CXCR3 is a gliadin receptor

Abstract
Gliadin is identified as a physiological receptor for CXCR3. Assays for determining modulators of CXCR3 signaling are provided. Fragments of gliadin which function as inhibitors of CXCR3 signaling can be determined. Methods for treating diseases relating to gluten and/or autoimmunity by targeting CXCR3 are provided. Such diseases include celiac disease, gluten sensitivity, gluten allergy, rheumatoid arthritis, multiple sclerosis, immune-mediated or type 1 diabetes mellitus, inflammatory bowel diseases, systemic lupus erythematosus, psoriasis, scleroderma, and autoimmune thyroid diseases.
Description
TECHNICAL FIELD OF THE INVENTION

This invention is related to the area of autoimmune diseases. In particular, it relates to treatment and drug screening and discovery for autoimmune diseases.


BACKGROUND OF THE INVENTION

Environmental stimuli, such as microorganisms and gluten, induce zonulin release in the intestine, brain, heart, and other organs. Zonulin release causes an increase in permeability of epithelia as measured by a decrease in trans-epithelial electrical resistance (TEER) (ex vivo) or the Lactulose/mannitol test (in vivo). Presumably, the environmental stimuli interact with the surface of cells, possibly by binding to a receptor on the cell surface. However, such a receptor has not been identified.


Many inflammatory diseases are thought to be autoimmune. These include rheumatoid arthritis, multiple sclerosis, immune-mediated or type 1 diabetes mellitus, inflammatory bowel diseases, systemic lupus erythematosus, psoriasis, scleroderma, and autoimmune thyroid diseases. Prolonged inflammation is often associated with these diseases, although the inflammation is thought to be a sequela rather than a primary pathological insult.


CXCR3 is a G protein-coupled receptor which is known to bind to three chemokines, IP10 (interferon-γ-inducible 10 kDa protein), MIG (monokine induced by interferon-γ) and I-TAC (interferon-inducible T cell α-chemoattractant). IP10, MIG and I-TAC are termed CXC chemokines, because they contain a CXC sequence motif. CXCR3 has been linked to integrin activation, cytoskeletal changes, and chemotaxis. CXCR3 is prominently expressed in inflamed tissues.


There is a continuing need in the art for methods to treat autoimmune diseases more effectively and to discover or identify drugs which are suitable for treating autoimmune diseases.


SUMMARY OF THE INVENTION

One embodiment of the invention provides a method to screen for modulators of CXCR3 signaling. Gliadin or a fragment of gliadin (e.g. a fragment comprising at least six amino acid residues) is contacted with CXCR3. Binding of the gliadin or fragment of gliadin to CXCR3 is determined. A fragment of gliadin which binds to CXCR3 is identified as a modulator of CXCR3 signaling.


Another embodiment of the invention provides a method to screen for modulators of CXCR3 signaling. Gliadin or a fragment of gliadin comprising at least six amino acid residues is contacted with a first cell which expresses CXCR3 and with a second cell which does not express CXCR3. Binding of the gliadin or fragment to the first and second cells is determined. A fragment of gliadin which binds preferentially to the first cell relative to the second cell is identified as a modulator of CXCR3 signaling.


Still another embodiment of the invention provides a method to screen for modulators of CXCR3 signaling. Gliadin or fragment of gliadin comprising at least six amino acid residues is contacted with CXCR3 or another CXCR3 ligand, such as IP10, MIG, or ITAC. Inhibition of binding of ligand to CXCR3 caused by the fragment of gliadin is determined. A fragment of gliadin which inhibits binding of ligand to CXCR3 is identified as a modulator of CXCR3 signaling.


Yet another aspect of the invention is a method to screen for modulators of CXCR3 signaling. A fragment of gliadin comprising at least six amino acid residues is contacted with a cell which expresses CXCR3 or another CXCR3 ligand, such as IP10, MIG, or ITAC. Binding of ligand to the cell is determined. A fragment of gliadin which inhibits binding of ligand to the cell is identified as a modulator of CXCR3 signaling.


Also provided by the present invention is a method to screen for modulators of zonulin release. A test compound is contacted with CXCR3. Binding of the test compound to CXCR3 is determined. A test compound which binds to CXCR3 is identified as a modulator of zonulin release.


Another embodiment provided by the present invention is a method to screen for modulators of zonulin release. A test compound is contacted with CXCR3. Binding of the gliadin to CXCR3 in the presence and absence of the test compound is determined. A test compound which inhibits binding of gliadin to CXCR3 is identified as a modulator of zonulin release.


Still another embodiment of the invention is a method to screen for modulators of zonulin release. A test compound is contacted with a first cell which expresses CXCR3 and with a second cell which does not express CXCR3. Binding of the test compound to the first and second cells is determined. A test compound which binds preferentially to the first cell relative to the second cell is identified as a modulator of zonulin release.


Even a further embodiment is a method to screen for modulators of zonulin release. A test compound is contacted with a cell which expresses CXCR3 . Binding of gliadin to the cell is determined. A test compound which inhibits binding of gliadin to the cell is identified as a modulator of zonulin release.


Another embodiment of the invention is a method of treating a patient with a disease selected from the group consisting of celiac disease, gluten allergy, gluten sensitivity, and gluten ataxia. An antibody which specifically binds to CXCR3 is administered to the patient. Zonulin release is thereby inhibited.


A further embodiment of the invention is a method of treating a patient with an autoimmune or inflammation-associated disease. Typically, these diseases will be characterized by an undesired CXCR3 signaling. The disease is selected from the group consisting of type 1 diabetes, celiac disease, autoimmune hepatitis, multiple sclerosis, autism, dermatitis herpetiformis, IgA nephropathy, primary biliary cirrhosis, rheumatoid arthritis, systemic lupus erythematosus, Grave's disease, Hashimoto's disease, and depression. An antibody which specifically binds to CXCR3 is administered to the patient. CXCR3 signaling is thereby inhibited.


In some embodiments, the present invention provides methods of identifying a CXCR3 ligand comprising contacting a cell expressing CXCR3 with gliadin or a fragment thereof and a compound to be tested and determining the amount of gliadin or fragment thereof bound to the cell. For example, one or more gliadins or fragments thereof may be labeled with one or more fluorescent moieties. A CXCR3-expressing cell may then be brought into contact with the fluorescently labeled gliadin or fragment in the presence of the compound to be tested. The binding of the gliadin or fragment thereof and CXCR3 may be determined using standard techniques. Suitable techniques include, but are not limited to, fluorescence activated cell sorting (FACS), fluorescent microscopy, and fluorescence spectrophotometry. Optionally the gliadin or fragment thereof may be contacted with CXCR3 -expressing cells in the absence of compound to be tested and the amount of binding of gliadin or fragment thereof to CXCR3-expressing cell may be determined. The amount of binding in the presence of compound to be tested and in the absence of compound to be tested may be compared. Other techniques known to those skilled in the art may be used to quantify the gliadin or fragment thereof binding. For example, cells expressing CXCR3 may be fixed to a solid surface, for example, a microtiter plate or a bead (e.g., a magnetic bead) and contacted with fluorescently labeled gliadin or fragment thereof. The amount of bound fluorescently labeled gliadin may be determined. In some embodiments, gliadin may be labeled with a detectable moiety other than a fluorescent moiety, for example, with biotin or digoxigenein and, detected with a suitable reagent, for example, streptavidin or anti-digoxigenin antibody.


