that is 40-45%, 45-50%, 50-55%, 55-60%, at least about 65%, generally at least less about 70-75%, more generally at least about 80 to 25 85%, and even more generally at least about 90 to 95% or more homologous to the nucleotide sequence illustrated in SEQ ID NO : 2 or a fragment of this sequence. Said nucleic acid molecules can be easily identified as having the ability to hybridize under severe conditions, to the nucleotide sequence shown in SEQ ID NO: 2 or a sequence fragment. Variants 14618 comprise a nucleotide sequence encoding a receptor that is 40-45%, 45-50%, 50-55%, 55-60% at least about 65%, generally at least about 70-75 %, more generally of at least about 80-85%, and even more generally of at least about 90-95%, or more homologous to the nucleotide sequence illustrated in SEQ ID NO: 4 or a fragment of this sequence. Said nucleic acid molecules can be easily identified as having the ability to hybridize under severe conditions, to the nucleotide sequence shown in SEQ ID NO: 4. or a sequence fragment. Variants 15334 comprise a nucleotide sequence encoding a receptor that is 45-50%, 50-55%, 55-60% at least about 65%, generally at least about 70-75%, more generally at least about 80-85%, and even more generally at least about 90-95%, or more homologous to the nucleotide sequence shown in SEQ ID NO: 6 or a fragment of this sequence. Said nucleic acid molecules can be easily identified as having the ability to hybridize under severe conditions, to the nucleotide sequence shown in SEQ ID NO: 6. or a fragment of the sequence. It should be understood that severe hybridization does not indicate substantial homology where it is due to general homology, such as poly A sequences, or sequences common to all or most of the proteins all GPCRs, or the entire family of GPRs I, since still to all the purinoceptors. Furthermore, it should be understood that the variants do not include any of the amino acid sequences that have been disclosed prior to the present invention. As used in the present invention, the term "hybrid under severe conditions" is intended to describe conditions for hybridization and washing under which the nucleotide sequence encoding a receptor polypeptide is at least 50-55%, % homoloqa one of the other generally remain hybridized with each other. The conditions can be such that the sequences in • at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 90%, at least about 95% or more identical one from the other they remain hybridized with each other. Such severe conditions are known to those skilled in the art and can be found in the publication Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), pages 6.3.1 to 6.3.6, incorporated in the present description as reference. An example of the severe hybridization conditions are hybridization in sodium citrate / sodium chloride 6X (SSC) at a temperature of about 45 ° C followed by one or more washes in 0.2 X SSC, 01% SDS at a temperature of 50- 65 ° C. In In another non-limiting example, the nucleic acid molecules are allowed to hybridize in sodium citrate / sodium chloride 6X (SSC) at a temperature of about 45 ° C followed by one or more washings of low stringency in 0.2x SSC / 0.1. % SDS at temperature environment, or by one or more moderate washes in 0. 2X SSC / 0. 1% SDS at a temperature of 42 ° C or washed at 0. 2X SSC / 0. 1% SDS at a temperature of 65 ° C for a high severity. In one modality, the molecule • Isolated nucleic acid receptor that hybridizes under severe conditions to the sequence of SEQ ID NO: S: 2, 4, and 6 corresponds to a nucleic acid molecule that occurs naturally. As used in the present invention, a nucleic acid molecule that "occurs naturally" refers to • the ARD molecule or DNA having a nucleotide sequence that occurs in nature (eg, that encodes a natural protein). As understood by those skilled in the art, the exact conditions can be They are determined empirically and depend on the ionic strength, temperature and concentration of the stabilizer acids such as formide or denaturing agents such as SDS. Other factors that are considered in determining the The desired hybridization conditions include the length of the nucleic acid sequences, the base composition, the percentage of equality between the hybridization sequences and the frequency of the occurrence of subsets of the sequences within other sequences that are not identical. In this way, equivalent conditions can be determined by varying one or more of those parameters while maintaining a similar degree of identity or similarity between the two nucleic acid molecules. The present invention also provides isolated nucleic acids containing a fragment or portion of a single strand of double strand that hybridizes under severe conditions to a nucleotide sequence selected from the group consisting of SEQ ID N0: S: 2, • 10 4, and 6 and the complements of SEQ ID NO: S: 2, 4, and 6. In another embodiment, the nucleic acid consists of a portion of a nucleotide sequence selected from the group consisting of SEQ ID NOS.
NO: S: 2, 4 and 6 and the complements. The nucleic acid fragments of the present invention have a length of at least about 15, • preferably at least about 18, 20, 23 or 25 nucleotides, and may be 30, 40, 50, 100, 200 or more nucleotides. Longer fragments, for example, of a length of 30 or more nucleotides, which encode the antigenic proteins or polypeptides described herein are useful. In addition, the present invention provides polynucleotides comprising a fragment of full length receptor polynucleotides. The fragment may be single or double stranded and may comprise DNA or RNA. The fragment can be derived from either a coding or non-coding sequence. In one embodiment, the receptor nucleic acid 2838 isolated from nucleotide 1 around nucleotide 990 has a length of at least 16 nucleotides that hybridizes under severe conditions to the molecule of • Nucleic acid comprising the nucleotide sequence of SEQ ID NO: 2. In another embodiment, the nucleic acid of about 1487 to 1617 nucleotides is at least 20 nucleotides. In other embodiments, the nucleic acid is of a length of at least 40.50,100, 250 or 500 nucleotide or greater. In another embodiment, the 14168 receptor nucleic acid isolated from about nucleotide 1 to • approximately nucleotide 911 has a length of at least 8 nucleotides and hybridizes under severe conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO: 4. In other embodiments, the nucleic acid has a length of at least 40, 50, 100, 250 or 500 nucleotides or greater.
In another embodiment, the receptor nucleic acid 15334 isolated from nucleotide 1 approximately nucleotide 1355 has a length of about • 18 nucleotides and hybridizes under severe conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO: 6. In another embodiment, the nucleic acid of about nucleotide 868 to about 1355 has at least 11 nucleotides . In other modalities, the nucleic acid • 10 is of a length of at least 40, 50, 100, 250 or 500 nucleotides or greater. In another embodiment, an isolated 2838 receptor nucleic acid encodes the entire coding region from amino acid 1 to amino acid 319. In another In one embodiment, the isolated 2838 receptor nucleic acid encodes a sequence corresponding to the mature protein from about amino acid 6 to about amino acid 319. In another embodiment, the isolated 14618 receptor nucleic acid encodes the The entire coding region from amino acid 1 to amino acid 337. In another embodiment, the isolated 14618 receptor nucleic acid encodes a sequence corresponding to the mature protein from about amino acid 6 to about Amino acid 337. In another embodiment, an isolated 15334 receptor nucleic acid encodes the entire coding region from amino acid 1 to amino acid 372. In another embodiment, nucleic acid • Isolated receptor 15334 encodes a sequence corresponding to the mature protein from about amino acid 6 to about amino acid 372. Other fragments of all three receptors include nucleotide sequences encoding the P 10 amino acid fragments described herein. In addition, the fragments may include their fragments of the specific fields or sites described in the present invention. The fragments also include nucleic acid sequences corresponding to specific amino acid sequences described above or fragments thereof. It should not be considered that • the nucleic acid fragments, according to the present invention, comprise those fragments that may have been disclosed prior to the present invention. However, it should be understood that a receptor fragment includes any nucleic acid sequence and does not include complete qen.
The nucleic acid fragments of the 2838 receptor further include sequences corresponding to the fields described herein, subregions also described, and specific functional sites. The nucleic acid fragments of the 2838 receptor include nucleic acid molecules that encode a polypeptide comprising the extracellular field of the amino terminal including amino acid residues from 1 to about 24, a polypeptide comprising the region extending along of the transmembrane field (amino acid residues from about 25 to about 292), a polypeptide comprising the intracellular field of the carboxy terminus (amino acid residues from about 293 to 319), and a polypeptide encoding the receptor signature of G protein (118-120 or amino acid residues surrounding it from about 107 to about 123), nucleic acid molecules that encode any of the seven transmembrane segments, intracellular or extracellular spirals, deglicosylation sites, phosphorylation sites, sites of myristoylation and amidation sites. The nucleic acid fragments of the 14618 receptor include nucleic acid molecules that encode a polypeptide comprising the extracellular field of the amino terminal including the amino acid residues from 1 to • approximately 28, a polypeptide comprising the region extending along the transmembrane field (amino acid residues from about 29 to about 297), a polypeptide comprising the intracellular field of the carboxy terminal, (residues of amino acids from • about 298 to about 337), a polypeptide encoding the G protein receptor signature (120-122 or surrounding amino acid residues from about 110 to about 132), nucleic acid molecules that encode any of the seven transmembrane segments, extracellular or intracellular spirals, glycosylation sites and phosphorylation sites. The nucleic acid fragments of the receptor 15334 include nucleic acid molecules that encode a polypeptide comprising the extracellular field of the amino terminus including the amino acid residues from 1 to about 25, a polypeptide comprising the Region that extends along the transmembrane field (amino acid residues from about 26 to about 299), a polypeptide comprising the intracellular field of the carboxy terminus (amino acid residues from about 300 to about 372), and a polypeptide that encodes the G-protein receptor signature (118 to 120 or surrounding amino acid residues from about 110 to about 130), the nucleic acid molecules that encode any of the seven transmembrane segments, the extracellular or intracellular spiral, qylosylation sites, kinase C phosphorylation sites of the protein, cAMP, cGMP, and casein kinase II, and myristoylation sites. When the location of the fields has been predicted by computer analysis, a person skilled in the art would appreciate that the amino acid residues that constitute these fields may vary depending on the criteria used to define the fields. The receptor nucleic acid fragments also include combinations of the fields, segments, coils and other functional sites that have been described above. Thus, for example, a receptor nucleic acid would include sequences corresponding to the extracellular field of the amino terminal and a transmembrane element. A person skilled in the art would be warned of the many permutations that are possible. Where the location of the fields or sites has been predicted in computer analysis, a person skilled in the art would appreciate that the amino acid residues that constitute these fields may vary depending on the criteria used to define the fields. The present invention also provides receptor amino acid fragments, which encode epitope-bearing regions of the receptor protein described herein. The polynucleotide sequences of the isolated receptor, and especially the fragments, are useful as DNA test substances and primers. For example, the coding region of a receptor gene can be isolated using the known nucleotide sequence to synthesize an oligonucleotide test substance. A labeled test substance can then be used to select a cDNA, genomic, or mRNA library to isolate the nucleic acid corresponding to the coding region. In addition, primers can be used in PCR reactions to clone specific regions of the receptor genes. A test substance / preparation generally comprises that the oligonucleotide is substantially purified. Oligonucleotide 2838 generally comprises a region in the nucleotide sequence from 1 to 990, or from 1487 to 1617 that hybridizes under severe conditions to at least about 16 and 20, respectively generally about 25, more generally about 40, 50 or 75 nucleotides consecutive of SEQ ID NO: 2, sense or antisense strands or other receptor polynucleotides. Oligonucleotide 14618 generally comprises a region of the nucleotide sequence 1 to 911 that hybridizes under severe conditions to at least about 9., 12, generally about 25 and more generally about 40, 50 or 75 consecutive nucleotides of SEQ ID NO: 4, and sense or antisense or other receptor polynucleotides. Oligonucleotide 15334 generally comprises a region of the nucleotide sequence from 868 to 1355 that hybridizes under severe conditions to at least about 11.12, usually about 25, more generally about 40, 50, or 75 consecutive nucleotides of the SEQ ID NO: 6, and sense or antisense or other receptor polynucleotides. Oligonucleotide 15334 also generally comprises a region of the nucleotide sequence of nucleotide 1 through 1355 that hybridizes under severe conditions to at least about 18, typically about 25, and more generally about 40, 50 or 75 consecutive nucleotides of SEQ. ID NO: 6, and sense or antisense or other receptor polynucleotides.
Uses of the Polynucleotide. The nucleic acid sequences of the present invention can also be used "sequence of doubt" to carry out a search against public databases to, for example, identify other members of the family or related sequences. Said searches can also be carried out using the NBLAST and XBLAST (version 2.0) methods of Altschul and Associates (1990) J. Mol. Biol. 215: pages 403 to 410. BLAST searches of nucleotides can be performed with the NBLAST program, score = 100, word length = 12 to obtain homologs of nucleotide sequences to the nucleic acid molecules of the present invention.
To obtain differentiated alignments for comparison purposes, the differentiated BLAST can be used as described by Altschul and Associates (1997) Nucleic Acids Res. 25 (17): pages 3389 to 3402. When BLAST and differentiated BLAST programs are used, the default parameters of the respective programs can be used (for example XBLAST and NBLAST). Consult http: / www. nebi nlm. nih gov. The nucleic acid fragments of the present invention provide test preparation substances in the assays such as those described below. The "test substances" oligonucleotides that hybridize in a specific manner in the base to a complementary thread of the nucleic acid. Said test substances include polypeptide nucleic acids, as described by Nielsen and Associates (1991) Science 254: pages 1497 to 1500. Generally, a test substance comprises a region of the nucleotide sequence that hybridizes under highly stringent conditions to at least about 15, usually about 20 to 25, and more generally about 40, 50? 75 consecutive nucleotides of a nucleic acid selected from the group consisting of SEQ ID NO: S: 2, 4 and 6, and the complements thereof. More generally, the assay sequence further comprises a label, e.g., radioisotope, fluorescent compound, enzyme, or enzyme cofactor. As used in the present invention, the term "preparer" refers to a single-stranded oligonucleotide which acts as a starting point for template-driven DNA synthesis using well known methods (eg, PCR, LCR) including but not limited to those described herein. The appropriate length of the preparer depends on the particular use, but is generally in a range of about 15 to 30 nucleotides. He term "preparer site" refers to the area of Target DNA to which the preparer hybridizes. He "Term" preparer pair "refers to a set of preparers that includes a 5 'preparer. (ascending) that hybridizes with the 5 'terminal of the nucleic acid sequence to be amplified and a 3 'preparer (downstream) that hybridizes with the complement of the sequence to be amplified. The receptor polynucleotides are useful for test substances, preparers, and in assays biological.
