Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS FOR SCREENING DRUGS FOR REDUCING CD28 COSTIMULATION OF T CELLS
Document Type and Number:
WIPO Patent Application WO/2014/173840
Kind Code:
A1
Abstract:
The present invention relates to methods for screening drugs for reducing CD28 costimulation of T cells in a subject in need thereof. In particular, the present invention relates to a method for screening a plurality of test substances useful for reducing CD28 costimulation of T cells in a subject in need thereof comprising the steps consisting of a1) testing each of the test substances for its ability to bind to a RLTR polypeptide and a2) positively selecting the test substances that are able to bind to the RLTR polypeptide.

Inventors:
MALISSEN BERNARD (FR)
MALISSEN MARIE (FR)
Application Number:
PCT/EP2014/058006
Publication Date:
October 30, 2014
Filing Date:
April 18, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INSERM INST NAT DE LA SANTÉ ET DE LA RECH MÉDICALE (FR)
UNIVERSITÉ D AIX MARSEILLE (FR)
CENTRE NAT RECH SCIENT (FR)
International Classes:
G01N33/50
Other References:
MATSUZAKA Y ET AL: "Identification, expression analysis and polymorphism of a novel RLTPR gene encoding a RGD motif, tropomodulin domain and proline/leucine-rich regions", GENE, ELSEVIER, AMSTERDAM, NL, vol. 343, no. 2, 22 December 2004 (2004-12-22), pages 291 - 304, XP004704089, ISSN: 0378-1119, DOI: 10.1016/J.GENE.2004.09.004
QIAO GUILIN ET AL: "T-cell receptor-induced NF-kappaB activation is negatively regulated by E3 ubiquitin ligase Cbl-b", MOLECULAR AND CELLULAR BIOLOGY, AMERICAN SOCIETY FOR MICROBIOLOGY, WASHINGTON, US, vol. 28, no. 7, 1 April 2008 (2008-04-01), pages 2470 - 2480, XP002540681, ISSN: 0270-7306, DOI: 10.1128/MCB.01505-07
T. KIM ET AL: "Mechanism for CARMIL Protein Inhibition of Heterodimeric Actin-capping Protein", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 287, no. 19, 4 May 2012 (2012-05-04), pages 15251 - 15262, XP055067834, ISSN: 0021-9258, DOI: 10.1074/jbc.M112.345447
C-C HSU ET AL: "Identifying LRRC16B as an oncofetal gene with transforming enhancing capability using a combined bioinformatics and experimental approach", ONCOGENE, vol. 30, no. 6, 10 February 2011 (2011-02-10), pages 654 - 667, XP055067857, ISSN: 0950-9232, DOI: 10.1038/onc.2010.451
SMITH; WATERMAN, J. THEOR. BIOL., vol. 91, no. 2, 1981, pages 370 - 380
NEEDLEMAN; WUNSCH, J. MIOL. BIOL., vol. 48, no. 3, 1972, pages 443 - 453
PEARSON; LIPMAN, PNAS, USA, vol. 85, no. 5, 1988, pages 2444 - 2448
"Current Protocols in Molecular Biology", vol. 1, 1994, JOHN WILEY & SONS, INC.
ED HARLOW; DAVID LANE: "Antibodies, A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
OKKENHAUG, K. ET AL.: "A point mutation in CD28 distinguishes proliferative signals from survival signals", NATURE IMMUNOLOGY, vol. 2, 2001, pages 325 - 332, XP002960866, DOI: doi:10.1038/86327
HOLDORF, A.D.; LEE, K.H.; BURACK, W.R.; ALLEN, P.M.; SHAW, A.S.: "Regulation of Lck activity by CD4 and CD28 in the immunological synapse", NATURE IMMUNOLOGY, vol. 3, 2002, pages 259 - 264
ISAKOV, N.; ALTMAN, A.: "PKC-theta-mediated signal delivery from the TCR/CD28 surface receptors", FRONT IMMUNOL, vol. 3, 2012, pages 273
RAAB, M.; PFISTER, S.; RUDD, C.E.: "CD28 signaling via VAV/SLP-76 adaptors: regulation of cytokine transcription independent of TCR ligation", IMMUNITY, vol. 15, 2001, pages 921 - 933
PAGAN, A.J.; PEPPER, M.; CHU, H.H.; GREEN, J.M.; JENKINS, M.K.: "CD28 Promotes CD4+ T cell clonal expansion during infection independently of its YMNM and PYAP motifs", JOURNAL OF IMMUNOLOGY, vol. 189, 2012, pages 2909 - 2917
THOME, M.; CHARTON, J.E.; PELZER, C.; HAILFINGER, S: "Antigen receptor signaling to NF-kappaB via CARMAI, BCL10, and MALT1", COLD SPRING HARBOR PERSPECTIVES IN BIOLOGY, vol. 2, 2010, pages A003004
WANG, X.; CHUANG, H.C.; LI, J.P.; TAN, T.H.: "Regulation of PKC-theta function by phosphorylation in T cell receptor signaling", FRONT IMMUNOL, vol. 3, 2012, pages 197
JIANG, C.; LIN, X.: "Regulation of NF-kappaB by the CARD proteins", IMMUNOLOGICAL REVIEWS, vol. 246, 2012, pages 141 - 153
YOKOSUKA, T. ET AL.: "Spatiotemporal regulation of T cell costimulation by TCR-CD28 microclusters and protein kinase C theta translocation", IMMUNITY, vol. 29, 2008, pages 589 - 601, XP002733105, DOI: doi:10.1016/j.immuni.2008.08.011
DUSTIN, M.L.: "The cellular context of T cell signaling", IMMUNITY, vol. 30, 2009, pages 482 - 492
VARDHANA, S.; CHOUDHURI, K.; VARMA, R.; DUSTIN, M.L.: "Essential role of ubiquitin and TSG101 protein in formation and function of the central supramolecular activation cluster", IMMUNITY, vol. 32, 2010, pages 531 - 540
YOKOSUKA, T. ET AL.: "Spatiotemporal basis of CTLA-4 costimulatory molecule-mediated negative regulation of T cell activation", IMMUNITY, vol. 33, 2010, pages 326 - 339
AGUADO, E. ET AL.: "Induction of T Helper Type 2 Immunity by a Point Mutation in the LAT Adaptor", SCIENCE, vol. 296, 2002, pages 2036 - 2040, XP002207182, DOI: doi:10.1126/science.1069057
SOMMERS, C.L. ET AL.: "A LAT mutation that inhibits T cell development yet induces lymphoproliferation", SCIENCE, vol. 296, 2002, pages 2040 - 2043, XP002249670, DOI: doi:10.1126/science.1069066
WANG, Y. ET AL.: "Th2 lymphoproliferative disorder of LatY136F mutant mice unfolds independently of TCR-MHC engagement and is insensitive to the action of Foxp3+ regulatory T cells", JLMMUNOL, vol. 180, 2008, pages 1565 - 1575
MINGUENEAU, M. ET AL.: "Loss of the LAT adaptor converts antigen-responsive T cells into pathogenic effectors that function independently of the T cell receptor", IMMUNITY, vol. 31, 2009, pages 197 - 208
CHEVRIER, S.; GENTON, C.; MALISSEN, B.; MALISSEN, M.; ACHA-ORBEA, H: "Dominant Role of CD80-CD86 Over CD40 and ICOSL in the Massive Polyclonal B Cell Activation Mediated by LAT(Y136F) CD4(+) T Cells", FRONT IMMUNOL, vol. 3, 2012, pages 27
BEUTLER, B. ET AL.: "Genetic analysis of resistance to viral infection", NAT REV IMMUNOL, vol. 7, 2007, pages 753 - 766
COOK, M.C.; VINUESA, C.G.; GOODNOW, C.C: "ENU-mutagenesis: insight into immune function and pathology", CURRENT OPINION IN IMMUNOLOGY, vol. 18, 2006, pages 627 - 633, XP025079067, DOI: doi:10.1016/j.coi.2006.07.011
LIANG, Y.; NIEDERSTRASSER, H.; EDWARDS, M.; JACKSON, C.E.; COOPER, J.A.: "Distinct roles for CARMIL iso forms in cell migration", MOL BIOL CELL, vol. 20, 2009, pages 5290 - 5305
HERNANDEZ-VALLADARES, M. ET AL.: "Structural characterization of a capping protein interaction motif defines a family of actin filament regulators", NAT STRUCT MOL BIOL, vol. 17, 2010, pages 497 - 503
FUJIWARA, I.; REMMERT, K.; HAMMER, J.A., 3RD: "Direct observation of the uncapping of capping protein-capped actin filaments by CARMIL homology domain 3", JBIOL CHEM, vol. 285, 2010, pages 2707 - 2720
YANG, C. ET AL.: "Mammalian CARMIL inhibits actin filament capping by capping protein", DEV CELL, vol. 9, 2005, pages 209 - 221
NG, A.C. ET AL.: "Human leucine-rich repeat proteins: a genome-wide bioinformatic categorization and functional analysis in innate immunity", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 108, no. 1, 2011, pages 4631 - 4638