In some embodiments, the present invention provides methods of identifying a CXCR3 ligand comprising contacting a purified CXCR3 or fragment thereof with gliadin or a fragment thereof and a compound to be tested and determining the amount of gliadin or fragment thereof bound to the CXCR3. For example, one or more gliadins or fragments thereof may be labeled with one or more fluorescent moieties. CXCR3 may then be brought into contact with the fluorescently labeled gliadin or fragment in the presence of the compound to be tested. The binding of the gliadin or fragment thereof and CXCR3 may be determined using standard techniques. Suitable techniques include, but are not limited to, ELISA and fluorescence spectrophotometry. Optionally the gliadin or fragment thereof may be contacted with CXCR3 in the absence of compound to be tested and the amount of binding of gliadin or fragment thereof to CXCR3 cell may be determined. The amount of binding in the presence of compound to be tested and in the absence of compound to be tested may be compared. Other techniques known to those skilled in the art may be used to quantify the gliadin or fragment thereof binding. For example, CXCR3 may be fixed to a solid surface, for example, a microtiter plate or a bead (e.g., a magnetic bead) and contacted with fluorescently labeled gliadin or fragment thereof. The amount of bound fluorescently labeled gliadin may be determined. In some embodiments, gliadin may be labeled with a detectable moiety other than a fluorescent moiety, for example, with biotin or digoxigenein and, detected with a suitable reagent, for example, streptavidin or anti-digoxigenin antibody. In some embodiments, gliadin or fragment thereof may be attached to a solid support and labeled CXCR3 or fragment thereof may be detected in the presence and absence of a compound to be tested. CXCR3 may be labeled and detected using any techniques known in the art, for example, using the techniques described above for labeling and detecting gliadin.


These and other embodiments which will be apparent to those of skill in the art upon reading the specification provide the art with methods of screening for useful therapeutic agents and with methods of treating autoimmune and inflammation-associated diseases.




BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A and 1B show results when jejunal intestinal fragments from wild-type mice mounted in microsnapwells were challenged with gliadin. FIG. 1A shows the amount of zonulin released after challenge. Gliadin challenge causes an increase in zonulin released. FIG. 1B shows the Trans Epithelial Electrical Resistance (TEER) after challenge. Gliadin challenge causes a decrease in TEER.



FIG. 2 shows the zonulin released after gliadin challenge of endoscopic jejunal biopsies from CXCR3-deficient mice. Gliadin challenge failed to lead to zonulin release. Compare to FIG. 1A.



FIG. 3 shows the TEER after gliadin challenge of endoscopic jejunal biopsies from CXCR3-deficient mice. Gliadin challenge failed to decrease TEER. Compare to FIG. 1B.



FIG. 4 shows the results of the protein bound to the gliadin affinity column is CXCR3, according to protein database search.



FIG. 5A shows the zonulin levels and TEER of B6 wild type mice challenged with gliadin and FIG. 5B shows the zonulin levels and TEER of CXCR3 knock-out mice (n=20) challenged with gliadin. The CXCR3 knock-out mice do not respond to gliadin and do not release zonulin, and accordingly, do not exhibit an increase in intestinal permeability. In contrast, the wild-type cohort responds to gliadin, releases zonulin and, therefore, does exhibit an increase in intestinal permeability.



FIG. 6 shows the results of CXCR3 transfected HEK293 cells probed with anti-CXCR3 mAb (red trace) and IgG1 isotype control (blue trace). FIG. 6A shows cells transfected with vector. FIG. 6B shows cells transfected with vector expressing CXCR3.



FIG. 7 shows the results of fluorescence microscopy of cells transfected probed with DAPI (blue), RITC-labeled anti-CXCR3 monoclonal antibody (red), and FITC-labeled PT-gliadin (green). Panel A shows cells transfected with control vector and probed with DAPI and anti-CXCR3 monoclonal antibody, Panel B shows cells transfected with CXCR3 -expressing vector and probed with anti-CXCR3 monoclonal antibody, and Panel C shows cells transfected with CXCR3-expressing vector and probed with anti-CXCR3 monoclonal antibody and FITC-labeled PT-gliadin.



FIG. 8 shows the results of the effect of gliadin digested with pepsin and trypsin (PT-Gliadin) on HLA-DR expression in dendritic cells from normal volunteers.



FIG. 9 shows the effect of PT-gliadin on TEER and zonulin release in Black 6 wild type mice small intestine. FIG. 9A shows the TEER measurements made in snapwells and FIG. 9B shows the zonulin concentrations.



FIG. 10 shows the effect of PT-gliadin on TEER and zonulin release on small intestine from CXCR3 knockout mice. FIG. 10A shows the TEER measurements made in snapwells and FIG. 10B shows the zonulin concentrations.




DETAILED DESCRIPTION OF THE INVENTION

The inventors have discovered that the receptor known as CXCR3 is a physiological receptor for gliadin. This receptor not only binds to gliadin, but it also signals the release of zonulin and a decrease in trans-epithelial electrical resistance (TEER). These downstream effects indicate that the binding to gliadin is physiological.


Screening for modulators of CXRC3 signaling can be accomplished by a variety of techniques. Binding to CXRC3 to test compounds can be directly measured, or inhibition of binding of gliadin or another ligand to the receptor can be measured. Other ligands which can be used include IP10, MIG, and ITAC. Ligands can be labeled to facilitate measurement of binding. Assays may be in cell-free systems or in cell-based systems. Any binding assay format can be used, including formats where the receptor is attached to a solid support, either directly or indirectly.


Test compounds which can be tested are any compounds. The compounds may be tested as single compounds or in combinations of compounds. The compounds may be structurally identified or of unknown structure. The compounds may be novel or previously known. The compounds may be natural products or synthetic.


According to one embodiment of the invention the test compounds are fragments of gliadin. Gliadin is a family of proteins which are produced by wheat and other grains. Examples of gliadins are gliadin alpha, gamma, and omega. Gliadins are the aqueous alcohol-soluble storage proteins in the seed. There is great heterogeneity even within a single class of gliadins. At least six, seven, eight, nine, ten, eleven, fifteen, twenty, thirty, thirty-five, fifty, or seventy-five amino acid residues may be used in fragments of gliadin as test compounds. Fragments include any molecule which is less than full length. Fragments may be, e.g., synthesized or the result of proteolytic degradation. The following tables provide the sequences of a representative number of gliadins.