In cases where the polynucleotides are used to evaluate the properties and functions of the GPCR, such as in the assays described above, all or less than all of the complete cDNA 5 may be useful. In this case, even the arrangements that may have been known before the present invention are included. Thus, for example, assays directed specifically to GPCR functions, such as activities of agonist or antagonist evaluation, comprise the use of known fragments. In addition, diagnostic methods to evaluate the receptor function can also be practiced with any fragment, including those fragments that may have been known before the present invention. In a similar way, the methods that comprise the treatment of a receptor dysfunction, all fragments • are included, including those that may have been known in the field. The 2838 receptor polynucleotides are useful as hybridization assay substances for cDNA and qenomic DNA to isolate the full-length cDNA and the genomic clones encoding the polypeptide described in SEQ ID NO: 1, and to isolate the cDNA and the genomic clones corresponding to the variants that produce the same polypeptides shown in SEQ ID NO: 1, or the other variants described herein. The variants can be isolated from the same tissue and organism from which the polypeptide illustrated in SEQ ID NO: 1, different tissues of the same organism, or from different organisms was isolated. The 14618 receptor polynucleotides are useful as hybridization test substances for cDNA and genomic DNA to isolate the full-length cDNA and the genomic clones encoding the polypeptide described in SEQ ID NO: 3, and to isolate the cDNA and the qenomic clones. which correspond to the variants that produce the same polypeptide shown in SEQ ID NO: 3 or the other variants described herein. The variants can be isolated from the same tissue and organism, of which the polypeptide shown in SEQ ID NO: 3 was isolated, different tissues of the same organism, or from different organisms. The 15334 receptor polynucleotides are useful as a hybridization assay substance for the cDNA and genomic DNA to isolate the full-length cDNA and the genomic clones encoding the polypeptide described in SEQ ID NO: 5 and to isolate the cDNA and the clones. genomes corresponding to the variants that produce the same polypeptide as shown in SEQ ID NO: 5 or the variants described herein. The variants can be isolated from the same tissue and organism from which the polypeptide shown in SEQ ID NO: 5, different tissues of the same organism, or from different organisms was isolated. This method is useful for isolating genes and cDNAs that are controlled in a developmental fashion and therefore can be expressed in the same tissue or different tissues at different points in the development of an organism. The sample substance may correspond to any sequence along the entire length of the gene encoding the receptor. Accordingly, it could be derived from the 5 'non-coding regions, the coding region, and the 3' non-coding regions. The substance demonstrates, however, that it should not be construed as corresponding to any of the sequences that may have been known prior to the present invention. The nucleic acid assay substance 2838 may be, for example, the full-length cDNA of SEQ ID NO: 1 or a fragment thereof, such as an oligonucleotide of at least 11, 16, 18, 20, 30 , 50, 100, 250 or 500 nucleotides in length and sufficient to hybridize specifically under severe conditions to mRNA or DNA. The nucleic acid test substances 14618 can be, for example, the full length cDNA of SEQ ID NO: 3 or a fragment thereof, such as an oligonucleotide of at least 8, 12, 15, 30, 50 , 100, 250, or 500 nucleotides in length and sufficient to hybridize specifically under severe conditions to mRNA or DNA. The nucleic acid test substance 15334 can be, for example, the full-length cDNA of SEQ ID NO: 5, or a fragment thereof, such as an oligonucleotide of at least 11, 12, 18, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to hybridize specifically under severe conditions to mRNA or DNA. The fragments of the polynucleotides that are described in the present invention are also useful for synthesizing larger fragments, or the full length polynucleotides described herein. For example, a fragment can be hybridized to any portion of a mRNA and a larger or full length cDNA can be produced. The fragments are also useful for synthesizing antisense molecules of the desired length and sequence.
The antisense nucleic acids of the present invention can be designated using the nucleotide sequence of SEQ ID NO: S: 2, 4 and 6 constructed using chemical synthesis and enzymatic ligation reactions using methods known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or modified nucleotides in various manners designed to increase the biological stability of the molecule or to increase the physical stability of the duplex formed between the molecules. antisense and sense nucleic acids, for example, phosphorothioate derivatives and nucleotides substituted by acridine can be used. Examples of the modified nucleotides that can be used to generate antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5- (carboxyhydroxymethyl) uracil, 5-carboxymethylaminomethyl -2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, -methycytosine, N6-adenine, 7-methylguanine, 5-ethyl laminometauracil, 5-methoxyminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyl luracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, acid of uracil-5-oxyacetic acid (v), wibutoxosin, pseudouracil, kerosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methyl ester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-3 (3-amino-3-N -2-carboxypropyl) uracil, (acp3) w, and 2,6-diaminopin. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector within which the nucleic acid has been subcloned in an antisense orientation (eg, the RNA transcribed from an inserted nucleic acid will be antisense-oriented to the target nucleic acid of interest. ). Additionally, the nucleic acid molecules of the present invention can be modified in the base portion, sugar portion or phosphate structure to improve, for example, the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate structure of the nucleic acids can be modified to generate nucleic acid peptides (see Hyrup and Associates (1996) Bioorganic &Medicinal Chemistry 4: page 5). As used in the present invention, the term "peptide nucleic acids" or "PANs" refers to nucleic acid mimics, for example, DNA mimics, in which the deoxyribose phosphate structure is replaced by a pseudopeptide structure and only the four natural nucleobases are retained. It has been shown that the neutral structure of the PANs allows the specific hybridization of DNA and RNA under conditions of low ionic strength. The synthesis of the PNA oligomers can be carried out using standard synthesis protocols of solid base peptides as described by Hyrup and Associates (1996), mentioned above; Perry-0 'Keefe and Associates (1996) Proc. Nati Acad. Sci. USA 93: page 14670.
The PANs can be further modified, for example, to increase their stability, specificity or cellular assimilation, by adhering lipophilic or other auxiliary groups to the PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other administration techniques. of drugs known in the art. The synthesis of the PNA-DNA chimeras can be carried out as described by Hyrup (1996), mentioned above Finn and Associates (1996) Nucleic Acids Res. 24 (17): pages 3357 to 3363, Mag and Associates ( 1989) Nucleic Acids. Res. 17: page 5973 and Peterser and Associates (1975) Bioorganic • Med. Chem. Lett. 5: page 1119. The nucleic acid molecules and fragments of the present invention may also include other adhered groups such as peptides (eg, receptors in vi vo of target host cells), or agents that facilitate membrane-wide transport. cell phone (see, for example, Letsinger and Associates (1989) Proc. Nati, Acad. Sci. USA 86: pages 6553 to 6556; Lemaitre and Associates (1987) Proc. Nati. Acad. Sci. USA 84: pages 648 to 652; PCT publication No. WO 88/0918) or the blood barrier of the brain (see for example, PCT publication No. WO 89/10134). In addition, the oligonucleotides can be modified with washing agents detonated by hybridization (see for example, Krol and Associates (1998) Bio-Techniques 6: pages 958 to 976) or intercalation agents (see for example Zon). (1988) Pharm Res. 5: pages 539 to 549). The receptor polynucleotides are also preparers for the PCR test in order to amplify a particular region of a receptor polynucleotide.
The receptor polynucleotides are also useful for the construction of recombinant vectors. Such vectors include expression vectors that express a portion of, or all of the receptor polypeptides. The vectors also include insertion vectors, used to integrate into other polynucleotide sequences, such as within the cell genome, to alter the expression and in si t of the receptor genes and gene products. For example, a coding sequence of the endogenous gene can be replaced by homologous recombination by all or part of the coding region that contains one or more specifically introduced mutations. The receptor polynucleotides are also useful for expressing antigenic peptides. The regions of the peptides having a high index of antigenicity are shown in Figures 2, 6 and 10. The receptor polynucleotides are also useful as a test substance for determining the position of the chromosomes of the receptor polynucleotides by means of hybridization methods. you, see for example. FISH (for a review of this technique, see Verma and Associates (1998) Human Chromosomes: A Manual of Basic Techniques (Pergamon Press, New York), and the development of PCR maps of somatic cell hybrids. the sequences to chromosomes, is an important first step in the co-relation of these frequencies with the genes • associated with the disease. 5 Reagents can be used for chromosome mapping individually to mark a single chromosome, or a single chromosome site, or reagent panels can be used to mark multiple sites and / or multiple chromosomes. The • 10 reagents corresponding to the non-coding regions of the genes are currently preferred for mapping purposes. The coding sequences are more likely to be conserved within the gene families, thus increasing the opportunity for cross-hybridizations during the elaboration of the chromosomal map. Once the map of a sequence has been treated at a precise chromosomal location, the The physical position in the chromosome sequence can be correlated with the genetic map data. (Such data are found, for example, in V. McKusick, Mendelian Inheritance in Man, available on the line through Jhons Hopkins University Welch Medical Library). In relation between a gene and a disease, plotted on a map to the same chromosomal region then it can be identified through a linkage analysis (co-inheritance, or genes) • physically adjacent), described in for example, 5 Egeland and Associates (1987) Nature 325: pages 783 to 787. In addition, differences in DNA sequences between affected and unaffected individuals can be determined with a disease associated with a gene • 10 specific. If any mutation is observed in any or all of the affected individuals but not in any of the unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. The comparison of individuals affected and unaffected usually comprises first observing the structural alterations in the chromosomes, such as deletions or translocations that are in a visible form of chromosome broadcasts, or detectable using the PCR based on that DNA sequence. Finally, an elaboration of complete sequences of the qen of several individuals can be carried out to confirm the presence of a mutation and to distinguish the mutations of the polymorphisms.
The test substances of the receptor polynucleotide are also useful for determining patterns of the presence of the gene encoding the receptors • and its variants with respect to tissue distribution, for example, if gene duplication has occurred, and if duplication occurs in all or only a subset of tissues. Genes may occur naturally or may have been introduced into a cell, tissue, or organism in a hexogenic manner.
• The receptor polynucleotides are also useful for designing ribozymes corresponding to all or a portion of mRNA produced from the genes encoding the polynucleotides described herein. The receptor polynucleotides are also useful for constructing host cells that express a part, or all of the receptor polynucleotides and polypeptides. The receptor polynucleotides are also useful for constructing transgenic animals expressing all or part of the receptor polynucleotides or polypeptides. The receptor polynucleotides are also useful for making vectors that express part or all of the receptor polypeptides.