JUNTTILA, M.R.; SAARINEN, S.; SCHMIDT, T.; KAST, J.; WESTERMARCK, J.: "Single-step Strep- tag purification for the isolation and identification of protein complexes from mammalian cells", PROTEOMICS, vol. 5, 2005, pages 1199 - 1203
BACHMANN, M.F. ET AL.: "T cell responses are governed by avidity and costimulatory thresholds", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 26, 1996, pages 2017 - 2022
SHAHINIAN, A. ET AL.: "Differential T cell costimulatory requirements in CD28-deficient mice", SCIENCE, vol. 261, 1993, pages 609 - 612, XP002952111, DOI: doi:10.1126/science.7688139
HOGQUIST, K.A. ET AL.: "T cell receptor antagonist peptides induce positive selection", CELL, vol. 76, 1994, pages 17 - 27, XP024245323, DOI: doi:10.1016/0092-8674(94)90169-4
KAYE, J. ET AL.: "Selective development of CD4+ T cells in transgenic mice expressing a class II MHC-restricted antigen receptor", NATURE, vol. 341, 1989, pages 746 - 749
TAI, X.; COWAN, M.; FEIGENBAUM, L.; SINGER, A.: "CD28 costimulation of developing thymocytes induces Foxp3 expression and regulatory T cell differentiation independently of interleukin 2", NATURE IMMUNOLOGY, vol. 6, 2005, pages 152 - 162
ROMAN, E.; SHINO, H.; QIN, F.X.; LIU, Y.J.: "Cutting edge: Hematopoietic-derived APCs select regulatory T cells in thymus", JOURNAL OF IMMUNOLOGY, vol. 185, 2010, pages 3819 - 3823
VANG, K.B. ET AL.: "Cutting edge: CD28 and c-Rel-dependent pathways initiate regulatory T cell development", JOURNAL OF IMMUNOLOGY, vol. 184, 2010, pages 4074 - 4077
SALEK-ARDAKANI, S. ET AL.: "B cell-specific expression of B7-2 is required for follicular Th cell function in response to vaccinia virus", JOURNAL OF IMMUNOLOGY, vol. 186, 2011, pages 5294 - 5303
NGUYEN, A.W.; DAUGHERTY, P.S.: "Evolutionary optimization of fluorescent proteins for intracellular FRET", NAT BIOTECHNOL, vol. 23, 2005, pages 355 - 360, XP002488502, DOI: doi:10.1038/nbt1066
CAMPI, G.; VARMA, R.; DUSTIN, M.L.: "Actin and agonist MHC-peptide complex- dependent T cell receptor microclusters as scaffolds for signaling", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 202, 2005, pages 1031 - 1036
YOKOSUKA, T. ET AL.: "Newly generated T cell receptor microclusters initiate and sustain T cell activation by recruitment of Zap70 and SLP-76", NAT IMMUNOL, vol. 6, 2005, pages 1253 - 1262
ZANIN-ZHOROV, A. ET AL.: "Protein kinase C-theta mediates negative feedback on regulatory T cell function", SCIENCE, vol. 328, 2010, pages 372 - 376
SINGLETON, K.L. ET AL.: "Spatiotemporal patterning during T cell activation is highly diverse", SCI SIGNAL, vol. 2, 2009, pages RAL5
SINGLETON, K. ET AL.: "A large T cell invagination with CD2 enrichment resets receptor engagement in the immunological synapse", JOURNAL OF IMMUNOLOGY, vol. 177, 2006, pages 4402 - 4413
DEFORD-WATTS, L.M. ET AL.: "The CD3 zeta subunit contains a phosphoinositide-binding motif that is required for the stable accumulation of TCR-CD3 complex at the immunological synapse", JOURNAL OF IMMUNOLOGY, vol. 186, 2011, pages 6839 - 6847
BURKHARDT, J.K.; CARRIZOSA, E.; SHAFFER, M.H.: "The actin cytoskeleton in T cell activation", ANNU REV IMMUNOL, vol. 26, 2008, pages 233 - 259
CEFAI, D. ET AL.: "CD28 receptor endocytosis is targeted by mutations that disrupt phosphatidylinositol 3-kinase binding and costimulation", JOURNAL OF IMMUNOLOGY, vol. 160, 1998, pages 2223 - 2230
BADOUR, K. ET AL.: "Interaction of the Wiskott-Aldrich syndrome protein with sorting nexin 9 is required for CD28 endocytosis and cosignaling in T cells", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 104, 2007, pages 1593 - 1598
BARNES, M.J. ET AL.: "Commitment to the regulatory T cell lineage requires CARMAI in the thymus but not in the periphery", PLOS BIOL, vol. 7, 2009, pages E51
MOLINERO, L.L. ET AL.: "CARMAI controls an early checkpoint in the thymic development of FoxP3+ regulatory T cells", JOURNAL OF IMMUNOLOGY, vol. 182, 2009, pages 6736 - 6743
JUNG, G.; REMMERT, K.; WU, X.; VOLOSKY, J.M.; HAMMER, J.A., 3RD: "The Dictyostelium CARMIL protein links capping protein and the Arp2/3 complex to type I myosins through their SH3 domains", J CELL BIOL, vol. 153, 2001, pages 1479 - 1497
TAKEDA, S. ET AL.: "Actin capping protein and its inhibitor CARMIL: how intrinsically disordered regions function", PHYS BIOL, vol. 8, 2011, pages 035005
HUTCHINGS, N.J.; CLARKSON, N.; CHALKLEY, R.; BARCLAY, A.N.; BROWN, M.H.: "Linking the T cell surface protein CD2 to the actin-capping protein CAPZ via CMS and CIN85", JBIOL CHEM, vol. 278, 2003, pages 22396 - 22403
MATSUZAKA, Y. ET AL.: "Identification, expression analysis and polymorphism of a novel RLTPR gene encoding a RGD motif, tropomodulin domain and proline/leucine-rich regions", GENE, vol. 343, 2004, pages 291 - 304, XP004704089, DOI: doi:10.1016/j.gene.2004.09.004
POINTON, J.J. ET AL.: "The chromosome 16q region associated with ankylosing spondylitis includes the candidate gene tumour necrosis factor receptor type I-associated death domain (TRADD", ANN RHEUM DIS, vol. 69, 2010, pages 1243 - 1246
SEDER, R.A.; PAUL, W.E.; DAVIS, M.M.; FAZEKAS DE ST GROTH, B: "The presence of interleukin 4 during in vitro priming determines the lymphokine-producing potential of CD4+ T cells from T cell receptor transgenic mice", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 176, 1992, pages 1091 - 1098
MALISSEN, M. ET AL.: "Altered T cell development in mice with a targeted mutation of the CD3-epsilon gene", EMBO J, vol. 14, 1995, pages 4641 - 4653
ORDONEZ-RUEDA, D. ET AL.: "A hypomorphic mutation in the Gfil transcriptional repressor results in a novel form of neutropenia", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 42, 2012, pages 2395 - 2408
HARAKALOVA, M. ET AL.: "Multiplexed array-based and in-solution genomic enrichment for flexible and cost-effective targeted next-generation sequencing", NAT PROTOC, vol. 6, 2011, pages 1870 - 1886, XP009168553, DOI: doi:10.1038/nprot.2011.396
KITAMURA, T. ET AL.: "Retrovirus-mediated gene transfer and expression cloning: powerful tools in functional genomics", EXP HEMATOL, vol. 31, 2003, pages 1007 - 1014, XP009118675, DOI: doi:10.1016/j.exphem.2003.07.005
HEAVEY, B.; CHARALAMBOUS, C.; COBALEDA, C.; BUSSLINGER, M.: "Myeloid lineage switch of Pax5 mutant but not wild-type B cell progenitors by C/EBPalpha and GATA factors", THE EMBO JOURNAL, vol. 22, 2003, pages 3887 - 3897
MORITA, S.; KOJIMA, T.; KITAMURA, T.: "Plat-E: an efficient and stable system for transient packaging of retroviruses", GENE THER, vol. 7, 2000, pages 1063 - 1066, XP002932672, DOI: doi:10.1038/sj.gt.3301206
RESKE-KUNZ, A.B.; RUDE, E.: "Insulin-specific T cell hybridomas derived from (H-2b x H-2k)Fl mice preferably employ Fl-unique restriction elements for antigen recognition", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 15, 1985, pages 1048 - 1054
TOKUNAGA, M.; KITAMURA, K.; SAITO, K.; IWANE, A.H.; YANAGIDA, T.: "Single molecule imaging of fluorophores and enzymatic reactions achieved by objective-type total internal reflection fluorescence microscopy", BIOCHEM BIOPHYS RES COMMUN, vol. 235, 1997, pages 47 - 53, XP001086476, DOI: doi:10.1006/bbrc.1997.6732
Attorney, Agent or Firm:
HIRSCH, Denise (7 rue Watt, Paris, FR)
Download PDF:
Claims:
CLAIMS:

1. A method for screening a plurality of test substances useful for reducing CD28 costimulation of T cells in a subject in need thereof comprising the steps consisting of al) testing each of the test substances for its ability to bind to a RLTR polypeptide and a2) positively selecting the test substances that are able to bind to the RLTR polypeptide.

2. The method according to claim 1 which further comprises the steps consisting of bl) testing each of the substances positively selected at step a2) for its ability to reduce the CD28 costimulation signalling in a T cell and b2) positively selecting the test substances that are able to reduce the CD28 costimulation in a T cell.

Description:
METHODS FOR SCREENING DRUGS FOR REDUCING CD28 COSTIMULATION

OF T CELLS

FIELD OF THE INVENTION:

The present invention relates to methods for screening drugs for reducing CD28 costimulation of T cells in a subject in need thereof.

BACKGROUND OF THE INVENTION:

T cell proliferation is the normal component of the immune reaction toward an antigen

(e.g. a pathogen antigen). However in certain circumstances T cell proliferation appears deleterious. For example, organ transplantation elicits a complex series of immunologic processes that are generally categorized as inflammation, immunity, tissue repair and structural reinforcement of damaged tissues. Typically T cell proliferation leads to inflammation by the secretion of proinflammatory cytokines, e.g., interleukin-2 (IL-2). Accordingly, the skilled man in the art has tried to develop immunosuppressive agents. In the "two-signal model" of T cell activation, the first signal is delivered via the T cell antigen receptor (TCR) following recognition of antigenic peptides bound to class I and II molecules of the major histocompatibility complex (MHC). This signal confers antigen specificity on T cell responses. The second "costimulatory" signal is delivered by the CD28 molecule upon binding of the CD80 and CD86 ligands on antigen presenting cells (APC). CD28 delivers signals via its cytoplasmic tail that has no intrinsic catalytic activity, but possesses several protein-protein interaction motifs enabling it to associate with effector molecules. By synergizing with the TCR, CD28 activates the classical NF-κΒ signaling pathway and leads to the transcription of genes involved in T cell survival and proliferation. TCR- CD28-induced NF-KB activation is thought to be triggered by the phosphorylation of the Carmal (Cardl l) adaptor by protein kinase C (PKC) family kinases, primarily PKC-Θ. This results in a conformational change in Carmal allowing it to recruit the ΙκΒ kinase complex and to stimulate the NF-κΒ signaling pathway. Accordingly, the selective inhibition of the agonist signal delivered to the T cell by CD28, for example by means of specific blocking of the CD28/CD80/CD86 interaction, has been explored in the prior art to prevent T lymphocyte activation, and thus to promote the induction of tolerance in the case of organ transplantation and also in the case of the treatment of autoimmune diseases. SUMMARY OF THE INVENTION:

The present invention relates to methods for screening drugs for reducing costimulation of T cells in a subject in need thereof.

DETAILED DESCRIPTION OF THE INVENTION:

Using an N-ethyl-N-nitrosourea- mutagenesis screen, the inventors identified a mutation in the Rltpr gene. They demonstrated that Rltpr is a lymphoid cell-specific, actin- uncapping protein essential for CD28 costimulation and regulatory T cell development. TCR- CD28 engagement at the immune synapse resulted in the colocalization of CD28 with both wild-type and mutant Rltpr forms. However, the connection between CD28 and protein kinase C-θ and Carmal, two key effectors of CD28 costimulation, was abrogated in T cells with the Rltpr mutation and CD28 costimulation did not occur. The findings provide a more complete model of CD28 costimulation in which Rltpr plays a key role. Accordingly, the present invention relates to a method for screening a plurality of test substances useful for reducing CD28 costimulation of T cells in a subject in need thereof comprising the steps consisting of al) testing each of the test substances for its ability to bind to a RLTR polypeptide and a2) positively selecting the test substances that are able to bind to the RLTR polypeptide.

As used herein the term "Rltpr" has its general meaning in the art and refers to RGD motif, leucine rich repeats, tropomodulin domain and proline-rich containing. The human exemplary amino sequence is SEQ ID NO: 1(RLTPR-001 ENST00000334583 http://www.ensembl.org/Homo_sapiens/Info/Index) and the murine exemplary amino sequence is SEQ ID NO: 2.

SEQ ID NO: l :

MAQTPDGISCELRGEITRFLWPKEVELLLKTWLPGEGAVQNHVLALLRWRAY

LLHTTCLPLRVDCTFSYLEVQAMALQETPPQVTFELESLRELVLEFPGVAALE

QLAQHVAAAIK VFPRSTLGKLFRRPTPASMLARLERSSPSESTDPCSPCGGFL

ETYEALCDYNGFPFREEIQWDVDTIYHRQGCRHFSLGDFSHLGSRDLALSVAA

LSYNLWFRCLSCVDMKLSLEVSEQILHMMSQSSHLEELVLETCSLRGDFVRRL

AQALAGHSSSGLRELSLAGNLLDDRGMTALSRHLERCPGALRRLSLAQTGLTP

RGMRALGRALATNAAFDSTLTHLDLSGNPGALGASEDSGGLYSFLSRPNVLSF LNLAGTDTALDTVRGCSVGGWMTGRADWRAGRGGLGPPAGVANSLPPQLF AAVSRGCCTSLTHLDASRNVFSRTKSRAAPAALQLFLSRARTLRHLGLAGCKL PPDALRALLDGLALNTHLRDLHLDLSACELRSAGAQVIQDLVCDAGAVSSLD LADNGFGSDMVTLVLAIGRSRSLRHVALGRNFNVRCKETLDDVLHRIVQLMQ DDDCPLQSLSVAESRLKLGASVLLRALATNPNLTALDISGNAMGDAGAKLLA KALRVNSRLRSVVWDRNHTSALGLLDVAQALEQNHSLKAMPLPLNDVAQAQ RSRPELTARAVHQIQACLLRNNRADPASSDHTTRLQPLGLVSDPSEQEVNELC QSVQEHVELLGCGAGPQGEAAVRQAEDAIQNANFSLSILPILYEAGSSPSHHW QLGQKLEGLLRQVGEVCRQDIQDFTQATLDTARSLCPQMLQGSSWREQLEGV LAGSRGLPELLPEQLLQDAFTRLRDMRLSITGTLAESIVAQALAGLSAARDQL VESLAQQATVTMPPALPAPDGGEPSLLEPGELEGLFFPEEKEEEKEKDDSPPQK WPELSHGLHLVPFIHSAAEEAEPEPELAAPGEDAEPQAGPSARGSPSPAAPGPP AGPLPRMDLPLAGQPLRHPTRARPRPRRQHHHRPPPGGPQVPPALPQEGNGLS ARVDEGVEEFFSKRLIQQDRLWAPEEDPATEGGATPVPRTLRKKLGTLFAFK PRSTRGPRTDLETSPGAAPRTRKTTFGDLLRPPTRPSRGEELGGAEGDTSSPDP AGRSRPRYTRDSKAYSMILLPAEEEATLGARPDKRRPLERGETELAPSFEQRV QVMLQRIGVSRGSGGAEGKRKQSKDGEIKKAGSDGDIMDSSTEAPPI SIKSRTHSVSADPSCRPGPGSQGPESATWKTLGQQLNAELRSRGWGQQDGPGPPSPG QSPSPCRTSPSPDSLGLPEDPCLGPRNEDGQLRPRPLSAGRRAVSVHEDQLQAPAERPL RLQRSPVLKRRPKLEAPPSPSLGSGLGTEPLPPQPTEPSSPERSPPSPATDQRGGGPNP