TABLE 1Amino acid sequence of alpha-gliadin fromTriticum aestivum (NCBI accession no. CAB76964,(SEQ ID NO:1))1MVRVPVPQLQ PQNPSQQQPQ EQVPLVQQQQ FPGQQQPFPPQQPYPQPQPF PSQQPYLQLQ61PFPQPQLPYP QPQLPYPQPQ LPYPQPQPFR PQQPYPQSQPQYSQPQQPTS QQQQQQQQQQ121QQKQQQQQQQ QILQQILQQQ LIPCRDVVLQ QHSIAYGSSQVLQQSTYQLV QQLCCQQLWQ181IPEQSRCQAI HNVVHAIILH QQQQQQQQQQ QQPLSQVSFQQPQQQYPSGQ GSFQPSQQNP241QAQGSVQPQQ LPQFEEIRNL ALETLPAMCN VYIPPYCTIAPVGIFGTNYR










TABLE 2








Amino acid sequence of alpha-gliadin precursor



from Triticum turgidum subsp. durum (NCBI


accession no. CAI35909, (SEQ ID NO:2))

















1
MKTFLILALL AIVATTATTA VRVPVPQLQR QNPSQQQPQE




QVPLVQQQQF LGQQQPFPPQ





61
QPYPQPQPFP SQQPYLQLQP FPQPQLPYSQ PQPFRPQQPY



PQPQPRYSQP QQPISQQQQQ





121
QHQQHQQHHQ EQQILQQILQ QQLIPCMDVV LQQHNIAHRR



SQVLQQSTYQ LLQELCCQHL





181
WQIPEQSQCQ AIHNVVHAII PHQQQKQQQQ PSSQFSFQQP



LQQYPLGQGS FRPSQQNPQA





241
QGSVQPQQLP QFEEIRNLAL QTLPAMCNVY IPPYCTIAPF



GIFGTN

















TABLE 3








Amino acid sequence of alpha/beta-gliadin



precursor from Triticum aestivum (NCBI


accession no. AAA34280, (SEQ ID NO:3))

















1
MKTFLILVLL AIVATTATTA VRFPVPQLQP QNPSQQQPQE




QVPLVQQQQF LGQQQPFPPQ





61
QPYPQPQPFP SQLPYLQLQP FPQPQLPYSQ PQPFRPQQPY



PQPQPQYSQP QQPISQQQQQ





121
QQQQQQQQQQ QQQILQQILQ QQLIPCMDVV LQQHNIAHGR



SQVLQQSTYQ LLQELCCQHL





181
WQIPEQSQCQ AIHNVVHAII LHQQQKQQQQ PSSQVSFQQP



LQQYPLGQGS FRPSQQNPQA





241
QGSVQPQQLP QFEEIRNLAL QTLPAMCNVY IPPYCTIAPF



GIFGTN

















TABLE 4








Amino acid sequence of Gamma-gliadin



precursor from Triticum aestivum (NCBI accession


no. P21292, (SEQ ID NO:4))

















1
MKTLLILTIL AMATTIATAN MQVDPSGQVQ WPQQQPFPQP




QQPFCQQPQR TIPQPHQTFH





61
HQPQQTFPQP QQTYPHQPQQ QFPQTQQPQQ PFPQPQQTFP



QQPQLPFPQQ PQQPFPQPQQ





121
PQQPFPQSQQ PQQPFPQPQQ QFPQPQQPQQ SFPQQQQPAI



QSFLQQQMNP CKNFLLQQCN





181
HVSLVSSLVS IILPRSDCQV MQQQCCQQLA QIPQQLQCAA



IHSVAHSIIM QQEQQQGVPI





241
LRPLFQLAQG LGIIQPQQPA QLEGIRSLVL KTLPTMCNVY



VPPDCSTINV PYANIDAGIG





301
GQ

















TABLE 5








Amino acid sequence of Gamma-gliadin B precursor



from Triticum aestivum (NCBI accession no.


P06659, (SEQ ID NO:5))

















1
MKTLLILTIL AMAITIATAN MQADPSGQVQ WPQQQPFLQP




HQPFSQQPQQ IFPQPQQTFP





61
HQPQQQFPQP QQPQQQFLQP RQPFPQQPQQ PYPQQPQQPF



PQTQQPQQPF PQSKQPQQPF





121
PQPQQPQQSF PQQQPSLIQQ SLQQQLNPCK NFLLQQCKPV



SLVSSLWSII LPPSDCQVMR





181
QQCCQQLAQI PQQLQCAAIH SVVHSIIMQQ EQQEQLQGVQ



ILVPLSQQQQ VGQGILVQGQ





241
GIIQPQQPAQ LEVIRSLVLQ TLPTMCNVYV PPYCSTIRAP



FASIVASIGG Q

















TABLE 6








Amino acid sequence of Gamma-gliadin (Gliadin



B-III) from Triticum aestivum (NCBI accession no.


P04730, (SEQ ID NO:6))

















1
PQQPFPLQPQ QSFLWQSQQP FLQQPQQPSP QPQQVVQIIS




PATPTTIPSA GKPTSAPFPQ





61
QQQQHQQLAQ QQIPVVQPSI LQQLNPCKVF LQQQCSPVAM



PQRLARSQML QQSSCHVMQQ





121
QCCQQLPQIP QQSRYQAIRA IIYSIILQEQ QQVQGSIQSQ



QQQPQQLGQC VSQPQQQSQQ





181
QLGQQPQQQQ LAQGTFLQPH QIAQLEVMTS IALRILPTMC



SVNVPLYRTT TSVPFGVGTG





241
VGAY

















TABLE 7








Amino acid sequence of Gamma-gliadin precursor



from Triticum aestivum (NCBI accession no.


P08453, (SEQ ID NO:7))

















1
MKTLLILTIL AMAITIGTAN IQVDPSGQVQ WLQQQLVPQL




QQPLSQQPQQ TFPQPQQTFP





61
HQPQQQVPQP QQPQQPFLQP QQPFPQQPQQ PFPQTQQPQQ



PFPQQPQQPF PQTQQPQQPF





121
PQQPQQPFPQ TQQPQQPFPQ LQQPQQPFPQ PQQQLPQPQQ



PQQSFPQQQR PFIQPSLQQQ





181
LNPCKNILLQ QSKPASLVSS LWSIIWPQSD CQVMRQQCCQ



QLAQIPQQLQ CAAIHSVVHS





241
IIMQQQQQQQ QQQGIDIFLP LSQHEQVGQG SLVQGQGIIQ



PQQPAQLEAI RSLVLQTLPS





301
MCNVYVPPEC SIMRAPFASI VAGIGGQ

















TABLE 8








Amino acid sequence of Gamma-gliadin B-I



precursor from Triticum aestivum (NCBI accession


no. P04729, (SEQ ID NO:8))

















1
MKTFLVFALI AVVATSAIAQ METSCISGLE RPWQQQPLPP




QQSFSQQPPF SQQQQQPLPQ





61
QPSFSQQQPP FSQQQPILSQ QPPFSQQQQP VLPQQSPFSQ



QQQLVLPPQQ QQQQLVQQQI





121
PIVQPSVLQQ LNPCKVELQQ QCSPVAMPQR LARSQMWQQS



SCHVMQQQCC QQLQQIPEQS





181
RYEAIRAIIY SIILQEQQQG FVQPQQQQPQ QSGQGVSQSQ



QQSQQQLGQC SFQQPQQQLG





241
QQPQQQQQQQ VLQGTFLQPH QIAHLEAVTS IALRTLPTMC



SVNVPLYSAT TSVPFGVGTG





301
VGAY

















TABLE 9








Amino acid sequence of Gamma-gliadin precursor



from Triticum aestivum (NCBI accession no.