The receptor polynucleotides are also useful as a hybridization assay substance to determine the level of receptor nucleic acid expression. Accordingly, the test substances can be used to detect the presence of, or to determine levels of, receptor nucleic acids in cells, tissues and in organisms. The nucleic acid whose level is determined can be DNA or RNA. Accordingly, the sample substances corresponding to the polypeptides described herein can be used to evaluate the number of copies of the gene in a given cell, tissue or organism. This is particularly important in the case where there has been an amplification of the receptor genes. Alternatively, the test substance can be used in a context of in si t u hybridization. To evaluate the position of extra copies of the receptor genes, as in extrachromosomal elements or as integrated within chromosomes in which the receptor gene is not normally found, for example as in a region of homogeneous coloration. These uses are important for the diagnosis of the disease comprising an increase or decrease in the expression of the receptor in relation to normal results, such as a proliferative disease, a differential or developmental disease, or a hematopoietic disease. Therefore, the present invention provides a method for identifying a disease or condition associated with an expression or aberrant activity of the receptor nucleic acid, in which the test sample is obtained from a subject and the nucleic acid (e.g., mRNA, genomic DNA) of detected, wherein the presence of the nucleic acid is a diagnosis for a subject having or at risk of developing a disease or condition associated with the expression or aberrant activity of the nucleic acid. One aspect of the present invention relates to diagnostic assays for determining nucleic acid expression, as well as activity in the context of a biological sample (e.g., blood, serum, cells, tissues) to determine whether an individual has a disease or condition, or is at risk of developing a disease or condition, associated with the expression or aberrant activity of the nucleic acid. Said assays can be used for prognostic or predictive purposes to treat them prophylactically to an individual before the presentation of a condition characterized by or associated with the expression or activity of the nucleic acid molecules. The in vi tro techniques for mRNA detection include Northern hybridizations, and in si t u hybridizations. The in vi tro techniques for determining DNA include Southern hybridizations and in si t u hybridizations. The test substances can be used as part of a diagnostic test kit to identify cells or tissues expressing a receptor protein, such as by measuring a level of a nucleic acid encoding the receptor in a sample of cells from a subject , for example, the mRNA or genomic DNA or the determination of whether a receptor gene has been mutated. Nucleic acid expression assays are useful for the selection of drugs to identify compounds that modulate the expression of the receptor nucleic acid (eg, antisense, polypeptides, peptide mimetizations, small molecules or other drugs). A cell is contacted with a candidate compound, and expression of the mRNA is determined. The level of expression of the receptor mRNA in the presence of the candidate compound is compared to the level of expression of the receptor mRNA in the absence of the candidate compound. The candidate compound can then be • identified as a modulator of nucleic acid expression based on this comparison and used, for example, to treat a condition characterized by an aberrant expression of the nucleic acid. The modulator can be linked to the nucleic acid or indirectly modulate the expression, such as by interaction with another cellular component that affects the expression of the nucleic acid. Modulatory methods can be performed in vi tro, (for example, by cultivating the cell with the agent) or, alternatively in vi, (for example, administering the agent to a subject) in patients or in transgenic animals. Therefore the present invention provides a method for the identification of a compound that can be used to treat a condition associated with the expression of the nucleic acid of the receptor gene. The method generally includes testing the ability of the compound to modulate the expression of the receptor nucleic acid and thus identify A compound that can be used to treat a condition characterized by an unwanted expression of the receptor nucleic acid. Assays can be carried out, in cell-based and cell-free systems, cell-based assays include cells that naturally express the receptor nucleic acid, or recombinant cells genetically engineered to express the specific nucleic acid sequences. Alternatively, candidate compounds can be tested in patients or in transgenic animals. Assays of receptor nucleic acid expression may comprise direct assays of nucleic acid levels, such as mRNA levels, or a collateral compound comprised in the signal path (such as a cyclic AMP, or a phosphatidylnositol rotation) . In addition, the expression of genes that are sub-classified in response to the signal path of the receptor protein can also be tested. In this embodiment, the regulatory regions of these genes can be operably linked to a reporter gene such as luciferase. In this way, modulators of receptor gene expression can be identified in a method where a cell is contacted with a candidate compound, and the expression of the mRNA is determined. The level of expression of the receptor mRNA in the presence of the candidate compound is compared to the level of expression of the receptor mRNA in the absence of the candidate compound. The candidate compound can then be identified as a modulator of nucleic acid expression based on this comparison and used, for example, to treat a condition characterized by an aberrant expression of the nucleic acid. When the expression of mRNA is statistically significantly greater in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulant in the expression of the nucleic acid. When expression of the nucleic acid is statistically significantly lower in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of nucleic acid expression. Accordingly, the present invention provides methods of treatment, with nucleic acid as a target, using a compound identified through the selection of drugs as a modulator of the gene for modulating the expression of the receptor nucleic acid. Modulation includes both overqualification (for example activation or agonization) or down-regulation (deletion or antagonization) or • effects on nucleic acid activity (for example, when the nucleic acid is mutated or modified incorrectly). Treatment is the conditions characterized by the expression or aberrant activity of the nucleic acid. Alternatively, a modulator of the expression of the P receptor nucleic acid can be a small molecule or a drug identified using the screening assays described herein to the extent that the drug or small molecule inhibits the expression of the receptor nucleic acid. The receptor polynucleotides are also useful for monitoring the effectiveness of the modulator compounds in the expression or activity of the receptor gene in clinical trials, or in a treatment regimen. In this way, the expression pattern of the The gene can serve as a barometer for the continued effectiveness of the treatment with the compound, particularly with compounds in which the patient can develop resistance. The pattern of gene expression can also serve as a marker indicating a physiological response of the affected cells to the compound. Accordingly, such monitoring would allow either increased administration of the compound or administration of alternative compounds to which the patient has not become resistant. In a similar way, if the level of expression of the nucleic acid falls below the desirable level, the administration of the compound can be commensurately decreased. Supervision may be, for example, in the following manner: i) obtaining a sample prior to the administration of a subject prior to administration of the agent; ii) detection of the level of expression of a specified mRNA or a genomic DNA of the invention in the sample prior to administration; iii) obtain one or more samples subsequent to the administration of the subject; iv) detect the level of expression or activity of the mRNA or genomic DNA in the samples after administration; v) comparing the level of expression or activity of mRNA or genomic DNA in the sample or samples subsequent to administration; and vi) increase or decrease the administration of the agent to the subject in accordance with these results. The receptor polynucleotides are also useful in diagnostic assays for qualitative changes in the receptor nucleic acid, and particularly in qualitative changes leading to the pathology the polynucleotides can also be used for • detect mutations in the receptor genes and in the 5 gene expression products such as mANR. The polynucleotides can also be used as hybridization sample substances to detect genetic mutations that occur naturally in the recipient gene and by means of them determine whether a ^ P 10 subject with the mutation is at risk for a dse caused by the mutation. . Mutations include, deletion, addition or substitution of one or more nucleotides in the gene, chromosomal rearrangement, such as inversion or transposition, modification of genomic DNA, such as an aberrant pattern of methylation, or changes in the number of copies of the gene, such as amplification. The detection of a mutated form of the receptor gene associated with a dysfunction, provides a diagnostic tool for a Active dse or susceptibility to dse when the dse is the result of overexpression, subexpression, or altered expression of a receptor protein. Mutations in the receptor gene can be detected at the nucleic acid level by a variety of techniques. Genomic DNA can be analyzed directly, or it can be amplified by using PCR before analysis. The RNA or the cDNA can be used in the same way. In certain embodiments, detection of the mutation compr the use of a test substance / preparer in a polymerase chain reaction (PCR) (see, for example, U.S. Patent Nos. 4,683,195 and 4,683,202), such as P 10 as PCR anchor or PCR RACE, or alternatively, in a ligation chain reaction (LCR) (see for example, Landegran and Associates (1988) Science 241: pages 1077 to 1080, and Nakazawa and Associates (1994) PNAS 91: pages 360 to 364), the last of which may be particularly useful for the detection of the mutation points in the gene (see Abravaya and Associates (1995) g ^ Nucleic Acids Res. 23: 675 to 682). This method may include the steps of collecting a sample of cells from a patient, isolating the acid Nucleic acid (eg, genomic, mRNA or both) of the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridizes to a gene under conditions so that hybridization or amplification of the qen occurs.25 (if present) and the detection of the presence or absence of an amplification product, or the detection of the size of the amplification product and the comparison of the length as a control sample. Suppressions and insertions can be detected by a change in the size of the amplified product purchased with a normal genotype. Mutation sites can be identified by hybridizing amplified DNA to a normal sequence RNA or antisense DNA. It is anticipated that PCR and / or LCR may be desirable to be used as a preliminary amplification step in conjunction with any of the techniques used for the detection of mutations described herein. Alternative methods of amplification include: duplication of self-sustaining sequences (Guatelli and Associates (1990) Proc. Nati. Acad. Sci.
USA 87: pages 1874 to 1878) transcription amplification system (Kwoh and Associates (1989) Proc. Nati, Acad. Sci. USA 86: pages 1173 to 1177), Replicase Q-Beta (Lizardi and Associates (1988) Bio / Technology 6: page 1197), or other methods of nucleic acid amplification, followed by the detection of amplified molecules using techniques well known to those skilled in the art. These detection schemes are especially useful for the detection of nucleic acid molecules, if said molecules are present in very low numbers. Alternatively, mutations of a gene The receptor can be identified directly, for example, by altering the restriction enzyme digestion patterns determined by gel electrophoresis. In addition, the specific ribozymes of the sequence ^ P 10 (US Patent No. 5,498,531) can be used to qualify the presence of specific mutations by developing or losing a ribozyme wash system. The perfectly collated sequences can be distinguished from the unequal sequences by the nuclease washing digestion assays or by differences in melting temperatures. • Sequence changes at specific locations can also be evaluated through the 20 nuclease protection assays, such as RNase and SI protection, or chemical washing methods. In addition, sequence differences between the mutant receptor gene and a wild-type gene can be determined by sequence processing of direct DNA. A variety of automated sequencing procedures can be used when carrying out diagnostic tests ((1995) Biotechniques 19: page 448), including the elaboration of sequences by mass spectrometry (see for example the international publication PCT No WO 94/16101; Cohen and Associates (1996) Adv. Chromatogr. 36: pages 127 to 162; and Griffin and Associates (1993) Appl. Biochem. Biotechnol. 38: pages 147 to 159). Other methods for the detection of mutations in the gene include methods in which the protection of washing agents is used to detect uneven bases in the RNA / RNA duplexes or in the RNA / DNA duplex (Myers and Associates (1985) Science 230 : page 1242); (Cotton and Associates (1988) PNAS 85: pages 4397; Saleeba and Associates (1992) Meth. Enzimol. 217: pages 286 to 295), the electrophoretic mobility of a mutant and a wild type nucleic acid is compared (Orita y Asociados (1989) PNAS 86: page 2766, Cotton and Associates (1993) Mutat, Res. 285: pages 125 to 144, and Hayashi and Associates (1992) Genet, Anal. Tech. Appl. 9: pages 73 to 79), and the movement of fragments of a mutant or wild type in the polycrylamide gels containing a denaturation gradient is tested using denaturing gradient gel electrophoresis (Myers and Associates (1985) Nature 313: page 495). The sensitivity of the assay can be increased by using RNA (instead of DNA) in which the secondary structure is more sensitive to a change in sequence. In a modality, the method uses heteroduplex analysis to separate double stranded heteroduplex molecules based on the change in electrophoretic mobility (Keen and Associates (1991) Trends Genet, 7: page 5). Examples of other techniques for detecting mutant sites include, selective oligonucleotide hybridization, selective amplification, and selective extension of the separator. In other embodiments, genetic mutations can be identified by the hybridization of a sample and the control nucleic acid, for example, RNA or DNA, in high density arrays containing hundreds or thousands of oligonucleotide test substances (Cronin and Associates). (1996) Human Mutation 7: pages 244 to 255; Kozal and Associates (1996) Nature Medicine 2: pages 753 to 759). For example, genetic mutations can be identified in bi-dimensional distributions containing DNA test substances generated by light as described in Cronin and Associates, previously mentioned. Briefly, a first hybridization distribution of the test substance • can be used to scan through 5 long stretches of DNA in a sample, and control to identify base changes between sequences by making linear distributions of test substances that overlap the sequence. This step allows the identification of the points of ^ P 10 mutation. The step is followed by a second hybridization arrangement that allows the characterization of specific mutations using specialized, smaller test substance adjuncts complementary to all variants or mutations detected. Each mutation distribution is composed of sets of parallel test substance, one complementary to the wild-type gene and the other complementary to the mutant gene. The receptor polynucleotides are also useful to test an individual for a genotype that while not necessarily causing the disease, nevertheless affects the treatment modality. In this way, the polynucleotides can be used to study the relationship between the genotype of the individual, and the response of the individual to a compound used for the treatment (far acogenomics relationship). In the present case, for example, a mutation in the receptor gene that results in an altered affinity for the binder may result in an excessive or decreased effect of the drug with standard concentrations of the binder that activates the receptor. Accordingly, the receptor polynucleotides described herein can be used to evaluate the content of the recipient gene mutation in an individual for the purpose of selecting an appropriate compound, and a dosage regimen for the treatment. Therefore, polynucleotides that exhibit genetic variations that affect the treatment provide a diagnostic objective that can be used to design the treatment in an individual. Accordingly, the production of recombinant cells, and animals containing these polymorphisms will allow the clinical design effect of the treatment compounds and dosage regimens. The methods may comprise obtaining a biological sample from a control subject, which is in contact with the control sample with a compound or agent capable of detecting the mRNA or genomic DNA, so that the presence of the mRNA or mRNA is detected. the genomic DNA in the biological sample, and compare the presence of the mRNA or genomic DNA in the control sample with the presence of the mRNA or DNA genomic in the test sample. The receptor polynucleotides are also useful for the identification of chromosomes, when the sequence is identified with an individual chromosome and at a particular location on the chromosome. "First, the presence of DNA is matched to the chromosome by hybridization in itself or another specific hybridization of the chromosome, and the sequences can be correlated with specific chromosomes, preparing PCR primers that can be used for PCR selection of somatic cell hybrids, which contain individual chromosomes of the desired species. Only hybrids that contain the chromosome that contains the homologous of the gene to the preparer, will produce a fragment amplified. Sublocalization can be achieved, using chromosomal fragments. Other strategies include, preselection with chromosomes classified by marked flow, pre-selection by hybridization to libraries specific chromosomes. In addition, mapping strategies include fluorescence hybridization in si t u, which allows hybridization with test substances shorter than those traditionally used. Reagents 5 can be used for the elaboration of chromosome maps individually to mark a single chromosome, or a single site on the chromosome, or reagent panels can be used to mark multiple sites and / or multiple chromosomes. The reagents corresponding to • 10 non-coding regions of genes are currently preferred for mapping purposes. The coding sequences are more likely to be conserved within the families of queens, thereby increasing the opportunity for cross-hybridizations during the elaboration of chromosomal maps. The receptor polynucleotides can also be • used to identify individuals from small biological samples. This can be done for example, using restriction fragment length polymorphism (RFLP), to identify an individual. Thus, the polynucleotides described herein are useful as DNA markers for RFLP (See US Patent No. 5,272,057).
In addition, the receptor sequence can be used to provide an alternative technique which determines the actual DNA sequence of the selected fragments in an individual's genome. Therefore, the receptor sequences described herein can be used to prepare two PCR primers from the 5 'and 3' ends of the sequences. Then these primers can be used to amplify the DNA of an individual, for the subsequent elaboration of sequences. Panels of the corresponding DNA sequences of individuals prepared in this way can provide unique individual identifications, since each individual will have a unique set of said DNA sequences. It is estimated that allelic variations in humans occur at a frequency of about once per 500 bases. Allelic variations occur to some degree in the coding regions of these sequences, and to a greater degree in the non-coding regions. The receptor sequences can be used to obtain said identification sequences of individuals and tissues. The sequences represent unique fragments of the human genome. Each of the sequences described herein can, to some extent, be used as a standard against which the individual DNA can be compared for identification purposes. If a panel of reagents of the sequences is used to create a database of unique identification for an individual, those same reagents can be used later to identify the tissues of said individual. The use of unique identification databases, positive identification of the individual, living or dead, can be made from extremely small tissue samples. The receptor polynucleotides can also be used in forensic identification procedures. PCR technology can be used to amplify DNA sequences taken from very small biological samples, such as a single hair follicle, body fluids, (eg, blood, saliva, or semen). Then the amplified sequence can be compared with a standard, allowing the identification of the origin of the sample. The receptor polynucleotides can therefore be used to provide polynucleotide reagents, for example, PCR primers focused on the specific location in the human genome, which can increase the reliability of forensic identification based on DNA, for example, by providing another " "identification tag" (for example another DNA sequence that is unique to a particular individual). As described above, the current information of the base sequence can be used for identification as an exact alternative for patterns formed by restriction fragments enzymes. The sequences focused on the non-coding reqion are particularly useful since a greater polymorphism occurs in the non-coding regions, making it easier to differentiate individuals using this technique. The fragments are at least 12 bases.
The receptor polynucleotides can be used additionally, to provide polynucleotide reagents, e.g., labeled, or screened, test substances, in which, for example, an in si t u hybridization technique can be used to identify a specific tissue. This is useful in the case in which the forensic pathology is presented with a tissue of unknown origin. The panels of receptor assay substances can be used to identify tissues by species and / or by type of organ. In a similar embodiment, these primers and test substances can be used to screen tissue cultures for contamination (e.g., to select the presence of a mixture of different types of cells in a culture). Alternatively, the receptor polynucleotides can be used directly to block the transcription or translation of the receptor sequences by means of the antisense or ribozyme constructs. Therefore, in a disease characterized by an abnormally high or undesirable expression of the receptor gene, the nucleic acids can be used directly for treatment. The receptor polynucleotides are therefore useful as antisense constructs to control the expression of the receptor gene in cells, tissues and organisms. An antisense DNA polynucleotide is designed to be complementary to a region of the gene comprised in transcription, preventing transcription and thus production of the receptor protein. An antisense RNA or DNA polynucleotide would hybridize to the mRNA and thus block the translation of the mRNA within the receptor protein. Examples of the antisense molecules useful for inhibiting the expression of the nucleic acid include 5 antisense molecules complementary to a fragment of the 5 'region not translated from the IDS. SEC NOS: 2, 4, or 6 which also includes the start codon, and the antisense molecules which are complementary to a fragment of the 3 'region. transferred from the IDS. SEC NOS: 2, 4, or 6. Alternatively, a class of antisense molecules can be used to inactivate the mRNA in order to decrease the expression of the receptor nucleic acid. Therefore, these molecules can to treat a condition characterized by abnormal or undesired expression of the receptor nucleic acid. This technique comprises washing by means of ribozymes containing the sequences of nucleotides complementary to one or more regions in the mRNA that decrease the ability of the mRNA to be transferred. Possible regions include the coding regions and particularly the coding regions corresponding to the catalytic activities and other functional activities of the receptor protein, such as binding to the binder.