SEQ ID NO:2: Mouse Rltpr mRNA sequence: Accession#: HF678090 http://www.ebi.ac.uk/ena/data/view/HF678090

1 MAQT PDDI SC ELRGE I TRFL WPKEAELLLK TWLPQEGAEQ SH I LALLRWR AYLLHTCLPL

61 RVDCTFSYLE VQAMALQET P PRVTFELE SL PELVLEFPCV AALEQLAQHV AAAI KKVFPR

12 1 STLGKLFRKP T PS SLLARLE RSHPLE ST I P S S PCGGFLET YEALCDYNGF PFREE I QWDV

1 8 1 DT I YHRQGCR HFCLGDFSHF GSRDLALSVA ALSYNLWFRR LSCEDMKLSL EVSEQ I LHMT

2 4 1 SQS SYLEELV LEACGLRGDF VRRLAQALAG HFNSGLRELS LSGNLLDDRG MAALSRHLEH

3 0 1 CPGALRRLSL AQTGLT PRGM RALGRALATN ATFDSTLTHL DLSGNPGALG PSQDSGGLYT

3 61 FLSRPNVLAY LNLAGTDATL GTLFTALAGG CCS SLTHLEA SRNI FSRMKS QAAPAALQRF

42 1 LGGTRMLRHL GLAGCKLPPE ALRALLEGLA LNTQ I HDLHL DLSACELRSV GAQVI QDLVC 4 8 1 DAGALS SLDL SDNGFGSDMV TLVLAI GRSR SLKHVALGRN FNVRCKETLD DVLHRIAQLM 541 QDDDCPLQSL SVAESRLKQG ASILIRALGT NPKLTALDIS GNAIGDAGAK MLAKALRVNT

601 RLRSVIWDRN NTSALGLLDV AQALEQNHSL KSMPLPLNDV TQAHRSRPEL TTRAVHQIQA

661 CLWRNNQVDS TSDLKPCLQP LGLISDHSEQ EVNELCQSVQ EHMELLGCGA GPQGEVAVHQ

721 AEDAIQNANF SLSILPILYE AGRSPSHHWQ LQQKLESLLG QVGEICRQDI QDFTQTTLDT

781 TRSLCPQMLQ TPGWRKQLEG VLVGSGGLPE LLPEHLLQDA FSRLRDMRLS ITGTLAESIV

841 AQALAGLHAA RDRLVERLTQ QAPVTMAPAV PPLGGNELSP LETGGLEELF FPTEKEEERE

901 KDESSSWKWL EPSNCFHLVS SLHGAAEEAE RDPELAAPGE DAEPQAGPSA RGSPSPAAPG

961 PPAGPLPRMD LPPAGQPLRH PTRARPRPRR QHHHRPPPGG PQVPPALLQE GNGLTARVDE

1021 GVEEFFSKRL IQQDHFWAPE EDPATEGGAT PVPRTLRKKL GTLFAFKKPR STRGPRPDLE

1081 SPGAAARAR KSTLGDLLRP PARPGRGEEP GGAEGGTSSP DPARRNRPRY TRESKAYSMI

1141 LLPAEEEAAV GTRPDKRRPL ERGDTELAPS FEQRVQVMLQ RIGVSRASGG AESKRKQSKD

1201 GEIKKAGSDG DIMDSSTETP PISIKSRTHS VSADPSCRPG PGGQGPESAT WKTLGQQLNA

1261 ELRGRGWGQQ DGPGPPSPCP SPSPRRTSPA PDILSLPEDP CLGPRNEDGQ LRPRPLSAGR

1321 RAVSVHEDQL QAPAERPLRL QRSPVLKRRP KLEAPPSPSL GSGLGSKPLP PYPTEPSSPE

1381 RSPPSPATDQ RGGGPNP* 1397

The term "polypeptide" means herein a polymer of amino acids having no specific length. Thus, peptides, oligopeptides and proteins are included in the definition of "polypeptide" and these terms are used interchangeably throughout the specification, as well as in the claims. The term "polypeptide" does not exclude post-translational modifications that include but are not limited to phosphorylation, acetylation, glycosylation and the like. Especially, the term includes all phosphorylated forms of the polypeptide (e.g. all phosphorylated forms of Rltpr). Also encompassed by this definition of "polypeptide" are homologs thereof. Two amino acid sequences are "substantially homologous" or "substantially similar" when greater than 80 %, preferably greater than 85 %, preferably greater than 90 % of the amino acids are identical, or greater than about 90 %, preferably greater than 95 %, are similar (functionally identical). The term "sequence identity" refers to the identity between two peptides. Identity between sequences can be determined by comparing a position in each of the sequences which may be aligned for the purposes of comparison. When a position in the compared sequences is occupied by the same base or amino acid, then the sequences are identical at that position. A degree of sequence identity between nucleic acid sequences is a function of the number of identical nucleotides at positions shared by these sequences. A degree of identity between amino acid sequences is a function of the number of identical amino acid sequences that are shared between these sequences. To determine the percent identity of two amino acids sequences or two nucleic acid sequences, the sequences are aligned for optimal comparison. For example, gaps can be introduced in the sequence of a first amino add sequence or a first nucleic acid sequence for optimal alignment with the second amino acid sequence or second nucleic acid sequence. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences. In this comparison the sequences can be the same length or may be different in length. Optimal alignment of sequences for determining a comparison window may be conducted by the local homology algorithm of Smith and Waterman (J. Theor. Biol, 91 (2) pgs. 370-380 (1981), by the homology alignment algorithm of Needleman and Wunsch, J. Miol. Biol, 48(3) pgs. 443-453 (1972), by the search for similarity via the method of Pearson and Lipman, PNAS, USA, 85(5) pgs. 2444-2448 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA and TFASTA in the Wisconsin Genetics Software Package Release 7.0, Genetic Computer Group, 575, Science Drive, Madison, Wis.) or by inspection. The term "sequence similarity" means that amino acids can be modified while retaining the same function. It is known that amino acids are classified according to the nature of their side groups and some amino adds such as the basic amino acids can be interchanged for one another while their basic function is maintained. Accordingly, the term "Rltpr polypeptide" refers to the Rltpr protein or a fragment thereof that comprises the region around the amino acid residue at position 469 ins SEQ ID NO: l or the amino acid residue at 432 in SEQ ID NO:2 (both residues are in highlighted in bold). Typically a Rltpr polypeptide comprises the domain consisting of the eight leucine repeat regions (LRR) ranging from the amino acid residue at position 102 to the amino acid residue at position 677 in SEQ ID NO : 1 or ranging from the amino acid residue at position 65 to the amino acid residue at position 650 in SEQ ID NO : 2. Polypeptides of the invention may be produced by any technique known per se in the art, such as without limitation, any chemical, biological, genetic or enzymatic technique, either alone or in combination(s).

Knowing the amino acid sequence of the desired sequence, one skilled in the art can readily produce said polypeptides, by standard techniques for production of polypeptides. For instance, they can be synthesized using well-known solid phase method, preferably using a commercially available peptide synthesis apparatus (such as that made by Applied Biosystems, Foster City, California) and following the manufacturer's instructions.

Alternatively, the polypeptides of the invention can be synthesized by recombinant DNA techniques as is now well-known in the art. For example, these fragments can be obtained as DNA expression products after incorporation of DNA sequences encoding the desired (poly)peptide into expression vectors and introduction of such vectors into suitable eukaryotic or prokaryotic hosts that will express the desired polypeptide, from which they can be later isolated using well-known techniques.

A wide variety of host/expression vector combinations are employed in expressing the nucleic acids encoding for the polypeptides of the present invention. Useful expression vectors that can be used include, for example, segments of chromosomal, non-chromosomal and synthetic DNA sequences. Suitable vectors include, but are not limited to, derivatives of SV40 and pcDNA and known bacterial plasmids such as col EI, pCRl, pBR322, pMal-C2, pET, pGEX, pMB9 and derivatives thereof, plasmids such as RP4, phage DNAs such as the numerous derivatives of phage I such as NM989, as well as other phage DNA such as Ml 3 and filamentous single stranded phage DNA; yeast plasmids such as the 2 microns plasmid or derivatives of the 2 microns plasmid, as well as centomeric and integrative yeast shuttle vectors; vectors useful in eukaryotic cells such as vectors useful in insect or mammalian cells; vectors derived from combinations of plasmids and phage DNAs, such as plasmids that have been modified to employ phage DNA or the expression control sequences; and the like.