P08079, (SEQ ID NO:9))

















1
MKTLLILTIL AMAITIGTAN MQVDPSSQVQ WPQQQPVPQP




HQPFSQQPQQ TFPQPQQTFP





61
HQPQQQFPQP QQPQQQFLQP QQPFPQQPQQ PYPQQPQQPF



PQTQQPQQLF PQSQQPQQQF





121
SQPQQQFPQP QQPQQSFPQQ QPPFIQPSLQ QQVNPCKNFL



LQQCKPVSLV SSLWSMIWPQ





181
SDCQVMRQQC CQQLAQIPQQ LQCAAIHTII HSIIMQQEQQ



EQQQGMHILL PLYQQQQVGQ





241
GTLVQGQGII Q

















TABLE 10








Amino acid sequence of Alpha/beta-gliadin MM1



precursor (Prolamin) from Triticum aestivum (NCBI


accession no. P18573, (SEQ ID NO:10))

















1
MKTFLILALL AIVATTARIA VRVPVPQLQP QNPSQQQPQE




QVPLVQQQQF PGQQQPFPPQ





61
QPYPQPQPFP SQQPYLQLQP FPQPQLPYPQ PQLPYPQPQL



PYPQPQPFRP QQPYPQSQPQ





121
YSQPQQPISQ QQQQQQQQQQ QKQQQQQQQQ ILQQILQQQL



IPCRDVVLQQ HSIAYGSSQV





181
LQQSTYQLVQ QLCCQQLWQI PEQSRCQAIH NVVHAIILHQ



QQQQQQQQQQ QPLSQVSFQQ





241
PQQQYPSGQG SFQPSQQNPQ AQGSVQPQQL PQFEEIRNLA



LETLPAMCNV YIPPYCTIAP





301
VGIFGTN

















TABLE 11








Amino acid sequence of Alpha/beta-gliadin clone



PTO-A10 (Prolamin) from Triticum aestivum (NCBI


accession no. P04728, (SEQ ID NO:11))

















1
PQPQPQYSQP QQPISQQQQQ QQQQQQQQQQ EQQILQQILQ




QQLIPCMDVV LQQHNIAHGR





61
SQVLQQSTYQ LLQELCCQHL WQIPEQSQCQ AIHNVVHAII



LHQQQQKQQQ QPSSQFSFQQ





121
PLQQYPLGQG SFRPSQQNPQ AQGSVQPQQL PQFEIRNLAL



QTLPAMCNVY IPPYCTIAPF





181
GIFGTN

















TABLE 12








Amino acid sequence of Alpha/beta-gliadin clone



PW8142 precursor (Prolamin) from Triticum aestivum


(NCBI accession no. P04727, (SEQ ID NO:12))

















1
MKTFLILALV ATTATTAVRV PVPQLQPKNP SQQQPQEQVP




LVQQQQFPGQ QQQFPPQQPY





61
PQPQPFPSQQ PYLQLQPFPQ PQPFLPQLPY PQPQSFPPQQ



PYPQQRPKYL QPQQPISQQQ





121
AQQQQQQQQQ QQQQQQQQIL QQILQQQLIP CRDVVLQQHN



IAHASSQVLQ QSTYQLLQQL





181
CCQQLLQIPE QSRCQAIHNV VHAIIMHQQE QQQQLQQQQQ



QQLQQQQQQQ QQQQQPSSQV





241
SFQQPQQQYP SSQGSFQPSQ QNPQAQGSVQ PQQLPQFAEI



RNLALQTLPA MCNVYIPPHC





301
STTIAPFGIF GTN

















TABLE 13








Amino acid sequence of Alpha/beta-gliadin clone



PW1215 precursor (Prolamin) from Triticum aestivum


(NCBI accession no. P04726, (SEQ ID NO:13))

















1
MKTFLILALL AIVATTATTA VRVPVPQPQP QNPSQPQPQG




QVPLVQQQQF PGQQQQFPPQ





61
QPYPQPQPFP SQQPYLQLQP FPQPQPFPPQ LPYPQPPPFS



PQQPYPQPQP QYPQPQQPIS





121
QQQAQQQQQQ QQQQQQQQQQ QQILQQILQQ QLIPCRDVVL



QQHNIAHARS QVLQQSTYQP





181
LQQLCCQQLW QIPEQSRCQA IHNVVHAIIL HQQQRQQQPS



SQVSLQQPQQ QYPSGQGFFQ





241
PSQQNPQAQG SVQPQQLPQF EEIRNLALQT LPRMCNVYIP



PYCSTTIAPF GIFGTN

















TABLE 14








Amino acid sequence of Alpha/beta-gliadin A-IV



precursor (Prolamin) from Triticum aestivum (NCBI


accession no. P04724, (SEQ ID NO:14))

















1
MKTFLILALR AIVATTATIA VRVPVPQLQP QNPSQQQPQK




QVPLVQQQQF PGQQQPFPPQ





61
QPYPQQQPFP SQQPYMQLQP FPQPQLPYPQ PQLPYPQPQP



FRPQQSYPQP QPQYSQPQQP





121
ISQQQQQQQQ QQQQQQQILQ QILQQQLIPC RDVVLQQHSI



AHGSSQVLQQ STYQLVQQFC





181
CQQLWQIPEQ SRCQAIHNVV HAIILHQQQQ QQQQQQQQQQ



QPLSQVCFQQ SQQQYPSGQG





241
SFQPSQQNPQ AQGSVQPQQL PQFEEIRNLA LETLPAMCNV



YIPPYCTIAP VGIFGTN

















TABLE 15








Amino acid sequence of Alpha/beta-gliadin A-III



precursor (Prolamin) from Triticum aestivum (NCBI


accession no. P04723, (SEQ ID NO:15))

















1
MKTFLILALL AIVATTATSA VRVPVPQLQP QNPSQQQPQE




QVPLMQQQQQ FPGQQEQFPP





61
QQPYPHQQPF PSQQPYPQPQ PFPPQLPYPQ TQPFPPQQPY



PQPQPQYPQP QQPISQQQAQ





121
QQQQQQQTLQ QILQQQLIPC RDVVLQQHNI AHASSQVLQQ



SSYQQLQQLC CQQLFQIPEQ





181
SRCQAIHNVV HAIILHHHQQ QQQQPSSQVS YQQPQEQYPS



GQVSFQSSQQ NPQAQGSVQP





241
QQLPQFQEIR NLALQTLPAM CNVYIPPYCS TTIAPFGIFG



TN

















TABLE 16








Amino acid sequence of Alpha/beta-gliadin A-II



precursor (Prolamin) from Triticum aestivum (NCBI


accession no. P04722, (SEQ ID NO:16))

