The receptor polynucleotides also produce vectors for the kenotic therapy in patients that contain cells that have an aberrant expression in the receptor gene. In this way, the recombinant cells, which include the patient's cells that have been designed ex vivo and returned to the patient, are introduced into the individual where the cells produce the desired receptor protein to treat the individual. The present invention also comprises equipment for detecting the presence of a receptor nucleic acid in a biological sample. For example, the kit may comprise reagents such as a nucleic acid or labeled agent or which can be labeled with the ability to detect the receptor nucleic acid in a biological sample; means for determining the amount of receptor nucleic acid in the sample; and means for comparing the amount of receptor nucleic acid in the sample with a standard. The compound or agent can be packaged in a suitable container. The equipment may further comprise instructions for the use of the equipment to detect the mRNA or receptor DNA.
Computer-readable Media The nucleotide or amino acid sequences of the present invention are also pded in a variety of means to facilitate the use thereof. As used in the present invention, "pded" refers to a manufacturing, except an isolated nucleic acid or amino acid molecule, which contains a nucleotide or amino acid sequence of the present invention. Such fabrication pdes the nucleotide or amino acid sequences or a subset thereof (eg, a subset of open reading frames (ORFs)) in a manner which allows a person skilled in the art to examine manufacturing using means that do not. they are directly applicable to examine the nucleotide or amino acid sequences, or a subset thereof, since they exist in a natural or purified form. In an application of this embodiment, a nucleotide or amino acid sequence of the present invention can be qravada in a computer reading means. As used in the present invention, "means of computer readings" refers to any medium that can be read and accessed directly by a computer. Such means include, but are not limited to: magnetic storage media, such as floppy disks, hard disk storage media, and magnetic tapes; optical storage media such as CD-ROM; electrical storage means such as RAM and ROM; and hybrids of these categories such as magnetic / optical storage media. Those skilled in the art will readily appreciate the way in which any of the computer-readable media known today can be used to create a fabrication comprising a computer-readable medium that is recorded therein, a nucleotide or amino acid sequence of the present invention. As used in the present invention, "etching" refers to a process for storing information in a computer-readable medium. Those skilled in the art can readily adopt any of the currently known methods for measuring information in a computer-readable medium, for the purpose of generating fabrications comprising the nucleotide or amino acid sequence information of the present invention. .
A variety of these storage structures are available to those skilled in the art to create a computer readable medium which has a nucleotide or amino acid sequence of the present invention etched therein. The choice of data storage structure will generally be based on the means selected to access the stored information. In addition, a variety of data processing programs and formats can be used to store the nucleotide sequence information of the present invention in a computer readable medium. The sequence information can be represented in a word processing text file, formatted in software that can be obtained in the market such as a WordPerfect and Microsoft Word, or represented in the form of an ASCII code file, saved in a database application such as DB2, Sybase, Oracle, or similar. Those skilled in the art can easily adapt any of a number of data processing structuring formats (e.g., text file or database) in order to obtain computer readable media that has taxed thereon. the information of the nucleotide sequence of the present invention. By providing the nucleotide and amino acid sequences of the present invention in a computer-readable form, the skilled artisan can routinely access the sequence information for a variety of purposes. For example, a person skilled in the art can use the nucleotide or amino acid sequence of the present invention in a computer readable form to compare a target sequence or an objective structural pattern with the sequence information stored within the media. of data storage. Search means are used to identify fragments or regions of the sequences of the first invention, which coincide with a particular target sequence, or an objective pattern. As used in the present invention, an "objective sequence" can be any DNA sequence or amino acids of six or more nucleotides, or two or more amino acids. A person skilled in the art can easily recognize that the longer an objective sequence is, the less likely it is to be present as a random occurrence in the database. The most preferred length of the sequence of an objective sequence is from about 10 to 100 amino acids, or from about 30 to 300 nucleotide residues. However, it is well recognized that commercially important fragments, such as fragments of sequences comprised in gene expression and protein processing may still be of shorter length. As used in the present invention, a "target structural pattern" or "objective pattern" refers to any sequence or combination of rationally selected sequences in which the sequence (s) are selected based on a three-dimensional configuration which it is formed by bending the objective pattern. There is a variety of objective patterns known in the art. Target protein patterns include, but are not limited to, active sites of enzymes, and signal sequences. Target nucleic acid standards include, but are not limited to, promoter sequences, orquilla structures and inducible expression elements (protein binding sequences). A computer software is available publicly which allows an expert in the art to have access to the information of the sequence provided in a medium that can be read by computer for analysis and comparison with other sequences. A variety of known algorithms, and a variety of commercially known software for performing search means are disclosed publicly which are and can be used in computer-based systems of the present invention. Examples of such software include but are not limited to, MacPattern (EMBI), BLASTN and BLASTX (NCBIA). For example, the software which implements BLAST (Altschul and associates (1990) J. Mol. Biol. 215: pages 403 to 410) and BLAZE (Brutlaq and associates (1993) Comp.Chem. 17: pages 203 to 207) it looks for algorithms in a Sybase system which can be used to identify the open reading frames (ORFs) of the sequences of the present invention which contain homology with the ORFs or proteins of other libraries. Said ORFs are proteins that encode fragments and are useful in the production of commercially important proteins such as the enzymes used in various reactions and in the production of metabolites which are commercially useful.
Vectors / host cells The present invention also provides vectors containing the receptor polynucleotides. The term "vector" refers to a carrier, preferably a nucleic acid molecule, that can transport the receptor polynucleotides. When the vector is a nucleic acid molecule, the receptor polynucleotides are covalently linked to the vector nucleic acid. With this aspect of the present invention, the vector includes a plasmid, a single or double-stranded phage, a viral RNA or single-strand or double-stranded DNA vector, or artificial chromosome, such as a BAC, PAC, YAC, O MAC. A vector can be maintained in the host cell as an extrachromosomal element which duplicates and • produces additional copies of the receptor polynucleotides. Alternatively, the vector can be integrated into the genome of the host cell and produce additional copies of the receptor polynucleotides when the host cell is duplicated. The present invention provides vectors for maintenance (cloning vectors) or vectors for expression (expression vectors) of receptor polynucleotides. Vectors can function in prokaryotic or eukaryotic cells or in both (source vectors). Expression vectors contain, cis-regulatory regulatory regions that are operably linked in the vector to the receptor polynucleotides so that transcription of the polynucleotides in a host cell is allowed. The polynucleotides can be introduced into the host cells with a separate polynucleotide capable of affecting transcription. In this way, the second polynucleotide can provide a trans acting factor that interacts with the regulatory control region to allow transcription of the receptor nucleotides from the vector. Alternatively, a trans acting factor can be delivered by host cell. Finally, a trans acting factor can be produced from the vector itself. However, it should be understood that in some of the embodiments, transcription and / or translation of the receptor polynucleotides may occur in a cell-fsystem. The regulatory sequence to which the polynucleotides described in the present invention can be operably linked include promoters to direct the transcription of mRNA. These include but are not limited to, the left promoter of the bacteriophage ?, the lac, TRP promoters, and TAC of E. col i, the first and the last promoter from the SV40, the first immediate promoter of CMV, the early and later promoters of adenoviruses, and the long terminal repeats of retroviruses. In addition to controlling regions that promote transcription, expression vectors may also include transcriptionally modulating reqions, such as repressor binding sites, and enhancers. Examples include the SV40 enhancer, the cytomegalovirus immediate early enhancer, the polyoma enhancer, adenovirus enhancers, and LTR retrovirus enhancers. In addition to containing sites for the initiation and control of transcription, the expression vectors may also contain the sequences necessary for transcription termination and a ribosome binding site in the region transcribed for translation. Other regulatory control elements for expression include initiation and termination codons as well as polyadenylation signals. A person skilled in the art would be warned of the numerous regulatory sequences that are useful in expression vectors. Said regulatory sequences are described, for example, in the book by Sambrook and associated Molecular Cloning: A Laboratory Manual (Molecular Cloning: A Laboratory Manual) 2nd. edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, (1989). A variety of expression vectors can be used to express a receptor polynucleotide. Such vectors include chromosomal, episomal, and virus derivatives, for example vectors derived from bacterial plasmids, from bacteriophages, from yeast episomes, from yeast chromosomal elements, including artificial yeast chromosomes from viruses such as ba cul ovi ruses, papova viuses such as SV40, Vaccinia vi ruses, adenovi ruses, poxviuses, pseudorabi viruses, re troviuses. Vectors can also be released from combinations of these sources such as those derived from plasmids and bacteriophage genetic elements, e.g., cosmids and phagemids. The appropriate cloning and expression vectors for prokaryotic and eukaryotic hosts are described in the book by Sambrook and associates, Molecular Cloning: A Laboratory Manual (Molecular Cloning: A Laboratory Manual) 2nd. edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, (1989). The regulatory sequence can provide constitutive expression in one or more host cells (for example tissue-specific) or can provide inducible expression in one or more cell types such as by temperature, nutrient additive, or exogenous factor such as a hormone or other binder. A variety of vectors that provide constitutive or inducible expression in prokaryotic and eukaryotic hosts are well known to those skilled in the art. The receptor polynucleotides can be inserted into the vector nucleic acid by a well-known methodology. Generally, the DNA sequence will be finally expressed as linked to an expression vector by washing the DNA sequence, and the expression vector with one or more restriction enzymes and then the binding of the fragments together. The procedures for restriction enzyme digestion and ligation are well known to those skilled in the art. The vector containing the appropriate polynucleotide can be introduced into a host cell suitable for propagation or expression using well known techniques. Bacterial cells include, but are not limited to, E. col i, S trept omyces, and Sa lmonel l a typh im uri um. Eukaryotic cells include, but are not limited to, yeast, insect cells such as Drosophila, animal cells such as COS and CHO cells, and plant cells. As described in the present invention, it may be desirable to express the polypeptide as a fusion protein. Accordingly, the present invention provides fusion vectors that allow the production of the receptor polypeptides. The fusion vectors can increase the expression of a recombinant protein, increase the solubility of the recombinant protein, and aid in the purification of the protein by acting, for example, as a binder for affinity purification. A proteolytic wash site can be introduced at the junction of the fusion portion so that the desired polypeptide can finally be separated from the fusion portion.
Proteolytic enzymes include, but are not limited to, factor Xa, thrombin, and enterokinase. Typical fusion expression vectors include pGEX (Smith and associates (1988) Gene 67: pages 31 to 40), pMAL (New England Biolabs, Beverly, MA) and pRIT5 (Pharmacia, Piscataway, NJ) which fuse glutathione S-transferase (GST), the protein of maltose E bond, or protein A, respectively, to the target recombinant protein. Examples of inducible expression vectors that are not E expression. Col i inducible non-fusion include pTrc (Amann and associates (1988) Gene 69: pages 301 to 315) and pET lid (Studier and associates (1990) Gene Expression Technology: Methods in Enzymology (Gene Expression Technology: Methods in Enzymology) 185: pages 60 to 89). The expression of the recombinant protein can be maximized in a host bacterium by providing a genetic background where the host cell has a damaged ability to proteolytically clean the recombinant protein. (Go t tesman Gene Expression Technology: (Gene Expression Technology: Methods in Enzymology 185), Academic Press, San Diego, California (1990) pages 119 to 128). Alternatively, the sequence of the polynucleotide of interest can be altered to produce the preferential use of the codon for a specific host cell, for example E. col i. (Wada and associates (1992) Nucleic Acids Res. 20: pages 2111 to 2118).
The receptor polynucleotides can also be expressed by expression vectors that operate in yeast. Examples of expression vectors in yeast, for example S. cerevi if ae includes pYepSecl (Baldari, and associates (1987) EMBO J. 6: pages 229 to 234), pMFa (Kurjan and associates (1982) Cell 30: pages 933 to 943), pJRY88 (Schultz and associates (1987) Gene 54: pages 113 to 123), and pYES2 (Invitrogen Corporation, San Diego, CA). The receptor polynucleotides can also be expressed in insect cells using, for example, baculovirus expression vectors. Baculovirus vectors are available for the expression of proteins e? cultured insect cells (e.g., Sf9 cells), include the pAc series (Smith et al. (1983) Mol. Cell Biol. 3: pages 2156 to 2165) and the pVL series (Lucklow and associates (1989) Virology 170: pages 31 to 39). In certain embodiments of the present invention, the polynucleotides described herein are expressed in mammalian cells using mammalian expression vectors. Examples of mammalian expression vectors include pCDM8 (Seed (1987) Nature 329: page 840) and pMT2PC (Kaufman et al. (1987) EMBO J. 6: pages 187 to 195).