Consequently, mammalian and typically human cells, as well as bacterial, yeast, fungi, insect, nematode and plant cells an used in the present invention and may be transfected by the nucleic acid or recombinant vector as defined herein. Examples of suitable cells include, but are not limited to, VERO cells, HELA cells such as ATCC No. CCL2, CHO cell lines such as ATCC No. CCL61, COS cells such as COS-7 cells and ATCC No. CRL 1650 cells, W138, BHK, HepG2, 3T3 such as ATCC No. CRL6361, A549, PC12, K562 cells, 293T cells, Sf9 cells such as ATCC No. CRL1711 and Cvl cells such as ATCC No. CCL70. Other suitable cells that can be used in the present invention include, but are not limited to, prokaryotic host cells strains such as Escherichia coli, (e.g., strain DH5-[alpha]), Bacillus subtilis, Salmonella typhimurium, or strains of the genera of Pseudomonas, Streptomyces and Staphylococcus. Further suitable cells that can be used in the present invention include yeast cells such as those of Saccharomyces such as Saccharomyces cerevisiae.

Typically, step al) consists in generating physical values which illustrate or not the ability of said test substance to bind the Rltpr polypeptide. The "physical values" that are referred to above may be of various kinds depending of the binding assay that is performed, but notably encompass light absorbance values, radioactive signals and intensity value of fluorescence signal. The test substances may be assayed for binding to Rltpr polypeptide by method known in the art. Many different competitive binding assay format(s) can be used. For example, the binding assays include, but are not limited to, competitive assay systems using techniques such western blots, radioimmuno assays, ELISA, "sandwich" immunoassays, immunoprecipitation assays, precipitin assays, gel diffusion precipitin assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and complement-fixation assays. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994 Current Protocols in Molecular Biology, Vol. 1, John Wiley & sons, Inc., New York).

For example, the BIACORE® (GE Healthcare, Piscaataway, NJ) is one of a variety of surface plasmon resonance assay formats that are routinely used to epitope bin panels of monoclonal antibodies. Additionally, routine cross-blocking assays such as those described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane, 1988, can be performed. For example, step a) may include the use of an optical biosensor allowing the detection of interactions between molecules in real time, without the need of labelled molecules. This technique is indeed based on the surface plasmon resonance (SPR) phenomenon. Briefly, the Rltpr polypeptide may be attached to a surface (such as a carboxymethyl dextran matrix). Then the test substance is incubated with the previously immobilised polypeptide. Then the binding including the binding level, or the absence of binding is detected. For this purpose, a light beam is directed towards the side of the surface area where the polypeptide is attached and is reflected by said surface. The SPR phenomenon causes a decrease in the intensity of the reflected light with a combination of angle and wavelength. The binding of the test substance to the polypeptide causes a change in the refraction index on the substrate surface, which change is detected as a change in the SPR signal. In another one embodiment of the invention, the screening method includes the use of affinity chromatography.

In some embodiment, the screening method of the invention further comprise the steps consisting of bl) testing each of the substances positively selected at step a2) for its ability to reduce the CD28 costimulation signalling in a T cell and b2) positively selecting the test substances that are able to reduce the CD28 costimulation in a T cell.

Typically, step bl) may be performed by any in vitro or in vivo assay that the skilled man in the art can settle for testing the ability of the test substance to reduce the CD28 costimulation signalling in a T cell.

For example, the proliferation of a T cell after CD28-TCR co-stimulation may be assayed in the presence of the test substance. Typically, the skilled man in the art may used the method described in the EXAMPLE. Briefly, T cells are activated with a combination of anti-CD3 antibodies and anti-CD28 antibodies in the presence of the test substance. The proliferation level is then determined and is compared to the level determined in the absence of the test substance. It is concluded that the test substance reduce the CD28 costimulation signalling when the proliferation level determined in the presence of the test substance is lower that the proliferation level determined in the absence of the test substance.

Altenatively, the screening method may also comprise a step consisting of determining the ability of the test substance to prevent CD28 synergizing with the TCR in assays measuring IL-2 production and IFN-γ production by primary T cells. It is concluded that the test substance is able to inhibit the CD28 co-stimulation signal when the IL-2 production and/or the IFN-γ production determined in the presence of the test substance is lower when the IL-2 production and/or the IFN-γ production determined in the absence of the test substance. In some embodiments, the screening method of the invention may further comprise a step consisting of providing an animal (e.g. a mouse) having T cells expressing a TCR specific for an antigen, administering the animal with an amount of antigen and with an amount of the test substance, determining the production of IL-2 and/or of IFN-γ by the T cells expressing the TCR specific for the antigen, comparing production of IL-2 and/or of IFN-γ determined at the preceding step with the production determined in an animal that was not administered with the test substance and positively selecting the test substance when the production of IL-2 and/or of IFN-γ determined when the animal was administered with the test substance is lower than the production of IL-2 and/or of IFN-γ determined when the animal was not administered with the test substance. Typically, OT-I mice expressing a TCR specific for the OVA257-264 peptide bound to H-2K as described in the EXAMPLE may be used.

In some embodiment, the test substance may be evaluated for its ability to induce the functional defects as observed for T cells deficient for CD28 or for T cells harbouring a Rltpr loss of function mutation such as observed for Rltpr BAS mouse. For example, the proliferation level determined in the presence of the test substance may be compared with the proliferation level of purified T cells from Rltpr Bas or Cd28-/- mice as described in the EXAMPLE. Then the test substance may be selected when the proliferation level determined in the presence of the test substance is above the same as the proliferation level determined for the purified T cells from Rltpr Bas or Cd28-/- mice.

In some embodiments, the screening method may further comprises the steps consisting of administering an animal (e.g. a mouse) with an amount of the test substance, then determining the frequency of thymic natural Treg cells (i.e. Foxp3+ Treg cells) in the animal, comparing the frequency with the frequency of thymic natural Tregs determined in an animal that was not administered with the test substance and positively selecting the test substance when the frequency determined in the animal administered with the test substance is lower than the frequency determined in the animal that was not administered with the test substance.

In some embodiments, the screening method may further comprises the steps consisting of administering an animal (e.g. a mouse) with an amount of the test substance, then determining the frequency of thymic natural Treg cells (i.e. Foxp3+ Treg cells) in the animal, comparing the frequency with the frequency of thymic natural Tregs determined in a CD28 deficient animal or in a Rltpr Bas animal and positively selecting the test substance when the frequency determined in the animal administered with the test substance above the same as determined for the CD28 deficient animal or for the Rltpr Bas animal. In some embodiments, the screening method may further comprises the steps consisting of administering an animal (e.g. a mouse) with an amount of the test substance, then determining the frequency of TFH cells (e.g. CXCR5+PD1+ TFH cells) in the animal, comparing the frequency with the frequency of TFH cells determined in an animal that was not administered with the test substance and positively selecting the test substance when the frequency determined in the animal administered with the test substance is lower than the frequency determined in the animal that was not administered with the test substance.

In some embodiments, the screening method may further comprises the steps consisting of administering an animal (e.g. a mouse) with an amount of the test substance, then determining the frequency of TFH cells (e.g. CXCR5+PD1+ TFH cells) in the animal, comparing the frequency with the frequency of TFH cells determined in a CD28 deficient animal or in a Rltpr Bas animal and positively selecting the test substance when the frequency determined in the animal administered with the test substance above the same as determined for the CD28 deficient animal or for the Rltpr Bas animal.

In some embodiment, the screening method of the invention further comprises a step consisting of determining the ability of the test substance to inhibit the recruitment of PKC-Θ and Carmal at the T cell immunological synapse. Said recruitment may be assayed by any method known in the art. For example, the assay described in the EXAMPLE may be performed. Briefly, CD4+ T cells expressing PKC-9-EGFP or Carmal-EGFP are imaged using planar bilayers containing I-Ek-MCC88-103, ICAM-1 and CD80 in presence of the test substance. Then the accumulation of either PKC-Θ or Carmal at the cSMAC is then evaluated and it is concluded that the test substance is able to inhibit the recruitment of PKC-Θ and Carmal at the T cell immunological synapse when no accumulation is determined.

The test substances that have been positively selected at the end of any one of the embodiments screening method which has been described previously in the present specification may be subjected to further selection steps in view of further assaying their in vivo properties on animal model for a disease of interest or eventually in human in the context of clinical trials. Indeed the test substances that may be selected at the end of any one of the embodiments of the in vitro screening may find various applications.