1
MKTFPILALL AIVATTATTA VRVPVPQLQL QNPSQQQPQE




QVPLVQEQQF QGQQQPFPPQ





61
QPYPQPQPFP SQQPYLQLQP FPQPQLPYPQ PQPFRPQQPY



PQPQPQYSQP QQPISQQQQQ





121
QQQQQQQQQQ ILQQILQQQL IPCRDVVLQQ HNIAHGSSQV



LQESTYQLVQ QLCCQQLWQI





181
PEQSRCQAIH NVVHAIILHQ QHHHHQQQQQ QQQQQPLSQV



SFQQPQQQYP SGQGFFQPSQ





241
QNPQAQGSFQ PQQLPQFEEI RNLALQTLPA MCNVYIPPYC



TIAPFGIFGT N

















TABLE 17








Amino acid sequence of Alpha/beta-gliadin A-I



precursor (Prolamin) from Triticum aestivum (NCBI


accession no. P04721, (SEQ ID NO:17))

















1
MKTFLILALL AIVATTATTA VRVPVPQLQP QNPSQQQPQE




QVPLVQQQQF LGQQQPFPPQ





61
QPYPQPQPFP SQQPYLQLQP FLQPQLPYSQ PQPFRPQQPY



PQPQPQYSQP QQPISQQQQQ





121
QQQQQQQQQQ QQQQIIQQIL QQQLIPCMDV VLQQHNIVHG



KSQVLQQSTY QLLQELCCQH





181
LWQIPEQSQC QAIHNVVHAI ILHQQQKQQQ QPSSQVSFQQ



PLQQYPLGQG SFRPSQQNPQ





241
AQGSVQPQQL PQFEEIRNLA RK

















TABLE 18








Amino acid sequence of gamma gliadin from




Triticum aestivum (NCBI accession no. AAQ63860,



(SEQ ID NO:18))

















1
MNIQVDPSSQ VPWPQQQPFP QPHQPFSQQP QQTFPQPQQT




FPHQPQQQFS QPQQPQQQFI





61
QPQQPFPQQP QQTYPQRPQQ PFPQTQQPQQ PFPQSQQPQQ



PFPQPQQQFP QPQQPQQSFP





121
QQQPSLIQQS LQQQLNPCKN FLLQQCKPVS LVSSLWSMIL



PRSDCQVMRQ QCCQQLAQIP





181
QQLQCAAIHS IVHSIIMQQE QQEQRQGVQI LVPLSQQQQV



GQGTLVQGQG IIQPQQPAQL





241
EVIRSLVLQT LATMCNVYVP PYCSTIRAPF ASIVAGIGGQ



YR

















TABLE 19








Amino acid sequence of Omega-gliadin from




Triticum monococcum (NCBI accession no. P02865,



(SEQ ID NO:19))


















1
ARQLNPSDQE LQSPQQLYPQ QPYPQQPY










Fragments of gliadin that may be used in the practice of the invention include, but are not limited to, Leu-Gln-Leu-Gln-Pro-Phe-Pro-Gln-Pro-Gln-Leu-Pro-Tyr-Pro-Gln-Pro-Gln-Leu-Pro-Tyr-Pro-Gln-Pro-Gln-Leu-Pro-Tyr-Pro-Gln-Pro-Gln-Pro-Phe, which corresponds to amino acids 57-89 of the alpha-gliadin sequence of Table 1, and Leu-Gly-Gln-Gln-Gln-Pro-Phe-Pro-Pro-Gln-Gln-Pro-Tyr (SEQ ID NO:20), which corresponds to amino acids 32-44 of the alpha-gliadin sequence of Table 1 with the proline at position 32 of the wildtype alpha-gliadin sequence mutated to a leucine. Other suitable fragments of gliadin may be prepared, for example, by digesting a purified gliadin with proteolytic enzymes (e.g., pepsin, trypsin or mixtures thereof) and isolating peptides. Peptides may be isolated using any technique known in the art such as reverse phase high pressure liquid chromatography (RP-HPLC).


Modulators of CXCR3 signaling may be inhibitors, enhancers, or agonists. Inhibitors are useful for treating diseases characterized by inflammation, including autoimmune diseases and particularly including celiac disease. Enhancers or agonists can be used for increasing permeability of a tissue to a desired agent, e.g., a therapeutic agent which is less than optimally absorbed.


Antibodies to CXRC3 can be therapeutically by administration to patients in need thereof. Such patients include those with gluten-related diseases as well as diseases associated with inflammation and autoimmunity. Administration can be by any means known in the art for administration of antibodies. Such methods include, but are not limited to intravenous, intramuscular, and subcutaneous administration. Any form of antibodies known in the art can be used. The antibodies can be polyclonal or monoclonal. They can be, e.g., humanized or human or chimeric or recombinant. The antibodies can be of any isotype. They may be single chain antibodies, or fragments of antibodies such as F(ab′)2.


Signaling by CXCR3 can be measured by any means known in the art. Signaling events which can be determined include decrease in TEER, increase in zonulin release, microglia recruitment, tyrosine kinase phosphorylation and chemotaxis, and increase in MMP-2 and MMP-9 gelatinolytic activity in cell-conditioned media.


The invention provides methods of identifying agents, compounds or lead compounds for agents active at the level of CXCR3-ligand interaction. Generally, screening methods of the invention involve assaying for compounds which modulate the interaction of CXCR3 and ligand (e.g., gliadin or fragment thereof). A wide variety of assays for binding agents is provided including labeled in vitro protein-ligand binding assays, cell based assays, immunoassays, etc. A wide variety of formats may be used, including co-immunoprecipitation, 2-hybrid transactivation, fluorescent polarization, NMR, fluorescent resonance energy transfer (FRET), transcriptional activation, etc. For example, a wide variety of NMR-based methods are available to rapidly screen libraries of small compounds for binding to protein targets (Hajduk, P. J., et al. Quarterly Reviews of Biophysics, 1999. 32 (3): 211-40). In some embodiments, methods of the invention may be automated (e.g., high throughput screening) and may be used to screen chemical libraries for lead compounds. Identified compounds may be used to treat diseases involving CXCR3 signaling, for example, autoimmune diseases. Compounds identified by the methods of the invention may be further optimized to modulate CXCR3 signaling, for example, may derivatized. Multiple iterations of screening and derivatization may be employed to optimize the modulation of CXCR3 signaling.


In vitro ligand binding assays employ a mixture of components including CXCR3 or fragment thereof and ligand (e.g., gliadin or fragment thereof). CXCR3 and/or gliadin may be provided as fusion proteins (e.g., with purification tags such as 6-His). Assay mixtures typically further comprise a compound to be tested for CXCR3 modulating activity. Compounds to be tested may be of any kind known to those skilled in the art, for example, may be organic compounds, peptides, proteins, nucleic acids, lipids, carbohydrates and mixtures thereof. A variety of other reagents may also be included in the mixture including, but not limited to, salts, buffers, neutral proteins, e.g. albumin, detergents, protease inhibitors, nuclease inhibitors, antimicrobial agents, etc.