The expression vectors mentioned herein are provided by way of example only of the well-known vectors available to those • experts in the art that would be useful for expressing the receptor polynucleotides. Those skilled in the art will be informed of other suitable vectors for the maintenance, propagation or expression of the polynucleotides described herein. These are found, for example, in Sambrook's book, and ^ P 10 associated Molecular Cloning: A Laboratory Manual (T. Molecular Cloning: A Laboratory Manual) 2nd. edition, Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989. The present invention also comprises vectors in which the nucleic acid sequences described herein are cloned within the vector in reverse orientation, but which are linked operationally to a regulatory sequence that allows the transcription of antisense RNA. Therefore, an antisense transcript can be produced for all or a portion of the polynucleotide sequences described herein, including both coding regions and non-coding regions. coding. The expression of this antisense RNA is subject to each of the parameters described above in relation to the expression of sense RNA (regulatory sequences, constitutive or inducible expression, and tissue-specific expression). The present invention also relates to recombinant host cells containing the vectors described herein. Host cells therefore include prokaryotic cells, lower eukaryotic cells such as yeast, other eukaryotic cells such as insect cells, higher eukaryotic cells such as mammalian cells. The recombinant host cells are prepared by introducing the construction of the vector described in the present invention, into the cells by techniques that are readily available to a person skilled in the art. These include, but are not limited to, calcium phosphate transfection, DEAE-dexthand-transmitted transfection, cationic lipid-mediated transfection, electroporation, transduction infection, lipofection, and other techniques such as those found in Sambrook's book. and Associated Molecular Cloning: A Laboratory Manual (Mol ecul ar Cl oning: A Labora tory Manual), 2nd edition, (Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989.) Host cells can contain more than one vector, therefore different nucleotide sequences can be introduced into different vectors of the same cell. In a similar manner, the receptor polynucleotides can be introduced either alone, or with other polynucleotides that are not related to the receptor polynucleotides such as those that provide trans acting factors for the expression vectors. When more than one vector is introduced into a cell, the vectors can be introduced independently, co-introduced or bound to the vector of the receptor polynucleotide. In the case of bacteriophage and viral vectors, these can be introduced into the cells as vectors packaged or encapsulated by standard procedures for infection and transduction. Viral vectors may be competent for duplication, or defective for duplication. In the case where the vector is defective for duplication, duplication will occur in the host cells providing functions that complement the defects.
The vectors generally include selectable markers, which make possible the selection of the subpopulation of cells containing the ^ ("" * recombinant vector constructs. "Marker 5 may be contained in the same vector that contains the polynucleotides described by the present invention or may be in a separate vector Markers include tetracycline, or ampicillin resistance genes for the host cells prokaryotes and dihydriflate reductase or neomycin resistance for eukaryotic host cells. However, any marker that provides the selection for phenotypic treatment may be effective. 15 Although mature proteins can be produced in bacteria, yeast and mammalian cells, and other cells under the control of the appropriate sequences, cell-free transcription and translation systems can also be used to produce these proteins using the RNA derived from the DNA constructs described in the present invention. Where secretion of the polypeptide is desired, secretion signals are incorporated into the vector appropriate. The signal sequence may be endogenous to the receptor polypeptides, or heterologous to these polypeptides. Where polypeptides are not secreted inside • of the medium, the protein can be isolated from the host cell by standard interruption procedures, including freezing, sonication, mechanical interruption, using lysing agents and the like. The polypeptides can then be recovered and purified by well known purification methods, including ammonium sulfate precipitation, acid extraction, anion exchange or cation chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, Hydroxylate chromatography, lectin chromatography, or high performance liquid chromatography. It should also be understood that depending on the host cell, it is the recombinant production of the polypeptides described in the present invention, The polypeptides may have various glycosylation patterns, depending on the cell, or they may be non-glycosylated, as when they are produced in bacteria. In addition, the polypeptides may include an initial methionine modified in some cases as result of a process transmitted by the guest.
Uses of vectors and host cells It should be understood that the terms "cells "Host" and "recombinant host cells" refer not only to the particular subject cells, but also to the offspring or potential offspring of said cells, as certain modifications in successive cells may occur due either to mutation or to environmental influences. said descent may, in fact, not be identical to the originating cell, but may still be included within the scope of the term as used in the present invention.The host cells expressing the polypeptide described herein, and particularly the recombinant host cells have a variety of uses Firstly, the cells are useful for producing receptor proteins or polypeptides that can be further purified to produce the desired amounts of the protein or receptor fragments. Expression vectors are useful for the production of polypeptides. host cells are also useful for conducting tests based on cells comprising fragments of receptors or receptors. Thus, a recombinant host cell expressing a native receptor is useful for testing compounds that stimulate or inhibit receptor function. This includes ligand links, gene expression, transcription or translation levels, the interaction of the G protein, and components of the signal transduction pathway. The host cells are also useful for identifying mutant receptors in which these functions are affected. If the mutants occur naturally and cause a pathology, the host cells containing the feedback are useful for testing the compounds that have a desired effect on the mutant receptor (e.g., stimulation or inhibition of function) which can not be indicated by their effect on the native receiver. Recombinant host cells are also useful for expressing chimeric polypeptides described herein, to evaluate the compounds that activate or suppress activation by means of a heterologous extracellular field of the amino terminal (or other binding region). Alternatively, the heterologous region extending over the entire membrane field (or parts thereof) can be used to evaluate the effect of a desired extracellular amino terminal field (or other binding region) or any given host cell. In this embodiment, the region extending along the entire transmembrane field (or parts thereof) compatible with the specific host cell is used to make the chimeric vector. Alternatively, a heterologous intracellular carboxy terminal field, eg, signal transduction, can be introduced into the host cell. In addition, mutant receptors can be designed in which the increase or decrease of one or more of the different functions (eg, binding, or G protein binding) and used to increase or replace the receptor proteins in an individual is designed. . In this way, host cells can produce a therapeutic benefit by replacing an aberrant receptor or by providing an aberrant receptor that provides a therapeutic result. In one embodiment, the cells provide receptors that are abnormally active. In another embodiment, the cells provide receptors that are abnormally inactive. These receptors can fight with endogenous receptors in the individual.
In another embodiment, cells that express receptors that can not be activated are introduced into an individual for the purpose of • compete with the endogenous receptors for the binder. For example, in the case where the excessive binder is part of a treatment modality, it may be necessary to inactivate this binder at a specific point in the treatment. It would be beneficial to provide cells that compete for the binder, but which can not be affected by the activation of the receiver. Recombinant host cells can also be produced in a homologous manner that allow the in situ alteration of the endogenous sequences of receptor polynucleotides in a genome of the host cell. The host cell includes but is not limited to, a stable cell line, in vi ve cells, or cloned micro organisms. This technology is described more fully in the documents WO 93/09222, WO 91/12650, WO 91/06667, U.S. Patent 5,272,071 and U.S. Patent 5,641,670. Briefly, the specific polynucleotide sequences corresponding to the polynucleotides or nearby receptor sequences or distant to a receptor gene allows them to be integrated into the genome of the host cell by homologous recombination, where the expression of the gene can be affected. In one embodiment, regulatory sequences are introduced that either increase or decrease the expression of an endogenous sequence. Accordingly, a receptor protein can be produced in a cell that does not produce it in a normal manner. Alternatively, it may affect the increased expression of the receptor protein in a cell that normally produces the protein at a specific level. In addition, expression can be decreased or eliminated by introducing a specific regulatory sequence. The regulatory sequence can be heterologous to the sequence of the receptor protein, or it can be a homologous sequence with a desired mutation that affects the expression. Alternatively, the entire gene can be deleted. The regulatory sequence can be • specific to the host cell, or have the ability to function in more than one cell type. Also I know can introduce specific mutations into any desired region of the gene to produce mutant receptor proteins. Such mutations could be introduced, for example, within specific functional regions such as the site link to the binder.
In one embodiment, the host cell can be a fertilized stem or oocyte (ovum) or embryo cell that can be used to produce a transgenic animal that contains an altered receptor gene. Alternatively, the host cell can be a stem cell or other early tissue precursor that originates a specific subset of the cells and can be used to produce transgenic tissues in an animal. See also Thomas and Associates (1987) Cell 51: page 503, for a description of homologous recombination vectors. The vector is introduced into an embryonic stem cell line (for example by electroporation) and the cells in which the gene is introduced are recombined in a manner homologous to the endogenous receptor gene that is selected (see for example, Li and Associates ( 1992) Cell 69: page 915). Then the selected cells are injected into a blastocyst of an animal (for example, a mouse) to form aggregation chimeras (see for example, Bradley's publication (Teratocarcinomas, and Embryonic Stem Cells: A Practical Method) (Tera plays rcinoma s and Embryon i c Stem Cel l s: A Pra c ti ca l Approa ch) (E. Robertson ed. (IRL, Oxford, 1987) pp 113 to 152). Then a chimeric embryo can be implanted into a suitable pseudopregnant female breeding animal, and the embryo can be carried to term. It is possible to use the offspring that make the bridge of the recombinant DNA in a homologous way in their germ cells, to breed animals in which all the cells of the animal contain the recombinant DNA in a homologous manner by transmitting the transgene in the germline. Methods of constructing homologous recombination vectors, and recombinant homologous animals are further described in the publication by Bradley (1991) Curren t <; Opinion on Bi o technolgy 2: pages 823 to 829, and PCT International Publications Nos. WO 90/11354; WO 91/01140; and WO 93/04169. The genetically engineered host cells can be used to produce transgenic non-human animals. A transgenic animal is preferably a mammal, for example a rodent, such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. A transgene is an exogenous DNA which is integrated into the genome of a cell from which the transgenic animal develops and which remains in the genome of the mature animal in one or more of the cell types or tissues of the transgenic animal. These animals are useful for studying the function of a receptor protein and for identifying and evaluating activity modulators • of the receptor protein. 5 Other examples of transgenic animals include primates, sheep, dogs, cows, goats, chickens and non-human amphibians. In one embodiment, the host cell is a fertilized oocyte (ovum) or an embryonic stem cell ^ P 10 into which receptor polynucleotide sequences have been introduced. A transgenic animal can be produced by introducing the nucleic acid into the male polynucleus of a fertilized oocyte (ovum), for example, by microinjection, retroviral infection and allowing the oocyte (ovum) to develop into a pseudo-pregnant female breeding animal. Any of the receptor nucleotide sequences can be introduced as a transgene within the genome of a non-human animal, such as a mouse. Any of the regulatory sequences or of the other sequences useful in the expression vectors can be part of a transgenic sequence. This includes intron sequences, and polyadenylation signals, if not were already included. A tissue-specific regulatory sequence (s) can be operably linked to the transgene to direct expression of the receptor protein to particular cells. Methods • for the generation of transgenic animals by manipulation of the embryo and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in US Patent Nos. 4,736,866 and 4,870,009, both issued to Leder and ^ 10 Associates, U.S. Patent No. 4,873,191 issued to Wagner and Associates, and in Hogan's book: Manipulation of the Mouse Embryo, (Cold Spring Laboratory Press, Cold Spring Harbor, NY, 1986). Methods are used similar for the production of other transgenic animals. A transgenic founder animal can be identified based on the presence of the transgene in its genome and / or the expression of the transgenic mRNA in the tissues or cells of the animals. Then the animal Transgenic founder can be used for breeding additional animals that carry the transgene. In addition, transgenic animals that carry a transqen can additionally breed other transgenic animals that carry other transqenes. A The transgenic animal also includes animals in which the whole animal or animal tissues have been produced using the homologous recombinant host cells described in the present invention. In another embodiment, transgenic non-human animals can be produced in a manner that contains selected systems which allow regulated expression of the transgene. An example of such a system is the cre / xp oxP recombinase system of the bacteriophage Pl. For a description of the cre / l oxP recombinase system, see for example, the publication by Lakso and Associates (1992) PNAS 89: pages 6232 a 6236. Other examples of a recombinase system, is the FLP recombinase system of S. cerevi sia e (O'Gorman y Asociados (1991) Sci en ce 251: pages 1351 to 1355). If the cre / l oxP recombinase system is used to regulate the expression of the transgene, the animals containing the transgenes encode both the Cre recombinase and a selected protein that is required. Such animals can be produced through the construction of "double" transgenic animals, for example, by pairing two transgenic animals, one containing the gene encoding a selected protein and the other containing a transgene encoding a recombinase.
The clones of the non-human transgenic animals described in the present invention can also be produced according to the methods described in • Wilmut and Associates publication (1997) Na ture 385: 5 pages 810 to 813, and PCT international publications Nos. WO 97/07668 and WO 97/07669. Briefly, a cell, for example a somatic cell, from a transgenic animal can be isolated and induced to exit the growth cycle, and enter the cell. • 10 G ° phase. Then the inactive cell can be fused, for example, through the use of electrical impulses, to an oocyte (ovule) enucleated from an animal of the same species from which the inactive cell was isolated. The reconstructed oocyte (ovum) Then it is cultivated so that it develops the morula or blastocyst and is then transferred to a pseudopregnant female breeding animal. The offspring born of this female breeding animal will be a clone of the animal from which the cell was isolated, for example, the cell somatic. The transgenic animals that contain recombinant cells expressing the polypeptides described in the present invention are useful for performing the assays described herein, in a context, in vi vo. By Consequently, the different physiological factors that are present in vi and that could affect the binding of the ligand, the activation of the receptor and the transduction of the signal may not be evident in ^ P cell-free or cell-based assays. Therefore, it is useful to provide non-human transgenic animals for the in vitro testing of receptor functions, including the interaction of the binder, the effect of specific mutant receptors on receptor function and the interaction of the binder, and the effect of chimeric receptors. It is also possible to evaluate the effect of null mutations, this is mutations that substantially or completely eliminate one or more of the functions of the receptor. In general, methods for producing transgenic animals include the introduction of a nucleic acid sequence according to the present invention, the ability of the nucleic acid sequence to express the receptor protein in a transgenic animal, cell in culture or in vi vo. When it is introduced in vi, the nucleic acid is introduced into an intact organism so that one or more types of cells and, consequently, one or more types of tissues, express the nucleic acid that encodes the receptor protein. Alternatively, the nucleic acid can be introduced virtually into all cells in an organism, transfecting a cell in culture, such as an embryonic stem cell, as described in the present invention, for the production of transgenic animals, and This cell can be used to produce a complete transgenic organism. As described, in a further embodiment, the host cell can be a fertilized oocyte (ovum). These cells are allowed to develop in a female breeding animal to produce the transgenic organism.
Pharmaceutical Compositions The receptor nucleic acid molecules, proteins (particularly fragments such as the extracellular field of the amino terminal), modulators of the proteins and antibodies (also preferred in the present invention as "active compounds") can be incorporated into suitable pharmaceutical compositions for administration to a subject, for example a human. Said compositions generally comprise the nucleic acid molecule, protein, modulator or antibody in a pharmaceutically acceptable carrier.