For example, the test substances may constitute drug candidate for transplantation. For example, the selected test substance may be assayed for their ability to prevent or suppress immune responses associated with rejection of a donor tissue, cell, graft, or organ transplant by a recipient subject. Graft-related diseases or disorders include graft versus host disease (GVDH), such as associated with bone marrow transplantation, and immune disorders resulting from or associated with rejection of organ, tissue, or cell graft transplantation (e.g., tissue or cell allografts or xenografts), including, e.g., grafts of skin, muscle, neurons, islets, organs, parenchymal cells of the liver, etc. With regard to a donor tissue, cell, graft or solid organ transplant in a recipient subject, it is believed that the selected substances may be assayed to prevent acute rejection of such transplant in the recipient and/or for long-term maintenance therapy to prevent rejection of such transplant in the recipient (e.g., inhibiting rejection of insulin-producing islet cell transplant from a donor in the subject recipient suffering from diabetes). Thus the selected test substance may be useful for preventing Host- Versus-Graft-Disease (HVGD) and Graft- Versus-Host-Disease (GVHD). The CTPS1 inhibitor may be administered to the subject before and/or after transplantation (e.g., at least one day before transplantation, from one to five days after transplantation, etc.). In some embodiments, the CTPS1 inhibitor may be administered to the subject on a periodic basis before and/or after transplantation.

The test substances may also constitute drug candidate for the treatment of autoimmune diseases. As used herein, an "autoimmune disease" is a disease or disorder arising from and directed at an individual's own tissues. Examples of autoimmune diseases include, but are not limited to Addison's Disease, Allergy, Alopecia Areata, Alzheimer's disease, Antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis, Ankylosing Spondylitis, Antiphospho lipid Syndrome (Hughes Syndrome), arthritis, Asthma, Atherosclerosis, Atherosclerotic plaque, autoimmune disease (e.g., lupus, RA, MS, Graves' disease, etc.), Autoimmune Hemolytic Anemia, Autoimmune Hepatitis, Autoimmune inner ear disease, Autoimmune Lymphoproliferative syndrome, Autoimmune Myocarditis, Autoimmune Oophoritis, Autoimmune Orchitis, Azoospermia, Behcet's Disease, Berger's Disease, Bullous Pemphigoid, Cardiomyopathy, Cardiovascular disease, Celiac Sprue/Coeliac disease, Chronic Fatigue Immune Dysfunction Syndrome (CFIDS), Chronic idiopathic polyneuritis, Chronic Inflammatory Demyelinating, Polyradicalneuropathy (CIPD), Chronic relapsing polyneuropathy (Guillain-Barre syndrome), Churg-Strauss Syndrome (CSS), Cicatricial Pemphigoid, Cold Agglutinin Disease (CAD), COPD, CREST syndrome, Crohn's disease, Dermatitis, Herpetiformus, Dermatomyositis, diabetes, Discoid Lupus, Eczema, Epidermolysis bullosa acquisita, Essential Mixed Cryoglobulinemia, Evan's Syndrome, Exopthalmos, Fibromyalgia, Goodpasture's Syndrome, Hashimoto's Thyroiditis, Idiopathic Pulmonary Fibrosis, Idiopathic Thrombocytopenia Purpura (ITP), IgA Nephropathy, immunoproliferative disease or disorder (e.g., psoriasis), Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, Insulin Dependent Diabetes Mellitus (IDDM), Interstitial lung disease, juvenile diabetes, Juvenile Arthritis, juvenile idiopathic arthritis (JIA), Kawasaki's Disease, Lambert-Eaton Myasthenic Syndrome, Lichen Planus, lupus, Lupus Nephritis, Lymphoscytic Lypophisitis, Meniere's Disease, Miller Fish Syndrome/acute disseminated encephalo myeloradiculopathy, Mixed Connective Tissue Disease, Multiple Sclerosis (MS), muscular rheumatism, Myalgic encephalomyelitis (ME), Myasthenia Gravis, Ocular Inflammation, Pemphigus Foliaceus, Pemphigus Vulgaris, Pernicious Anaemia, Polyarteritis Nodosa, Polychondritis, Polyglandular Syndromes (Whitaker's syndrome), Polymyalgia Rheumatica, Polymyositis, Primary Agammaglobulinemia, Primary Biliary Cirrhosis/Autoimmune cholangiopathy, Psoriasis, Psoriatic arthritis, Raynaud's Phenomenon, Reiter's Syndrome/Reactive arthritis, Restenosis, Rheumatic Fever, rheumatic disease, Rheumatoid Arthritis, Sarcoidosis, Schmidt's syndrome, Scleroderma, Sjorgen's Syndrome, Stiff-Man Syndrome, Systemic Lupus Erythematosus (SLE), systemic scleroderma, Takayasu Arteritis, Temporal Arteritis/Giant Cell Arteritis, Thyroiditis, Type 1 diabetes, Type 2 diabetes, Ulcerative colitis, Uveitis, Vasculitis, Vitiligo, and Wegener's Granulomatosis. The test substance of may be selected from the group consisting of peptides, peptidomimetics, small organic molecules, or nucleic acids. For example the test substance according to the invention may be selected from a library of compounds previously synthetized, or a library of compounds for which the structure is determined in a database, or from a library of compounds that have been synthetized de novo. In a particular embodiment, the test substance may be selected form small organic molecules. As used herein, the term "small organic molecule" refers to a molecule of size comparable to those organic molecules generally sued in pharmaceuticals. The term excludes biological macromolecules (e.g.; proteins, nucleic acids, etc.); preferred small organic molecules range in size up to 2000da, and most preferably up to about 1000 Da.

The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention. EXAMPLE:

Material & Methods

Mice. To facilitate the mapping of mutations abrogating the Lat Y136F lymphoproliferative disorder, mice homozygous for a Lat Y136F mutant allele were constructed on a pure B6 background using Bruce 4 embryonic stem cells. They showed the same phenotype as that of the Lat Y136F mice that were previously established on a 129 background (data not shown).

5C.C7, AND and OT-I TCR transgenic mice, Cd28^ , Cd3e 5/ 5 and Cd3e 5/ 5 x

/ / 17 27 29 51 52

Cd8(F ~ Cd&6— mice have been described 1 '' J1 ' . Mice were maintained in specific pathogen- free conditions and all experiments were done in accordance with French and European guidelines for animal care. ENU mutagenesis and screening of mutant mice. The ENU mutagenesis screen was performed in a B6. Lat Y136F background as previously described 53 . Mutant mice were identified using blood samples collected from the retro-orbital sinus of sedated mice using heparinized microhematocrit tubes. Staining was performed on 30 μΐ of whole blood using Trucount tubes (BD Biosciences) and a combination of 8 antibodies consisting of PerCP- Cy5.5 conjugated anti-CD45.2 (104), APC conjugated anti-CD5 (PK136), PE-Cy7 conjugated anti-CD 19 (1D3/6D5), Pacific Blue conjugated anti-CD4 (RM4-5), Alexa-700 conjugated anti-CD8a (53.6.7), PE conjugated anti-CD62L (MEL-14), FITC conjugated CD44 (IM7) and Alexa-700 conjugated anti-IA/IE (M5/114.15.2) all from BD Biosciences. A lyse/no- wash procedure was performed using a BD FACS lysing solution (BD Biosciences). For each sample, a minimum of 30 000 CD45.2 + cells were analyzed. The absolute number of CD19 + B cells and of CD4 + and CD8 T cells per μΐ of blood was calculated using the formula: cell counts/bead counts x total Trucount beads/μΐ of blood.