In general, assay mixtures may be incubated under conditions in which, but for the presence of the compound to be tested, CXCR3 specifically binds the ligand (e.g., gliadin or fragment thereof) with a reference binding affinity. The mixture components can be added in any order that provides for the requisite bindings and incubations may be performed at any temperature which facilitates optimal binding. Incubation periods are likewise selected for optimal binding. In some embodiments, incubation periods may be minimized to facilitate rapid, high-throughput screening.


After incubation, the effect of the compound to be tested on the CXCR3-ligand binding may be detected by any convenient way. For example, CXCR3 or ligand may be immobilized, and the other labeled; then in a solid-phase format, any of a variety of methods may be used to detect the label depending on the nature of the label and other assay components, e.g. through optical or electron density, radiative emissions, nonradiative energy transfers, etc. or indirectly detected with antibody conjugates, etc.


A difference in the binding affinity of CXCR3 and the ligand in the absence of the compound to be tested as compared with the binding affinity in the presence of the compound to be tested indicates that the compound modulates the binding of CXCR3 to the ligand. A difference, as used herein, is statistically significant and preferably represents at least a 50%, 60%, 70%, 80%, or 90% difference.


The above disclosure generally describes the present invention. All references disclosed herein are expressly incorporated by reference. A more complete understanding can be obtained by reference to the following specific examples which are provided herein for purposes of illustration only, and are not intended to limit the scope of the invention.


EXAMPLE 1

In order to identify the putative receptor activated by gliadin, we performed experiments using a gliadin affinity column through which intestinal cell lysates were loaded. We eluted proteins with a step salt gradient. Three clear protein bands were observed on SDS-polyacrylamide gels with molecular weights of 97, 90, and 83 kDa. The observed proteins eluted at 0.2 M and 0.3 M NaCl off the affinity column. Mass spectrometry analysis of proteins that bound to the column identified XP125429 in the sequence database (see Table 20). This sequence includes a precursor of the R3 receptor and implicates the CXCR3 receptor as one of the proteins engaged by in (see FIG. 4).

TABLE 20Amino acid sequence of the protein identified byfragment sequencing (NCBI accession non.XP_125249 (SEQ ID NO:21))1MASGADSKGD DLSTAILKQK NRPNRLIVDE AINEDNSVVSLSQPKMDELQ LFRGDTVLLK61GKKRREAVCI VLSDDTCSDE KIRMNRVVRN NLRVRLGDVISIQPCPDVKY GKRIHVLPID121DTVEGITGNL FEVYLKPYFL EAYRPIRKGD IFLVRGGMRAVEFKVVETDP SPYCIVAPDT181VIHCEGEPIK REDEEESLNE VGYDDIGGCR KQLAQIKEMVELPLRHPALF KAIGVKPPRG241ILLYGPPGTG KTLIARAVAN ETGAFFFLIN GPEIMSKLAGESESNLRKAF EEAEKNAPAI301IFIDELDAIA PKREKTHGEV ERRIVSQLLT LMDGLKQRAHVIVMAATNRP NSIDPALRRF361GRFDREVDIG IPDATGRLEI LQIHTKNMKL ADDVDLEQVANETHGHVGAD LAALCSEAAL421QAIRKKMDLI DLEDETIDAE VMNSLAVTMD DFRWALSQSNPSALRETVVE VPQVTWEDIG481GLEDVKRELQ ELVQYPVEHP DKFLKFGMTP SKGVLFYGPPGCGKTLLAKA IANECQANFI541SIKGPELLTM WFGESEANVR EIFDKARVLF FDELDSIAKARGGNIGDGGG AADRVINQIL601TEMDGMSTKK NVFIIGATNR PDIIDPAILR PGRLDQLIYIPLPDEKSRVA ILKANLRKSP661VAKDVDLEFL AKMTNGFSGA DLTEICQRAC KLAIRESIESEIRRERERQT NPSAMEVEED721DPVPEIRRDH FEEAMRFARR SVSDNDIRKY EMFAQTLQQSRGFGSFRFPS GNQGGAGPSQ781GSGGGTGGSV YTEDNDDDLY G


Human CXCR3 has 368 amino acid residues and a calculated molecular weight of 40,459. The sequences of human CXCR3 and mouse CXCR3 are provided in the following tables.

TABLE 21Amino acid sequence of CXCR3 from Homo sapiens(NCBI accession no. AAH34403, (SEQ ID NO:22))1MVLEVSDHQV LNDAEVAALL ENFSSSYDYG ENESDSCCTSPPCPQDFSLN FDRAFLPALY61SLLFLLGLLG NGAVAAVLLS RRTALSSTDT FLLHLAVADTLLVLTLPLWA VDAAVQWVFG121SGLCKVAGAL FNINFYAGAL LLACISFDRY LNIVHATQLYRRGPPARVTL TCLAVWGLCL181LFALPDFIFL SAHHDERLNA THCQYNFPQV GRTALRVLQLVAGFLLPLLV MAYCYAHILA241VLLVSRGQRR LRAMRLVVVV VVAFALCWTP YHLVVLVDILMDLGALARNC GRESRVDVAK301SVTSGLGYMH CCLNPLLYAF VGVKFRERMW MLLLRLGCPNQRGLQRQPSS SRRDSSWSET361SEASYSGL










TABLE 21








Amino acid sequence of CXCR3 from Mus musculus



(NCBI accession no. NP_034040, (SEQ ID NO:23))

















1
MYLEVSERQV LDASDFAFLL ENSTSPYDYG ENESDFSDSP




PCPQDFSLNF DRTFLPALYS





61
LLFLLGLLGN GAVAAVLLSQ RTALSSTDTF LLHLAVADVL



LVLTLPLWAV DAAVQWVFGP





121
GLCKVAGALF NINFYAGAFL LACISFDRYL SIVHATQIYR



RDPRVRVALT CIVVWGLCLL





181
FALPDEIYLS ANYDQRLNAT HCQYNFPQVG RTALRVLQLV



AGFLLPLLVM AYCYAHILAV





241
LLVSRGQRRF RAMRLVVVVV AAFAVCWTPY HLVVLVDILM



DVGVLARNCG RESHVDVAKS





301
VTSGMGYMHC CLNPLLYAFV GVKFREQMWM LFTRLGRSDQ



RGPQRQPSSS RRESSWSETT





361
EASYLGL









CXCR3 is a G-protein coupled receptor which is known to function as a receptor of SCYB9, SCYB10, and SCYB11, also known as MIG, IP10, and ITAC, cytokines implicated in inflammation. The receptor is also identified as CD183, GPR9, CKR-L2. The amino acid sequence of the receptor is shown as SEQ ID NO: 23. Human variants are known such as a R292Q and an A363T polymorphisms see SEQ ID NO:22.


Methods


We linked a-gliadin (a gift from Dr. Donald D. Kasarda) to CarboxyLink™ (Pierce Biotechnology, Rockford, Ill.) coupling gel to form an affinity column.


We prepared human intestine mucous membranes using protease inhibitors and a standard protocol.