As used in the present invention, "pharmaceutically acceptable carrier" is intended to include any and all solvents, media, • dispersion, coatings, antibacterial and antifungal agents, isotonic delay and absorption agents, and the like, compatible with pharmaceutical administration. The use of said media and agents for pharmaceutically active substances is well known in the art. Except to the extent that any conventional means or agents are compatible with the active compound, said means may be used in the compositions of the present invention. Active compounds can also be incorporated into said compositions supplementary A pharmaceutical composition of the present invention is formulated to be compatible with its intended route of administration. Examples of administration routes - include parenteral, for example intravenous, intradermal, subcutaneous, oral (eg inhalation), transdermal (topical) transmucosal, and rectal administration. The solutions or suspensions used for parenteral, intradermal or subcutaneous application may include the following: components: a sterile diluent, such as aqua for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycols, or other synthetic solvents; antibacterial agents such as benzyl alcohol, or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfate; chelating agents such as ethylenediaminetetraacetic acid, buffers such as acetates, citrates or phosphates and tonicity adjusting agents such as sodium chloride or dextrose. The pH can be adjusted with acid or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be packaged in ampules, disposable syringes, or multiple dose containers made of glass or plastic. Pharmaceutical compositions suitable for use by injection include sterile aqueous solutions (where water is soluble) or sterile dispersions and powders for the extemporaneous preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, (Cremophor Eltm) (BASF, Parsippany, NJ) or phosphate regulated salt (PBS). In all cases, the composition must be sterile, and must be fluid to the point that it is easy to inject. It must also be stable under manufacturing and storage conditions and may be preserved against the action of contamination of microorganisms such as bacteria and fungi. The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (eg, glycerol, propylene glycol, liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion or by the use of a surfactant (surfactant). The prevention of the action of microorganisms can be achieved by different antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol, ascorbic acid, thimerosal and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be achieved by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incorporating the active compound (e.g., a receptor protein or an anti-receptor antibody) in the required amount in an appropriate solvent with one or a combination of ingredients as listed above, as required, followed by the filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the other ingredients required from those mentioned above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying, and drying by conqelation which produces a powder of the active ingredient plus any additional ingredients desired from a sterile solution previously filtered. thereof. Oral compositions generally include an inert diluent or an edible vehicle. They can also be stored in gelatin capsules, or compressed into tablets. For oral administration, the agent may be contained in enteric forms to survive the stomach or additionally coated or mixed to be delivered to a particular region of the gastrointestinal tract by known methods. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, pills or capsules. The oral compositions can also be prepared using a liquid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally, and sorbed and expectorated or swallowed. Pharmaceutically compatible binding agents, and / or adjuvant materials can be included as part of the composition. Tablets, pills, capsules, pills and the like can contain any of the following ingredients, or compounds of a similar nature: a linker such as microcrystalline cellulose, tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or wheat starch; a lubricant such as magnesium stearate or sterols; a bulking agent such as colloidal silicon dioxide; a sweetening agent, such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavor.
For administration by inhalation, the compounds are administered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant; for example, a gas such as carbon dioxide or a nebulizer. Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, appropriate penetrants are used for the barrier that is to be permeated in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be achieved through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated in ointments, ointments, gels, or creams as are generally known in the art. The compounds can also be prepared in the form of suppositories, (for example with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal administration.
In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as controlled release formulations, including, microencapsulated delivery systems or implants. Biodegradable, biocompatible polymers can be used, such as vinyl acetate, ethylene, polyanidrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. The methods for the preparation of said formulations will be appreciated by those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Suspensions of liposomes, (including liposomes that will be sent to monoclonal antibody-infected cells to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in US Pat. No. 4, 522, 811. It is especially advantageous to formulate the oral and parenteral compositions in dosage forms. unit for ease of administration and uniformity of dosage. The unit dosage form as used in the present invention refers to physically adapted separate units • as unit dosages for the subject who is going to be treated; each unit containing a predetermined amount of the active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the unit dose forms of the present invention are indicated by and directly depend on the exclusive characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the matter for the preparation of compounds of said active ingredient for the treatment of individuals. The nucleic acid molecules of the present invention can be inserted into vectors and used as gene therapy vectors. The gene therapy vectors can be administered to a subject for example, by intravenous injection, local administration (US Pat. No. 5,328,470) or by stereotactic injection (see for example, Chen et al.
Associates (1994) PNAS 91: pages 3054 to 3057). The pharmaceutical preparation of the gene therapy vector may include the gene therapy vector in an acceptable diluent, or may comprise, a slow release matrix in which the gene delivery vehicle has been embedded. Alternatively, where the intact whole gene delivery vector of recombinant cells is produced, for example retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system. The pharmaceutical compositions can be included in a container, package, or supplier along with the administration instructions. As defined in the present invention, the therapeutically effective amount of the protein or polypeptide (e.g., an effective dose) is in a range of about 0.001 to 30 mg / kg body weight, preferably about 0.01 a 25 mg / kg body weight, more preferably from about 0.1 to 20 mg / kg body weight and still more preferably from about 1 to 10 mg / kg, from 2 to 9 mg / kg, from 3 to 8 mg / kg kg, from 4 to 7 mg / kg, or from 5 to 6 mg / kg per body weight.
Those skilled in the art will appreciate that certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or condition, prior treatments, the general health and / or age of the subject, and other diseases present. In addition, treatment of a subject with a therapeutically effective amount of a protein, polypeptide or antibody may include a single treatment or, preferably, may include a series of treatments. In a preferred example, a subject is treated with the antibody, protein or polypeptide in a range of between about 0.1 to 20 mg / kg of body weight, once a week for a period of between about 1 to 10 weeks, preferably from 2 to 8 weeks, more preferably from 3 to 7 weeks, and even more preferably from about 4, 5 or 6 weeks. It will also be appreciated, that of the effective dose of the antibody, protein or polypeptide used for the treatment may increase or decrease during the course of a particular treatment. Changes in dosage may result and be appreciated from the results of the diagnostic tests described in the present invention.
The present invention comprises agents which modulate the expression or activity. An agent, for example, can be a small molecule. For example, said small molecule includes but is not limited to peptides, peptide mimetizations, amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, nucleotides, nucleotide analogs, organic or inorganic compounds (eg, including hetero-organic and organometallic compounds) which have a lower molecular weight of about 10,000 grams per mole, organic or inorganic compounds having a molecular weight of less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight of less than about 1000 grams per mole, organic or inorganic compounds which have a molecular weight of less than about 500 qurames per mole, and salts, esters and other pharmaceutically acceptable forms of said compounds. It should be understood that the appropriate dose of small molecule agents depends on a number of factors within the knowledge of an experienced physician, veterinarian or researcher. The dose of the small molecule will vary, for example, depending on the identity, size and condition of the subject or sample being treated, depending in addition on the route by means of which the composition is to be administered, if applicable, and the effect which the doctor wishing the small molecule in the nucleic acid or polypeptide of the present invention wishes. Exemplary doses include milligram or microgram quantities of the small molecule per kilogram of the subject or sample weight (e.g., about 1 microgram per gram). • 10 kilograms to approximately 500 milligrams per kilogram, from approximately 100 micrograms per kilogram to approximately 5 milligrams per kilogram, or from approximately one microgram per kilogram to approximately 500 micrograms per kilogram kilogram). It should also be understood that the appropriate dose of a small molecule depends on the potency of the small molecule with respect to the • expression or activity that is going to be modulated. Said appropriate dose can be determined using the tests described in the present invention. When one or more of these small molecules are to be administered to an animal (eg, a human), or in order to modulate the expression or activity of a polypeptide or nucleic acid of the present invention, A doctor, veterinarian or researcher can, for example, prescribe a relatively low dose at the beginning, increasing the dose subsequently until a dose has been obtained. • appropriate response. In addition, it must be understood, 5 that the specific dose level for a particular animal will depend on a variety of factors including the activity of the specific compound employed, age, body weight, general health, gender, and diet of the subject, time of administration, ^ 10 administration route, the range of excretion, any combination of the drug, and the degree of expression or activity that is going to be modulated. The present invention can be incorporated in many different forms, and should not be interpreted as being limited to the modalities described herein; furthermore, these modalities are provided so that the present description is fully understood by those skilled in the art. The experts in the field, Many modifications and other embodiments of the present invention will be in mind after having obtained the benefits of the teachings presented in the foregoing description. Although some specific terms have been used, they are used as they are used in the matter unless otherwise specified LIST OF SEQUENCES < 110 > María A. Glucksmann < 120 > Receivers Couplings new Protein G < 130 > 5800-11-1 < 140 > • 10 < 141: < 160 > 7 fifteen < 170 > Patentln Software Ver. 2.0 • < 210 > 1 20 < 211 > 319 < 212 > PRT < 213 > Homo sapiens < 400 > 1 25 Met Ser Gln Gln Asn Thr Ser Gly Asp Cys Leu Phe Asp Gly Val Asn 10 15 Glu Leu Met Lys Thr Leu Gln Phe Ala Val His lie Pro Thr Phe Val 25 30 Leu Gly Leu Leu Leu Asn Leu Leu Ala lie His Gly Phe Ser Thr Phe 40 45 Leu Lys Asn Arg Trp Pro Asp Tyr Wing Wing Thr Ser lie Tyr Met lie 50 55 60 Asn Leu Ala Val Phe Asp Leu Leu Leu Val Leu Ser Leu Pro Phe Lys 65 70 75 8 80 Met Val Leu Ser Gln Val Gln Ser Pro Phe Pro Ser Leu Cys Thr Leu 85 90 95 Val Glu Cys Leu Tyr Phe Val Ser Met Tyr Gly Ser Val Phe Thr lie 100 105 110 Cys Phe lie Ser Met Asp Arg Phe Leu Ala lie Arg Tyr Pro Leu Leu 115 120 125 Val Ser His Leu Arg Ser Pro Arg Lys lie Phe Gly lie Cys Cys Thr 130 135 140 lie Trp Val Leu Val Trp Thr Gly Ser lie Pro lie Tyr Ser Phe His 145 150 155 160 Gly Lys Val Glu Lys Tyr Met Cys Phe His Asn Met Ser Asp Asp Thr 165 170 175 Trp Ser Ala Lys Val Phe Phe Pro Leu Glu Val Phe Gly Phe Leu Leu 180 185 190 Pro Met Gly lie Met Gly Phe Cys Cys Ser Arg Ser lie His lie Leu 195 200 