Targeted sequencing and identification of genetic variation. The genetic variations induced by ENU-mutagenesis in the Basilic Lat Y136F mice were analyzed using targeted next- generation sequencing 54 , adapted for a SOLiD™ 4 sequencer. Briefly, short fragments (-150 nucleotide-long) from paired-end libraries were prepared using focussed acoustic fragmentation (Covaris TM S2) and according to the preparation guide from Life Technologies (cms_081748.pdf). Two custom-designed AgilentTM SurePrint G3 Mouse DNA Capture lxlM Arrays were designed to cover the targeted 16 Mb region on chromosome 8 and used to perform the library enrichment according to AgilentTM protocol #G4458-90000. Templated beads were prepared using the SOLiD™ EZ Bead™ System (EZ Bead E80) and sequenced according to standard Applied Biosystems Life Technologies protocols. The sequence reads were analyzed with the BioScope™ software suite (cms_4448431.pdf). SNPs and Small InDels were identified using the Genome Variant Analyzer pipeline developed at TGML Sequencing Platform (GeVarA). Flow cytometry. Stained cells were analyzed using a BD LSR II system (BD

Biosciences). Data were analyzed with Flow Jo software (Tree Star) or FCS Express™ 4, Diva. Cell viability was evaluated using SYTOX Blue (Life Technologies). Antibodies used were directed to CD5 (53-7.3), CD4 (RM4-5), CD8a (53-6.7), CD69 (H1.2F3), CD24 (Ml/69), TCR (H57-597), CD28 (37.51) and CXCR5 (2G8) all from BD Biosciences; to CD25 (PC61.5), CD279 (PD1, 29F.1A12) all from BioLegend and to GITR (DTA-1), CD 122 (TM- Bl) and FoxP3 (FJK-16s) all from eBioscience. CellTrace™ CFSE proliferation kit was purchased from Molecular Probes™.

Plasmids and cell transfection. Full-length cDNA encoding Rltpr and Rltpr as were appended at their 3 ' end to sequences coding for a One-Strep-Tag (OST) or a Yellow Fluorescent Protein for Energy Transfer (YPET) reporter. The Rltpr-OST and Rltpr Ba -OST cDNA were cloned into the pMX-IRES-eGFP retroviral expression vector 55 , whereas the Rltpr-YPET and Rltpr 8 ™ '-YPET were cloned into the pMSCV-IRES-hCD2t retroviral expression vector 56 . PLAT-E cells were used to produce retroviral particles 57 . 48 h after transfection, viral supernatants were harvested and used to transduce T cell hybridoma or briefly activated CD4 T cells from AND or 5C.C7 mice.

T cell isolation and stimulation. CD5 , CD4 or CD8 T cells were purified from pooled lymph nodes and spleens of WT, Rltpr Bas and Cd28 ~ ' ~ mice using mouse depletion Dynabeads untouched kits (Life Technologies) with a cell purity of over 90%. Purified cells were stimulated with plate-bound anti-CD3 antibody (145-2C11) with or without soluble anti- CD28 antibody (37-51) for 72 h. Alternatively, phorbol ester (PMA 10 ng/ml, Sigma- Aldrich) plus ionomycine (500 ng/ml, Sigma-Aldrich) were used to stimulate purified cells. Supernatants were collected at the indicated times and IL-2 and IFNy concentration were measured using BD™ Cytometric Bead Array or Milliplex Map mouse kit (Merck Millipore). Generation of antibodies against Rltpr. Polyclonal antisera against Rltpr were generated by immunizing rabbits with a glutathione S-transferase to which was appended a segment of Rltpr corresponding to aminocids 1147-1397.

Western blot and immunoblot analyses. Thymocytes and peripheral T cells were lysed by addition of lysis buffer (100 mM Tris pH 7.5, 274 mM NaCl, 1 mM EDTA, 10% glycerol, 0.2% n-Dodecyl- -Maltoside). Postnuclear supernatants were separated on 8% SDS- acrylamide gels, transferred to membrane and subjected to immunoblot analysis using antibodies against Rltpr (see above) or SLP-76 (4958, Cell Signaling). Membranes were incubated with anti-rabbit IgG (CF770, Biotium) and analyzed with an Odyssey Imaging System (LI-COR). The BI-141 T cell hybridoma 58 was retroinfected with an empty virus or with viruses expressing Rltpr-OST or Rltpr Bas -OST. Cell lysates were immunoprecipitated with Strep-Tactin sepharose beads (IBA GmbH). Beads were eluted with D-Biotin (Sigma- Aldrich). Eluates were separated on 8% SDS-acrylamide gels and subjected to immunoblot analysis using polyclonal rabbit antibodies specific for Rltpr and for the alpha subunit of CP (AB6016; Millipore).

Imaging at T cell-APC interfaces. Image acquisition and analysis were performed as described 38 . Briefly, T cell-APC interactions were imaged at 37°C. Every 20 seconds, 1 DIC and 21 fluorescence images that spanned 20 μιη in the z-plane at 1 μιη intervals were acquired. The acquisition and analysis software was Metamorph (Molecular Devices). The formation of a tight cell couple, time 0 in our analysis, was defined as either the first time point with a fully spread T cell-APC interface or 40 s after first membrane contact, whichever occurred first. A region of sensor accumulation was defined by an average fluorescence intensity of >135% of the background cellular fluorescence. To classify spatial accumulation features, six mutually exclusive interface patterns were used as defined by strict geometrical constraints 38 . Imaging at T cell-planar bilayer interface. Planar bilayers construction, confocal

9, 59 microscopy imaging and image processing and data analysis have been previously defined

CD28 internalization assay. Cells were incubated for 30 min on ice with anti-CD28 antibody (1 Mg/ml) followed by biotinylated goat anti-mouse antibody (2 g/ml) for another 30 min on ice. Cells then were washed and warmed to 37°C for 5, 10, or 15 min to allow internalization and then treated with 0.1% NaN 3 on ice. Cells were stained with PE- conjugated streptavidin, washed, and subjected to flow cytometry analysis (FACSCalibur; Becton Dickinson). The data were expressed as the percentage change in surface level CD28 over the time course.

Statistical analysis. The unpaired Student t test was used for statistical analyses with GraphPad Prism software. The p values were calculated by t test: *p, <0.05, **p, <0.01, ***/?,<0.001 and <0.0001. All data are presented as the arithmetic mean ± SEM or SD as indicated.

Results

The Basilic Lat Y 6F phenotype

We conducted an ENU- induced mutagenesis screen on C57BL/6J (B6) mice homozygous for the Lat Y136F mutation to identify mutations capable of abrogating their lymphoproliferative disorder. Compared to wild-type (WT) mice, the blood of Lat Yi36F mice contains large numbers of abnormal CD4 + T cells that show a CD44 high CD62L low T H 2 effector phenotype, and normal numbers of B cells that show an activated phenotype resulting from the action of the T R 2 effectors. By screening the blood of Lat Yi36F G3 offspring, we identified a mouse denoted Basilic Lat Yi36F , the blood of which contained 500- fold fewer CD44 high CD62L low CD4 + T cells. The B cells found in the blood of the Basilic Lat Y136F mouse had a resting phenotype, an observation consistent with the near absence of T H 2 effectors. A similar phenotype was also observed in the spleen. Therefore, the Basilic mutation abrogated the expansion of pathogenic Lat Y136F CD4 + T cells and the ensuing exaggerated B cell activation.

Characterization of the Basilic mutation The Basilic mutation was fixed in the Lat background and inherited as a fully penetrant recessive trait. To map it, we screened F2 mice from Basilic Lat Yi36F B6 mice x C3HeB/FeJ (C3H) intercrosses. 12 homozygous Lat Yi36F mice with a Basilic phenotype and 10 homozygous Zat Y136F mice without a Basilic phenotype were genotyped with a panel of 64 simple sequence length polymorphism (SSLP) markers corresponding to 2 to 6 SSLP markers per chromosome. This analysis revealed a linkage to chromosome 8 with the highest lod score of 3.97 associated with a marker (D8mit242) corresponding to position 50,07 cM . High-resolution haplotype mapping revealed that the Basilic mutation was localized in a 16-megabase (Mb) interval of chromosome 8. These 16 Mb of DNA were captured from Basilic Lat Yi36F mice using specially designed oligonucleotide arrays and then sequenced by next-generation sequencing. After comparison with a B6 reference sequence 3 single nucleotide polymorphism (SNP) were located within the coding regions of 3 distinct genes. Further analysis, excluded two of the three candidate genes and left us with a single coding region-associated SNP that involved the RGD, leucine-rich repeat, tropomodulin and proline- rich containing (Rltpr) gene. Rltpr is also known as leucine-rich repeat (LRR) containing 16c (Lrrcl6c).

Sequencing a full-length Rltpr cDNA isolated from B6 thymocytes and determining the exon-intron structure of the B6 Rltpr gene led us to re-annotate the available mouse sequence (ENSMUSG00000050357; http://mouse.ensembl.org/). The Rltpr gene is composed of 39 exons and codes for a 1397 amino acid protein. Consistent with this analysis, immunoblots of thymic and peripheral T cells with Rltpr-specific rabbit polyclonal antibodies detected a single protein species of approximately 150 kilodaltons. The mutation identified in the Rltpr gene of Basilic Lat Y 6F mice is denoted as C57BL/6-i?/t/?r Bas (Rltpr 3*8 in short). It affected exon 16 and corresponded to a T to C nucleotide transition at position 108,213,695.