One protocol which can be used involves the following steps: Tissues are washed with buffer D (20 mmol·L−1 Tris-HCl, 20 mmol·L−1 EDTA, 250 mmol·L−1 sucrose, pH 7.5) homogenized in buffer E (buffer D containing 5 mg·L−1 leupeptin, 2 mg·L−1 aprotinin, 1 mg·L−1 pepstatin, 10 mg·L−1 phenylmethylsulfonylfluoride (PMSF), and centrifuged at 5000×g, 4° C. for 10 min. Supernatants are centrifuged at 12000×g, 4° C. for 45 min. Precipitates are discarded and supernatants are centrifuged at 30000×g, 4° C. for an additional 90 min. Precipitates are dissolved in buffer E with 5 g·L−1 3[(3-cholamidopropyl) dimethylammonio]-1-propanesulfonate (CHAPS), sitting on ice for 60 min. with gentle mixing every five minutes.


EXAMPLE 2

Intestinal fragments isolated from normal mice, mounted on microsnapwells, and exposed to gliadin react by releasing zonulin. FIG. 1A. Following zonulin release, the intestinal permeability increases, suggesting a loss of the mucosal barrier function. FIG. 1B. This so-called “gluten effect” is detectable only when the protein is added to the surface (luminal side) of the intestine, suggesting that gliadin interacts with a receptor present on the enterocyte brush border.


To confirm the hypothesis that CXCR3 is the gliadin target receptor that needs to be activated in order to release zonulin, experiments were conducted using a CXCR3 knock out mouse model. Intestinal tissues isolated from these animals, mounted in the microsnapwell system, and exposed to gliadin failed to release zonulin and, consequently, no changes in intestinal permeability were detected. FIGS. 2 and 3. These results confirm the hypothesis that CXCR3 is a gliadin target receptor involved in zonulin release.


The “microsnapwell system,” a polarized model, is used to study the intestinal barrier function using human intestinal biopsies. The system evaluates the intestinal permeability of endoscopic jejunal biopsies by measuring the Trans Epithelial Electrical Resistance (TEER).


EXAMPLE 3

In vitro experiments using HEK cells transfected with CXCR3 were performed to study gliadin binding to the receptor by immunofluorescence (IF) microscopy. The in vitro IF experiments showed that gliadin bound on cells transfected with CXCR3 but not on cells transfected with vector alone.


As shown in FIG. 6, HEK293 cells transfected with vector expressing CXCR3 were specifically labeled with anti-CXCR3 mAb. The human CXCR3 sequence was inserted into pc DNA 3.1 (Invitrogen Corporation, Carlsbad, Calif.) under the control of a CMV1 promoter. Red trace shows results obtained with anti-CXCR3 mAb (marked with an arrow in 6B), blue trace shows results obtained with IgG1 isotype control. FIG. 6A shows the control transfection with vector alone while FIG. 6B shows the results obtained with vector expressing CXCR3. With vector alone, CXCR3 expression was 4.42%, mean 11.9 (FIG. 6A). In contrast, with cells transfected with vector expressing CXCR3, CXCR3 expression was 61.78%, mean 95.5.


When cells transfected with vector expressing CXCR3 were contacted with fluorescently labeled gliadin, the gliadin bound to the cells and not to the control cells that did not express CXCR3, thus PT-Gliadin co-localizes with CXCR3 in HEK293 transfected cells. In FIG. 7, nuclei were stained with DAPI (blue), CXCR3 were stained with monoclonal antibody labeled with RITC (red), and PT-Gliadin was labeled with FITC (green). Panel A shows nuclear staining only with cells transfected with control vector. Panel B shows the staining on the outside of the cells transfected with vector expressing CXCR3 and contacted with RITC-labeled monoclonal antibody specific for CXCR3. When the cells in Panel B were contacted with FITC-labeled gliadin, the gliadin co-localized with CXCR3.


EXAMPLE 4

Finally, the expression of co-stimulatory markers on peripheral blood mononuclear cells (PBMC) was studied in both normal subjects and patients affected by autoimmunity (celiac disease and type 1 diabetes). PBMC from autoimmune patients exposed to gliadin showed increase expression of co-stimulatory markers CD40, CD80, and CD86, and DR. The stimulation of DR expression (but not of the other markers) was prevented by blocking the CXCR3 receptor using specific antibodies. FIG. 8 shows the effect of PT-gliadin on HLA-DR expression in dendritic cells from normal volunteers is CXCR3 -dependent.


The antibodies used to measure costimulatory markers were all commercially available and were purchased from BD Biosciences and R&D Systems. From BD Biosciences: CD80 R-phycoerythrin (r-PE)-conjugated mouse anti-human monoclonal antibody (CD80 r-PE, cat.no. 557227), CD40 and CD86 fluorescein isothiocyanate (FITC)-conjugated mouse anti-human monoclonal antibodies (CD40 FITC, cat.no. 555588; CD86 FITC, cat.no 555657), HLA-DR PE-cy5-conjugated mouse anti-human monoclonal antibody (HLA-DR-cy5, cat.no. 555813). From R&D Systems: allophycocyanin-conjugated mouse monoclonal anti-human CXCR3 (CXCR3 APC, cat.no FAB160A). The antibodies used for blocking studies were: monoclonal anti-human CXCR3 antibody (cat.no.MAB160) and mouse IgG1 isotype control (cat.no.MAB002).


EXAMPLE 5

Ex vivo experiments to measure zonulin release and intestinal transepithelial electrical resistance (TEER) changes in response to gliadin exposure were performed using mouse small intestine mounted in microsnapwell chambers. The ex vivo experiments were conducted on both CXCR3 knock out (KO) and C57BL/6 wild-type (WT) mouse intestinal tissues mounted in microsnapwells. When exposed to PT-gliadin, intestinal segments obtained from WT mice (n=10) released zonulin (0.33±0.06 vs. 0.61±0.13 ng/mg protein, baseline vs. post-gliadin exposure, respectively; p<0.04, see FIG. 9B) and showed a significant TEER decrement (24.1±4.5 Ω/cm2 vs. 14.7±3.2 baseline vs. post-gliadin exposure, respectively; p<0.02, see FIG. 9A). Conversely, intestinal segments obtained from CXCR3 KO mice (n=18) exposed to PT-gliadin failed to release zonulin (0.56±0.15 vs. 0.45±0.13 ng/mg protein, baseline vs. post-gliadin exposure, respectively; p=N.S., see FIG. 10B) and showed no TEER changes (20.0±4.8 Ω/cm2 vs. 16.5±4.9, baseline vs. post-gliadin exposure, respectively; p N.S., see FIG. 10A).


Having now fully described the present invention in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious to one of ordinary skill in the art that the same can be performed by modifying or changing the invention within a wide and equivalent range of conditions, formulations and other parameters without affecting the scope of the invention or any specific embodiment thereof, and that such modifications or changes are intended to be encompassed within the scope of the appended claims. All publications, patents and patent applications mentioned in this specification are indicative of the level of skill of those skilled in the art to which this invention pertains, and are herein incorporated by reference to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference.