205 Leu Gly Arg Arg Asp His Thr Gln Asp Trp Val Gln Gln Lys Ala Cys 210 215 220 lie Tyr Ser lie Ala Ala Ser Leu Ala Val Phe Val Val Ser Phe Leu Q 225 230 235 240 Pro Val His Leu Gly Phe Phe Leu Gln Phe Leu Val Arg Asn Ser Phe 245 250 255 Lie Val Glu Cys Arg Ala Lys Gln Ser lie Be Phe Phe Leu Gln Leu 260 265 270 Ser Met Cys Phe Ser Asn Val Asn Cys Cys Leu Asp Val Phe Cys Tyr QP 10 275 280 285 Tyr Phe Val lie Lys Glu Phe Arg Met Asn lie Arg Ala His Arg Pro 290 295 300 Ser Arg Val Gln Leu Val Leu Gln Asp Thr Thr lie Ser Arg Gly 305 310 315 15 < 210 > 2 • < 211 > 1617 < 212 > DNA < 213 > Homo sapiens < 400 > 2 aggtacagcc tttggccatt agagaactaa ggcaggaacc tccaacctga ccttgctctt 60 gtggactgca gttgtgattc aatgggcatg aattgctgtg tgatgctggg aaggtgtttg 120 tgattcttga caaagtcatt tgaatccatc acttcaagag agtgaaagga gccccgtctg 180 atctgttggt gttgtaggaa gaaacatgag tcagcaaaac accagtgggg actgcctgtt 240 tgacggtgtc aacgagctga tgaaaaccct acagtttgca gtccacatcc ccaccttcgt 300 cctgggcctg ctcctcaacc tgctggccat ccatggcttt agcaccttcc ttaagaacag 360 • gtggcccgat tatgctgcca cctccatcta catgatcaac ctggcagtct ttgacctgct 420 5 gctggtgctc tccctcccat tcaagatggt cctgtcccag gtacagtccc ccttcccgtc 480 cctgtgcacc ctggtggagt gcctttactt cgtcagcatg tacggaagcg tcttcaccat 540 ctgcttcatc agcatggacc ggttcttggc catccgttac ccgctactgg tgagccacct 600 ccggtccccc aggaagatct ttgggatctg ctgcaccatc tgggtcctgg tgtggaccgg 660 aagcatccct atctacagtt tccatgggaa agtggaaaaa tacatgtgct tccacaacat 720 QP 10 gtctgatgat acctggagcg ccaaggtctt cttcccgctg gaggtgtttg gcttcctcct 780 tcccatgggc atcatgggct tctgctgctc caggagcatc cacatcctgc tgggccgccg 840 agaccacacc caggactggg tgcagcagaa agcctgcatc tacagcatcg cagccagcct 900 ggctgtcttc gtggtctcct tcctcccagt ccacctgggg ttcttcctgc agttcctggt 960 gagaaacagc tttatcgtag agtgcagagc caagcagagc atcagcttct tcttgcaatt 1020 gtccatgtgt ttctccaacg tcaactgctg cctggatgtt ttctgctact actttgtcat 1080 caaagaattc cgcatgaaca tcagggccca ccggccttcc agggtccagc tggtcctgca 1140 ^ ~ ggacaccacg atctcccggg gctaacggaa ggacatcctg ttcaggggaa gaaagccctg 1200 gccctgaatt ctggtaacgg atttcgcgtt ccagggtttt gatgtggtgg gatgatccgc 1260 accatcttca ctgatgtgct tccctttgat gcccattgag tgccagcttt gctcattata 1320 cttttttcca ccccaaagac ctgcccagac agcttatacc acccagtgtt cagggatctc 1380 tgaagaaccc acagaccagg tgaattactg atttctaagt ccaaaaacta tagagcagaa 1440 gaattgagaa agagaatgag accatgtcaa caaggctgtt tccaactctc cccattttcc 1500 tgttcgactg ggaggttctg gagagagaga gaaagaaaga aaagaggtaa aggagggagc 1560 caagagagtc agttattggg gagagtgtct tgggcagagg tggggtggta gggatga 1617 < 210 > 3 < 211 > 337 < 212 > PRT • < 213 > Homo sapiens < 400 > 3 Met Asp Glu Thr Gly Asn Leu Thr Val Ser Ser Ala Thr Cys His Asp 1 5 10 15 QP 10 Thr He Asp Asp Phe Arg Asn Gln Val Tyr Ser Thr Leu Tyr Ser Met 20 25 30 He Ser Val Val Gly Phe Phe Gly Asn Gly Phe Val Leu Tyr Val Leu 35 40 45 He Lys Thr Tyr His Lys Lys Ser Ala Phe Gln Val Tyr Met He Asn 15 50 55 60 Leu Ala Val Ala Asp Leu Leu Cys Val Cys Thr Leu Pro Leu Arg Val 65 70 75 80 Val Tyr Tyr Val His Lys Gly He Trp Leu Phe Gly Asp Phe Leu Cys 85 90 95 Arg Leu Ser Thr Tyr Wing Leu Tyr Val Asn Leu Tyr Cys Ser He Phe 100 105 110 Phe Met Thr Wing Met Ser Phe Phe Arg Cys He Wing He Val Phe Pro 115 120 125 Val Gln Asn He Asn Leu Val Thr Gln Lys Lys Wing Arg Phe Val Cys 25 130 135 140 Val Gly He Trp He Phe Val He Leu Thr Ser Ser Pro Phe Leu Met 145 150 155 160 Wing Lys Pro Gln Lys Asp Glu Lys Asn Asn Thr Lys Cys Phe Glu Pro Pro Gln Asp Asn Gln Thr Lys Asn His Val Leu Val Leu His Tyr Val 180 185 190 Ser Leu Val Gly Gly Phe He He Pro Phe Val He He He Val Cys 195 200 205 P 10 Tyr Thr Met He He Leu Thr Leu Leu Lys Lys Ser Met Lys Lys Asn 210 215 220 Leu Ser Ser His Lys Lys Wing He Gly Met He Met Val Val Thr Ala 225 230 235 240 Wing Phe Leu Val Ser Phe Met Pro Tyr His He Gln Arg Thr He His 245 250 255 Leu His Phe Leu His Asn Glu Thr Lys Pro Cys Asp Ser Val Leu Arg 260 265 270 Met Gln Lys Ser Val Val Th Th Leu Ser Leu Ala Wing Ser Asn Cys 275 280 285 20 Cys Phe Asp Pro Leu Leu Tyr Phe Phe Ser Gly Gly Asn Phe Arg Lys 290 295 300 Arg Leu Ser Thr Phe Arg Lys His Ser Leu Ser Ser Val Thr Tyr Val 305 310 315 320 Pro Arg Lys Lys Wing Ser Leu Pro Glu Lys Gly Glu Glu He Cys Lys 25 325 330 335 Val • < 210 > 4 < 211 > 135Í < 212 > DNA < 213 > Homo sapiens < 400 > 4 • 10 actttcaggc cagaattcgg cacgaggctg gtagatcgaa tttactgaag acttggagct 60 tgcttctgag aacaaacgca aaaggacagt aaactgtgga ccttgaagtt agcagcgtgg 120 atattacacc gcttcctcta ttgatcacca gtaaaaggca catttgtgaa taagaaggaa 180 ggtactccag tgccagaaag aggcacaaag cagacattcg tagagaaaca tggatgaaac 240 aggaaatctg acagtatctt ctgccacatg ccatgacact attgatgact tccgcaatca 300 agtgtattcc accttgtact ctatgatctc tgttgtaggc ttctttggca atggctttgt 360 ctcataaaaa gctctatgtc cctatcacaa gaagtcagcc ttccaagtat acatgattaa 420 tttagcagta gcagatctac tttgtgtgtg cacactgcct ctccgtgtgg tctattatgt 480 tcacaaaggc atttggctct ttggtgactt cttgtgccgc ctcagcacct atgctttgta 540 tgtcaacctc tattgtagca tcttctttat gacagccatg agctttttcc ggtgcattgc 600 aattgttttt ccagtccaga acattaattt ggttacacag aaaaaagcca ggtttgtgtg 660 tgtaggtatt tggatttttg tgattttgac cagttctcca tttctaatgg ccaaaccaca 720 aaaaataata aaaagatgag ccaagtgctt tgagccccca caagacaatc aaactaaaaa 780 tcatgttttg gtcttgcatt atgtgtcatt ggttgttggc tttatcatcc cttttgttat 840 tgttacacaa tataattgtc gaccttacta tgatcatttt tgaaaaaaaa aaaaaatcaa 900 tctgtcaagt cataaaaagg ctataggaat gatcatggtc gtgaccgctg cctttttagt 960 cagtttcatg ccatatcata ttcaacgtac cattcacctt cattttttac acaatgaaac 1020 taaaccctgt gattctgtcc ttagaatgca gaagtccgtg gtcataacct tgtctctggc 1080 tgcatccaat tgttgctttg accctctcct atatttcttt tctgggggta actttaggaa 1140 aaggctgtct acatttagaa agcattcttt gtccagcgtg acttatgtac ccagaaagaa 1200 ggcctctttg ccagaaaaag gagaagaaat atgtaaagta tagtttaaac ccatttccag 1260 tccaaaccaa tgaaaatagt ttcccaaata agtattttgt caaatcattt acaaaaaaaa 1320 taaaaatttt acttaaaaaa aaaaaaaaaa aaggaaaa 1358 < 210 > 5 < 211 > 372 < 212 > PRT < 213 > Homo sapiens < 400 > 5 Met Leu Ala Asn Being Ser Thr Asn Being Ser Val Leu Pro Cys Pro 1 5 10 15 Asp Tyr Arg Pro Thr His Arg Leu His Leu Val Val Tyr Ser Leu Val 20 25 30 Leu Ala Ala Gly Leu Pro Leu Asn Ala Leu Ala Leu Trp Val Phe Leu 35 40 45 Arg Ala Leu Arg Val His Ser Val Val Ser Val Tyr Met Cys Asn Leu 50 55 60 Ala Ala Ser Asp Leu Leu Phe Thr Leu Ser Leu Pro Val Arg Leu Ser 65 70 75 80 Tyr Tyr Ala Leu His His Trp Pro Phe Pro Asp Leu Leu Cys Gln Thr 85 90 95 Thr Gly Wing He Phe Gln Met Asn Met Tyr Gly Ser Cys He Phe Leu 100 105 110 Met Leu He Asn Val Asp Arg Tyr Wing Gly He Val His Pro Leu Arg 115 120 125 Leu Arg His Leu Arg Arg Ala Arg Val Ala Arg Leu Leu Cys Leu Gly 130 135 140 Val Trp Ala Leu He Leu Val Phe Ala Val Pro Ala Ala Arg Val His 145 150 155 160 Arg Pro Be Arg Cys Arg Tyr Arg Asp Leu Glu Val Arg Leu Cys Phe 165 170 175 Glu Ser Phe Ser Asp Glu Leu Trp Lys Gly Arg Leu Leu Pro Leu Val 180 185 190 Leu Leu Ala Glu Ala Leu Gly Phe Leu Leu Pro Leu Ala Ala Val Val 195 200 205 Tyr Ser Ser Gly Arg Val Phe Trp Thr Leu Wing Arg Pro Asp Wing Thr 210 215 220 Gln Ser Gln Arg Arg Arg Lys Thr Val Arg Leu Leu Leu Wing Asn Leu 225 230 235 240 Val He Phe Leu Leu Cys Phe Val Pro Tyr Asn Ser Thr Leu Ala Val 245 250 255 Tyr Gly Leu Leu Arg Ser Lys Leu Val Wing Ala Ser Val Pro Ala Arg 260 265 270 Asp Arg Val Arg Gly Val Leu Met Val Met Val Leu Leu Wing Gly Wing 275 280 285 Asn Cys Val Leu Asp Pro Leu Val Tyr Tyr Phe Ser Wing Glu Gly Phe 290 295 300 Arg Asn Thr Leu Arg Gly Leu Gly Thr Pro His Arg Wing Arg Thr Ser 305 310 315 320 Wing Thr Asn Gly Thr Arg Wing Wing Leu Wing Gln Ser Glu Arg Wing 325 330 335 Val Thr Thr Asp Wing Thr Arg Pro Asp Wing Wing Gln Gly Leu Leu 340 345 350 Arg Pro Ser Asp Ser His Ser Leu Ser Ser Phe Thr Gln Cys Pro Gln 355 360 365 Asp Ser Ala Leu 370 < 210 > 6 < 211 > 2559 < 212 > DNA < 213 > Homo sapiens < 400 > 6 ccatgacctc cctctgcttg ttttgggacc atgtctgtac agcctctagg ccccagcccc 60 ggaggtgaat gccatgccat gattctggtg tgctccatgg catccccagc ctagctccca 120 atcccacttt ggcacgatgt tagccaacag ctcctcaacc aacagttctg ttctcccgtg 180 tcctgactac cgacctaccc accgcctgca cttggtggtc tacagcttgg tgctggctgc 240 cgggctcccc ctcaacgcgc tagccctctg ggtcttcctg cgcgcgctgc gcgtgcactc 300 ggtggtgagc gtgtacatgt gtaacctggc ggccagcgac ctgctcttca ccctctcgct 360 • gcccgttcgt ctctcctact acgcactgca ccactggccc ttccccgacc tcctgtgcca 420 5 gacgacgggc gccatcttcc agatgaacat gtacggcagc tgcatcttcc tgatgctcat 480 caacgtggac cgctacgccg gcatcgtgca cccgctgcga ctgcgccacc tgcggcgggc 540 ccgcgtggcg cggctgctct gcctgggcgt gtgggcgctc atcctggtgt ttgccgtgcc 600 cgccgcccgc gtgcacaggc cctcgcgttg ccgctaccgg gacctcgagg tgcgcctatg 660 cttcgagagc ttcagcgacg agctgtggaa aggcaggctg ctgcccctcg tgctgctggc 720 ^ 10 cgaggcgctg ggcttcctgc tgcccctggc ggcggtggtc tactcgtcgg gccgagtctt 780 ctggacgctg gcgcgccccg acgccacgca gagccagcgg cggcggaaga ccgtgcgcct 840 cctgctggct aacctcgtca tcttcctgct gtgcttcgtg ccctacaaca gcacgctggc 900 ggtctacggg ctgctgcgga gcaagctggt ggcggccagc gtgcctgccc gcgatcgcgt 960 gcgcggggtg ctgatggtga tggtgctgct ggccggcgcc aactgcgtgc tggacccgct 1020 ggtgtactac tttagcgccg agggcttccg caacaccctg cgcggcctgg gcactccgca 1080 ccgggccagg acctcggcca ccaacgggac gcgggcggcg ctcgcgcaat ccgaaaggtc 1140 accgacgcca cgccgtcacc ccaggccgga tgccgccagt caggggctgc tccgaccctc 1200 cgactcccac tctctgtctt ccttcacaca gtgtccccag gattccgccc tctgaacaca 1260 catgccattg cgctgtccgt gcccgactcc caacgcctct cgttctggga ggcttacagg 1320 gtgtacacac aagaaggtgg gctgggcact tggacctttg ggtggcaatt ccagcttagc 1380 aacgcagaag agtacaaagt gtggaagcca gggcccaggg aaggcagtgc tgctggaaat 1440 ggcttcttta aactgtgagc acgcagagca ccccttctcc agcggtggga agtgatgcag 1500 aaagcccacc cgtgcagagg gcagaagagg acgaaatgcc tttgggtggg cagggcatta 1560 aactgctaaa agctggttag atggaccaga aaatgggcat tctggatttt aacccgccac 1620 gagttgaaga aggggcttga gcaccaggtt tggtggacaa agctactgag atgcctgttc 1680 atctgctgac ttctgtctag gctcatggat gccaccccct ttcattttgg cctaggcttc 1740 ccctgctcac cactgaggcc taatacaaga gttcctatgg acagaactac attctttctc 1800 gcatagtgac ttgtgacaat ttagacttgg catccagcat gggatagttg gggcaaggca 1860 aaactaactt agagtttccc cctcaacaac atccaagtcc aaaccctttt taggttatcc 1920 tttcttccat cacatcccct tttccaggcc tcctccattt taggtcctta atattctttc 1980 tttttctctc tctctcgttt ctctcttctc tctcctctcc tctctcttct cctcttctct 2040 ctctctccct ctctctcctt gtccagagta aggataaatt ctttctacta aagcactggt 2100 tctcaaactt tttggtctca gaccccactc ttagaaattg aggatctcaa agagctttgc 2160 ttatattttg ttcttttgat ctagaaatta acttaccata atttttaaaa aagcgaatac 2220 tgcacacatt taaatacaca acattagcca tgggagcaat aatgtcacca cacacacttc 2280 atgaagcctc tggaaaactc tacagtatac atgagagtga ttgtgagaga aagggacaaa 2340 taacatctgt gtagcagtat tatgaaaata gcttgacctc gtggacttcc tcagagggtt 2400 ggtccctgga tcacactttg agaaccatac ttgtcctgaa gtattggagt tcatgtctaa 2460 cttcttccca gggcattatg tacagtgctt tttattactg tggggagagg gcagtgctaa 2520 ataaattaat cactactgat agtcaaaaaa aaaaaaaaa 2559 < 210 > 7 < 211 > 269 < 212 > PRT < 213 > Unknown < 220 > < 223 > Description of Unknown Organism: Rhodopsin family Transmemran receiver < 400 > 7 Gly Asn He Leu Val He Trp Val He Cys Arg Tyr Arg Arg Met Arg 1 5 10 15 Thr Pro Met Asn Tyr Phe He Val Asn Leu Wing Val Wing Asp Leu Leu 20 25 30 5 Phe Ser Leu Phe Thr Met Pro Phe Trp Met Val Tyr Tyr Val Met Gly 35 40 45 Gly Arg Trp Pro Phe Gly Asp Phe Met Cys Arg He Trp Met Tyr Phe 50 55 60 Asp Tyr Met Asn Met Tyr Wing Ser He Phe Phe Leu Thr Cys He Ser f 10 65 70 75 80 He Asp Arg Tyr Leu Trp Wing He Cys His Pro Met Arg Tyr Met Arg 85 90 95 Trp Met Thr Pro Arg His Arg Wing Trp Val Met He He He He He Trp 100 105 110 15 Val Met Ser Phe Leu He Ser Met Pro Pro Phe Leu Met Phe Arg Trp 115 120 125 Ser Thr Tyr Arg Asp Glu Asn Glu Trp Asn Met Thr Trp Cys Met He 130 135 140 Tyr Asp Trp Pro Glu Trp Met Trp Arg Trp Tyr Val He Leu Met Thr 20 145 150 155 160 He He Met Met Gly Phe Tyr He Pro Met He Met Met Leu Phe Cys Tyr 165 170 175 Trp Arg He Tyr Arg He Wing Arg Leu Trp Met Arg Met He Pro Ser 180 185 190 25 Trp Gln Arg Arg Arg Arg Met Ser Met Arg Arg Glu Arg Arg He Val 195 200 205 Lys Met Leu He He Met Met Val Val Phe He He Cys Trp Leu Pro 210 215 220 Tyr Phe He Val Met Phe Met Asp Thr Leu Met Met Trp Trp Phe Cys P 225 230 235 240 Glu Phe Cys He Trp Arg Arg Leu Trp Met Tyr He Phe Glu Trp Leu 245 250 255 Wing Tyr Val Asn Cys Pro Cys He Asn Pro He He Tyr 260 265

Claims (43)