Claims
  • 1. A method to screen for modulators of CXCR3 signaling, comprising: contacting a gliadin or fragment of gliadin comprising at least six amino acid residues with CXCR3; determining binding of the fragment of gliadin to CXCR3; identifying a fragment of gliadin which binds to CXCR3 as a modulator of CXCR3 signaling.
  • 2. The method of claim 1 wherein the fragment is at least seven amino acid residues.
  • 3. The method of claim 1 wherein the fragment is at least eight amino acid residues.
  • 4. The method of claim 1 wherein the fragment is at least nine amino acid residues.
  • 5. The method of claim 1 wherein the fragment is at least ten amino acid residues.
  • 6. The method of claim 1 wherein the fragment is synthesized.
  • 7. The method of claim 1 wherein the fragment is a proteolytic product.
  • 8. A method to screen for modulators of CXCR3 signaling, comprising: contacting gliadin or a fragment of gliadin comprising at least six amino acid residues with a first cell which expresses CXCR3 and with a second cell which does not express CXCR3; determining binding of the fragment of gliadin to the first and second cells; identifying a fragment of gliadin which binds preferentially to the first cell relative to the second cell as a modulator of CXCR3 signaling.
  • 9. The method of claim 8 wherein the fragment is at least seven amino acid residues.
  • 10. The method of claim 8 wherein the fragment is at least eight amino acid residues.
  • 11. The method of claim 8 wherein the fragment is at least nine amino acid residues.
  • 12. The method of claim 8 wherein the fragment is at least ten amino acid residues.
  • 13. The method of claim 8 wherein the fragment is synthesized.
  • 14. The method of claim 8 wherein the fragment is a proteolytic product.
  • 15. A method to screen for modulators of CXCR3 signaling, comprising: contacting gliadin or a fragment of gliadin comprising at least six amino acid residues with CXCR3 in the presence of a ligand selected from the group consisting of gliadin, IP10, MIG, or ITAC; determining inhibition of binding of the ligand to CXCR3 caused by the fragment of gliadin; identifying a fragment of gliadin which inhibits binding of the ligand to CXCR3 as a modulator of CXCR3 signaling.
  • 16. The method of claim 15 wherein the fragment is at least seven amino acid residues.
  • 17. The method of claim 15 wherein the fragment is at least eight amino acid residues.
  • 18. The method of claim 15 wherein the fragment is at least nine amino acid residues.
  • 19. The method of claim 15 wherein the fragment is at least ten amino acid residues.
  • 20. The method of claim 15 wherein the fragment is synthesized.
  • 21. The method of claim 15 wherein the fragment is a proteolytic product.
  • 22. A method to screen for modulators of CXCR3 signaling, comprising: contacting in the presence of a ligand selected from the group consisting of gliadin, IP10, MIG, or ITAC, gliadin or a fragment of gliadin comprising at least six amino acid residues with a cell which expresses CXCR3; determining binding of the ligand to the cell; identifying a fragment of gliadin which inhibits binding of the ligand to the cell as a modulator of CXCR3 signaling.
  • 23. The method of claim 22 wherein the fragment is at least seven amino acid residues.
  • 24. The method of claim 22 wherein the fragment is at least eight amino acid residues.
  • 25. The method of claim 22 wherein the fragment is at least nine amino acid residues.
  • 26. The method of claim 22 wherein the fragment is at least ten amino acid residues.
  • 27. The method of claim 22 wherein the fragment is synthesized.
  • 28. The method of claim 22 wherein the fragment is a proteolytic product.
  • 29. A method to screen for modulators of zonulin release, comprising: contacting a test compound with CXCR3; determining binding of the test compound to CXCR3; identifying a test compound which binds to CXCR3 as a modulator of zonulin release.
  • 30. The method of claim 29 wherein the test compound is a fragment of gliadin comprising at least six amino acid residues.
  • 31. A method to screen for modulators of zonulin release, comprising: contacting a test compound with CXCR3; determining binding of gliadin or a fragment of gliadin to CXCR3 in the presence and absence of the test compound; identifying a test compound which inhibits binding of the gliadin or fragment of gliadin to CXCR3 as a modulator of zonulin release.
  • 32. The method of claim 31 wherein the test compound is a fragment of gliadin comprising at least six amino acid residues.
  • 33. A method to screen for modulators of zonulin release, comprising: contacting a test compound with a first cell which expresses CXCR3 and with a second cell which does not express CXCR3; determining binding of the test compound to the first and second cells; identifying a test compound which binds preferentially to the first cell relative to the second cell as a modulator of zonulin release.
  • 34. The method of claim 33 wherein the test compound is a fragment of gliadin comprising at least six amino acid residues.
  • 35. A method to screen for modulators of zonulin release, comprising: contacting a test compound with a cell which expresses CXCR3; determining binding of gliadin or a fragment of gliadin comprising at least six amino acids to the cell; identifying a test compound which inhibits binding of gliadin or fragment to the cell as a modulator of zonulin release.
  • 36. The method of claim 35 wherein the test compound is a fragment of gliadin comprising at least six amino acid residues.
  • 37. A method of treating a patient with celiac disease, comprising: administering to the patient an antibody which specifically binds to CXCR3, whereby zonulin release is inhibited.
  • 38. A method of treating a patient with a disease characterized by undesired CXCR3 signaling, comprising: administering to the patient an antibody which specifically binds to CXCR3, whereby CXCR3 signaling is inhibited.
  • 39. The method of claim 38, wherein the disease is diabetes.
  • 40. The method of claim 38, wherein the disease is autoimmune hepatitis.
  • 41. The method of claim 38, wherein the disease is multiple sclerosis.
  • 42. The method of claim 38, wherein the disease is autism.
  • 43. The method of claim 38, wherein the disease is gluten allergy.
  • 44. The method of claim 38, wherein the disease is dermatitis herpetiformis.
  • 45. The method of claim 38, wherein the disease is systemic lupus erythematosus.
  • 46. The method of claim 38, wherein the disease is Grave's disease.
  • 47. The method of claim 38, wherein the disease is Hashimoto's disease.
  • 48. The method of claim 38, wherein the disease is depression.
  • 49. The method of claim 38, wherein the disease is gluten ataxia.
  • 50. The method of claim 38, wherein the disease is biliary cirrhosis.
  • 51. The method of claim 38, wherein the disease is rheumatoid arthritis.
  • 52. The method of claim 38, wherein the disease is IgA nephropathy.
  • 53. A method of treating a patient with gluten sensitivity, comprising: administering to the patient an antibody which specifically binds to CXCR3, whereby zonulin release is inhibited.
  • 54. The method of any of claims 37-53, wherein the antibody is monoclonal.
  • 55. The method of any of claims 37-53, wherein the antibody is polyclonal.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. provisional patent application Ser. No. 60/653,118, filed Feb. 16, 2005, and to U.S. provisional patent application Ser. No. 60/741,998, filed Dec. 2, 2005, the contents of both of which are specifically incorporated herein by reference.

Provisional Applications (2)
Number Date Country
60653118 Feb 2005 US
60741998 Dec 2005 US