  1. NOVELTY OF THE INVENTION Having described the present invention, it is considered as a novelty and, therefore, the content of the following is claimed as property: CLAIMS 1. - An isolated polypeptide having an amino acid sequence selected from the group consisting of: (a) The amino acid sequence shown in SEQ ID NO: 1, the amino acid sequence shown in SEQ ID NO: 3, and the amino acid sequence shown in SEQ ID NO: 5; (b) The amino acid sequence of an allelic variant of the amino acid sequence shown in SEQ ID NO: 1, the amino acid sequence of the allelic variant of the amino acid sequence shown in SEQ ID NO: 3, and the sequence of amino acids of an allelic variant of the amino acid sequence shown in SEQ ID NO: 5; (c) The amino acid sequence of a variant of the amino acid sequence shown in SEQ ID NO: 1, wherein the variant of the sequence is encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule shown in FIG. SEQ ID NO: 2 under severe conditions, the sequence of ^ P amino acids of a sequence variant sequence 5 amino acids shown in SEQ ID NO: 3, wherein the variant of the sequence is encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule shown in SEQ ID NO: 4 under severe conditions, and the sequence of amino acids ^^ 10 a variant sequence of the amino acid sequence shown in SEQ ID NO: 5, wherein the variant of the sequence is encoded by a nucleic acid molecule that hybridizes to the nucleic acid molecule shown in SEQ ID NO: 6 under severe conditions; (D) A fragment of an amino acid sequence shown in SEQ ID NO: 1, wherein the fragment k comprises at least 8 contiguous amino acids 1 to 264, a fragment of the amino acid sequence shown in SEQ ID NO. : 3, where the fragment 20 comprises at least 9 contiguous amino acids from 1 to 244, and a fragment of an amino acid sequence shown in SEQ ID NO: 5, wherein the fragment comprises at least 8 contiguous amino acids; (e) The amino acid sequence of the polypeptide The mature receptor of about amino acid 6 to about amino acid 319, shown in SEQ ID NO: 1, the amino acid sequence of the mature receptor polypeptide of about amino acid 6 to about amino acid 337, shown in SEQ 5 ID NO: 3, and the amino acid sequence of the receptor polypeptide from about amino acid 6 to about amino acid 372, shown in SEQ ID NO: 5; (f) The amino acid sequence of the polypeptide • 10 shown in SEQ ID NO: 1 from about amino acid 1 to about amino acid 24, the amino acid sequence of the polypeptide shown in SEQ ID NO: 3 from about amino acid 1 to about amino acid 28, and the sequence of 15 amino acids of the polypeptide shown in SEQ ID NO: 5 from about amino acid 1 to about amino acid 25; • (g) The amino acid sequence of an epitope in the region where any of the 20 polypeptides from (a) to (f). 2. - An isolated antibody that selectively binds to a polypeptide (a) to (g), according to claim 1. 3. - The isolated nucleic acid molecule having a nucleotide sequence selected from the group consisting of: • (a) The nucleotide sequence shown in SEQ ID NO: 2, the nucleotide sequence shown in SEQ ID NO: 4 and the nucleotide sequence shown in SEQ ID NO: 6; (b) A nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO: 1, • a nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO: 3, and the nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO: 5; (c) A nucleotide sequence complementary to Any of the nucleotide sequences in (a) or (b). 4. - An isolated nucleic acid molecule having a nucleotide sequence selected from the group consisting of: (a) A nucleotide sequence encoding an amino acid sequence of a variant sequence of the amino acid sequence shown in SEQ ID NO: 1 that hybridizes to the nucleotide sequence shown in SEQ ID NO: 2 under severe conditions, a nucleotide sequence encoding an amino acid sequence of a variant sequence of the • amino acid sequence shown in SEQ ID NO: 3 5 that hybridizes to the nucleotide sequence shown in SEQ ID NO: 4 under severe conditions, and a nucleotide sequence that encodes an amino acid sequence of a sequence variant sequence of amino acids shown in SEQ ID NO: 5 which hybridizes to the nucleotide sequence shown in SEQ ID NO: 6 under severe conditions; (b) A nucleotide sequence complementary to the nucleotide sequence at point (a). 5. A nucleic acid molecule isolated from a polynucleotide having a nucleotide sequence selected from the group consisting of: (a) A nucleotide sequence that encodes a fragment of the amino acid sequence shown in SEQ ID NO: 1, wherein the fragment comprises at least 8 contiguous amino acids 1 to 264, a nucleotide sequence encoding a fragment of the amino acid sequence shown in SEQ ID NO: 3 wherein the fragment comprises at least 9 25 contiguous amino acids from 1 to 244, and a nucleotide sequence encoding a fragment of a sequence of the amino acid sequence shown in SEQ ID NO: 5, wherein the fragment comprises at least 8 • contiguous amino acids; (b) A nucleotide sequence complementary to the nucleotide sequence of point (a). 6. - A nucleic acid vector comprising the nucleic acid sequences according to • any one of claims 3 to 5. 7. - A host cell containing the vector according to claim 6. 8. A method for producing any of the polypeptides according to claim 1 comprising the introduction of a nucleotide sequence encoding any of the polypeptide sequences of points (a) to (f) within the 20 host cell, and culturing the host cell under conditions in which the proteins are expressed from the nucleic acid. 9. - A method for detecting the presence of any of the polypeptides according to claim 1 in a sample, said method comprising contacting said sample with an agent that specifically allows the detection of the • presence of the polypeptide in the sample and then detect the presence of the polypeptide. 10. - The method according to claim 9, wherein said agent has the ability of selective physical association with said • 10 polypeptide. 11. - The method according to claim 10, wherein said agent binds to said polypeptide. 12. The method according to claim 11, wherein said agent is an antibody. 13. The method according to claim 11, wherein said agent is a binder. 14. - A kit comprising reagents used for the method according to claim 9, wherein the reagents comprise an agent that binds specifically to said polypeptide. 15. A method for detecting the presence of any of the nucleic acid sequences according to any of claims 3 to 5 in a sample, the method comprising contacting the sample with an oligonucleotide that hybridizes to the sequences of nucleic acid under severe conditions and determines whether the oligonucleotide binds to the nucleic acid sequence in the sample. 16. - The method according to claim 15, wherein the nucleic acid, whose presence is detected, is mRNA. . { ^ k 17.- A device comprising reagents used for the method in accordance with the claim 15, wherein the reactants comprise a compound that 20 hybridizes under severe conditions to any of the nucleic acid molecules. 18. - A method for the identification of an agent that interacts with any of the polypeptides of According to claim 1, said method comprising contacting said anchor with a cell capable of allowing an interaction between said polypeptide and said agent so that • said polypeptide can interact with said agent and measure the interaction. 19. - A method of selecting a cell to identify an agent that interacts with any of the polypeptides according to the claim 10 1, said method comprising contacting said agent with a cell capable of allowing interaction between said polypeptide and said agent so that said polypeptide can interact with said agent and measure the interaction. 20. A method for the identification of an agent that modulates the level or activity of any of the polypeptides according to claim 1 in a cell, said method comprising putting in The aforementioned contact with a cell capable of expressing said polypeptide so that the level or activity of said polypeptide can be modulated in said cell by said agent and measuring the level or activity of said polypeptide. 25 21. - A method of selecting a cell to identify an agent that modulates the level or activity of any of the polypeptides according to claim 1 in a cell, said method comprising contacting said anchor with a cell capable of expressing said polypeptide so that the level or activity of said polypeptide can be modulated in said cell, by said agent and measuring the level or activity of said polypeptide. 22. - The method according to any of claims 19 or 20 wherein said interaction is the link. 23. The method according to claim 22, wherein a fragment of the polypeptide is contacted. 24. - A method for modulating the activity of any of the polypeptides according to claim 1, said method comprising contacting any of the polypeptides according to claim 1 with an agent under conditions that allow the polypeptide to modulate the activity of the polypeptide . 25. - A method to identify an agent that interacts with any of the acid molecules • nucleic of claims 3 to 5, said method comprising contacting said agent with a cell capable of allowing an interaction between said nucleic acid molecule and said agent so that said nucleic acid molecule can interact with said agent and measure the interaction. # 10 26.- A method of selecting a cell for identifying an agent that interacts with any of the nucleic acid molecules according to claims 3 to 5, said method comprising 15 contacting said agent with a cell capable of allowing an interaction between said nucleic acid molecule and said agent so that said nucleic acid molecule can interact with said agent and measure the interaction. 27.- A method for identifying an agent that modulates the level or activity of any of the nucleic acid molecules according to claims 3 to 5 in a cell, said method comprising contacting said agent with a cell with capacity. to express said nucleic acid molecule so that the level or activity of said nucleic acid molecule can be modulated in • said cell by said aid and measuring the activity or level of said nucleic acid molecule. 28. - A method of selecting a cell to identify a substance that modulates the level or activity of any of the nucleic acid molecules of According to claims 3 to 5 in a cell, said method comprising contacting said anchor with a cell capable of expressing said nucleic acid molecule, so that the level or activity of said acid molecule The nucleic acid can be modulated in said cell, by said agent and measure the level or activity of said nucleic acid molecule. • 29. - The method according to any of claims 25 or 26 wherein said interaction is binding. 30. - A method to modulate the activity of any of the nucleic acid molecules of According to claims 3 to 5, said method comprising contacting any of the nucleic acid molecules according to claim 1, with an • under conditions that allow the patient to modulate the activity of the nucleic acid molecule. 31. - A pharmaceutical composition containing any of the polypeptides according to claim 1 in a pharmaceutically acceptable vehicle. 32. - A pharmaceutical composition containing any of the nucleic acid molecules according to claims 3 to 5 in a pharmaceutically acceptable carrier. 33. - A non-human transgenic animal wherein one or more of the cells of said animal contains any of the nucleic acid sequences according to claims 3 to 5. 34. - A non-human transgenic animal in which one or more of the cells of said animal contains any of the nucleic acid sequences of According to claims 3 to 5 wherein said cell expresses any of the polypeptides according to claim 1. • A method for producing a transgenic animal according to claims 32 and 33, said method comprising introducing any of the nucleic acid sequences according to claim 1 into a cell, wherein said cell is present in said animal or gives origin to said animal. 36. - An identified agent using any of the methods according to claims 19 to 22 or 25 to 28. 15 - A method for the treatment of a condition in a subject comprising the administration of pharmaceutical compositions in accordance with any of the claims 31 20 or 32. 38. - A method for the treatment of a condition in a subject by modulating the level or expression of any of the polypeptides of 25 according to claim 1 in said subject. 39. - A method for the treatment of a condition in a subject by modulating the level or activity of any of the nucleic acid molecules according to claims 3 to 5 in said subject. 40. - The method according to any of claims 9 or 15 wherein said detection is in a cell derived from a subject having a condition comprising said cell. 41. - The method according to any of claims 24 or 30 wherein said modulation is in a subject having a condition comprising said cell. 42. - The method according to claim 39 wherein said condition is anemia, neutropenia or thrombocytopenia. 43. - The method according to claim 41 wherein said condition is anemia, neutropenia or thrombocytopenia.
MXPA/A/2001/003808A 1998-10-16 2001-04-16 G-protein coupled receptors MXPA01003808A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US09/173,869 1998-10-16

Publications (1)

Publication Number Publication Date
MXPA01003808A true MXPA01003808A (en) 2002-06-05

Family

ID=

Similar Documents

Publication Publication Date Title
US20030040052A1 (en) Novel G-protein coupled receptors
WO2000023588A9 (en) G-protein coupled receptors
US20030166042A1 (en) Novel seven-transmembrane proteins/G-protein coupled receptors
EP1246916A2 (en) 26904, 38911, and 39404, seven-transmembrane proteins / g-protein coupled receptors
US20020076784A1 (en) 40322, a novel human dynamin
US20010044130A1 (en) 39406 protein, a novel seven transmembrane protein
US20020068710A1 (en) 20685, 579, 17114, 23821, 33894 and 32613, novel human transporters
US20060205053A1 (en) 27411, a novel human PGP synthase
WO2000011170A1 (en) 14400 receptor, a g-protein coupled receptor
MXPA01003808A (en) G-protein coupled receptors
US20020042058A1 (en) 18057 protein, a novel seven transmembrane protein
US20010036649A1 (en) 26934, a novel cytidine deaminase-like molecule and uses thereof
US20030162247A1 (en) 32164 protein, a novel seven transmembrane protein
WO2001044453A1 (en) 25678, a novel human adenylate cyclase
US20080051322A1 (en) 14400, 2838, 14618, 15334, 14274, 32164, 39404, 38911, 26904, 31237, 18057, 16405, 32705, 23224, 27423, 32700, 32712 and 12216, novel seven-transmembrane proteins/G-protein coupled receptors
EP1135492A1 (en) 12216 receptor, a g-protein coupled receptor
WO2001032885A9 (en) Human lipase
US20030077626A1 (en) 20685, 579, 17114, 23821, 33894 and 32613, novel human transporters
AU1211100A (en) G-protein coupled receptors
WO2001059116A9 (en) 19459 protein, a human g-protein coupled seven transmembrane receptor