Rltpr Bas is a partial loss of function mutation

The mouse genome contains two Rltpr paralogs that are denoted as Lrrcl6a and Lrrl6b 20 . Orthologs of the Lrcc gene family can be found in vertebrates and invertebrates (Ensembl GeneTree ENSGT00390000014487). The vertebrate Lrcc gene family codes for cytosolic proteins that comprise concatenated LRRs, a capping protein (CP) interaction (CPI) motif and a region rich in proline residues. In its free form, CP has an affinity of 0.1-1 nM for the barbed ends of actin filaments. Upon binding to the CPI motif, the affinity of CP for actin filaments decreases by approximately 100-fold. It has thus been inferred that the whole vertebrate Lrcc gene family codes for actin-uncapping proteins that uncap actin filaments pre- capped by CP 21"23 .

The eighth LRR found in Rltpr comprises the 12-residue long LRR consensus sequence 24 . The non conservative L432P substitution found in Rltpr Bas likely disrupted the structure of the eighth LRR. This may account for the 1.5- and 2.5-fold decreased levels of Rltpr Bas observed in thymocytes and peripheral T cells, respectively, as compared to Rltpr. To verify the prediction that Rltpr is capable of constitutively associating with CP, Rltpr was fused to a One-Strep-Tag (OST) 25 and expressed in a T cell hybridoma. Pull-down experiments demonstrated that Rltpr constitutively associated with CP. Importantly, an Rltpr Bas -OST fusion protein was also capable of associating with CP. Therefore, the Rltpr Bas mutation preserved the actin- uncapping function of Rltpr and thus corresponded to a partial loss of function mutation of the Rltpr gene. Rltpr expression

We analyzed Rltpr expression in thymus, spleen, bone marrow (BM), brain and testis by quantitative RT-PCR analysis. Rltpr was expressed in large amounts in adult thymus and spleen, smaller amounts in BM and was undetectable in brain and testis. The Rltpr Bas mutation was without effect on the pattern and levels of transcription (data not shown). The BioGPS gene expression atlas database (http://biogps.org/) confirmed that Rltpr expression was restricted to lymphoid tissues. In contrast, the Lrrcl6a and Lrrcl6b mouse paralogs were predominantly expressed innon-lymphoid tissues (http://biogps.org .

Quantitative RT-PCR analysis of thymocyte subsets showed that Rltpr expression was upregulated in preselection CD69 DP cells and down-regulated in post-positive selection CD69 + DP cells and in CD4 + and CD8 + single positive thymocytes. Peripheral CD4 + and CD8 + T cells also expressed substantial amounts of Rltpr, a pattern consistent with the Immgen database (www.immgen.org and immunoblot analysis. Therefore, Rltpr is an actin-uncapping protein that is predominantly expressed in T cells.

Segregating Rltpr from La? libt mimicks a CD28 deficiency

Mice with a compound Rltpr Bas Lat Y136F mutation showed a phenotype highly reminiscent of that of Lat Y136F mice deprived of CD80 and CD86 ligands 17 . This led us to test whether Rltpr belonged to the signaling pathway responsible for CD28 costimulation. For that purpose, we segregated the Rltpr as mutation from the Lat mutation and determined whether mice homozygous for the sole Rltpr Bas mutation - called Rltpr Bas mice - functionally mimicked a CD28 deficiency. Akin to CD28-deficient (Cd28 ~ ' ~ ) mice, Rltpr Bas mice showed a normal sequence of T cell development and their periphery was populated with CD4 + and CD8 + T cells that showed a normal phenotype. No measurable effect on negative and positive selection was observed when OT-I, H-Y, OT-II and AND TCR transgenes were bred onto Rltpr Bas . Importantly, the Rltpr Bas mutation was without effect on the expression of CD28 in peripheral T cells. Moreover the decrease in CD28 expression that is normally associated with positive selection in the thymus was fully recapitulated in Rltpr Bas mice. Therefore, Rltpr Bas did not affect the expression of CD28.

To compare the importance of Rltpr and CD28 during T cell activation, negatively purified T cells from Rltpr Bas , Cd28^ ~ , and WT control mice were stimulated with graded doses of anti-CD3 antibody in the presence or absence of a fixed dose of anti-CD28 antibody. Rltpr Bas prevented CD28 synergizing with TCR-CD3 to the same extent as a lack of CD28. Both CD4 + and CD8 + T cells isolated from Rltpr B and Cd28^ mice and labeled for CFSE showed a reduced proliferation in response to anti-CD3 plus anti-CD28 antibody stimulation. The Rltpr Bas mutation also prevented CD28 synergizing with the TCR in assays measuring IL-2 production and IFN-γ production by primary T cells.

Similar to CD28-deficient CD4 + T cells 26 , addition of exogenous IL-2 overcame the inefficient proliferation manifested by T cells purified from Rltpr Bas mice in response to CD3 plus CD28 stimulation. As observed for CD28-deficient T cells 27 , T cells purified from Rltpr Ba mice also showed a dramatically reduced proliferation in mitogen (concanavalin A) responses.

To further support the view that Rltpr as and Cd28^ mice showed similar T cell functional defects, the Rltpr Bas allele was backcrossed onto OT-I mice expressing a TCR specific for the OVA257-264 peptide bound to H-2K b 28 ' 29 . OVA257-264 was capable of inducing OT-I Rltpr Bas CD8 + T cell proliferation to the same extent as OT-I Rltpr CD8 + T cells. In contrast, the presence of Rltpr Ba dramatically reduced the production of IL-2 and of IFN-γ by OT-I CD8 + T cells and the proliferation induced by the Q4 and T4 weak agonist variants of OVA257-264. Importantly, the Rltpr Ba mutation had an impact on OT-I proliferation similar to that resulting from the use of CD80/CD86-deficient APCs. Therefore, side-by-side comparison of Rltpr Bas and of Cd28 ~ ' ~ mice showed that Rltpr Bas by itself constituted a functional phenocopy of a Q 28 null mutation, suggesting that Rltpr constitutes a previously unrecognized and essential component of the CD28 signaling pathway.

Defective development of natural T REG cells and of T FH cells in Rltpr as mice

CD28 has an important cell-intrinsic role in the generation of thymic nTreg cells.

Cd28 ~ ' ~ mice or mice lacking both CD80 and CD86 have an approximately 80% reduction in the frequency of thymic nTreg cells with no changes in conventional T cell subsets 30"32 . Rltpr Bas mice showed a reduction in thymic nTreg cells as severe as Cd28 ~ ' ~ mice and this reduction was not associated with changes in other intrathymic T cell subsets. As a result, the spleens of Rltpr Bas and Cd28 ~ ' ~ mice contained approximately 5-fold reduced numbers of Foxp3 + T reg cells.

nTreg cells develop in the thymus in a two-step process. The first step is governed by TCR and CD28 signals and involves the differentiation of CD4 + single positive thymocytes into CD4 + CD25 + CD122 + GITR + Foxp3 nTreg cell progenitors. In the second step, upon stimulation via IL-2, these progenitors convert into CD4 CD25 CD122 + GITR Foxp3 + nTreg cells. CD28 is involved in the first stage of Treg differentiation 32 . Analysis of Rltpr Bas thymi revealed a similar reduction in the numbers of nTreg cell progenitors. T FH cells constitute a CD4 + T H lineage specialized in provision of help to B cells. CD28 costimulation has also been shown to be essential for T FH cell development 33 . Analysis of the spleen of Rltpr Bas mice, showed a reduction in CXCR5 PD1 + T FH cells that was similar to that of Cd28 ~ ' ~ mice. Therefore, analysis of the development of nT reg cells and of T FH cells further indicated that i?V Bas and Cd28 ~ '- mice showed the same T cell functional defects.

Rltpr and Rltpr Bas MCs colocalize with CD28 MCs

To determine the mechanism by which Rltpr Bas abrogates CD28-mediated costimulatory signals, we assessed the dynamic movement of Rltpr and Rltpr Bas at the IS with a high spatial resolution. For this purpose, we used glass-supported planar bilayers containing mobile ligands as APC substitutes. We first determined whether the Rltpr Bas mutation affected the generation of TCR and of CD28 MCs and their subsequent translocation to the cSMAC. The Rltpr as allele was first backcrossed onto AND mice that express a TCR specific for I-E k molecules bound to the MCC 8 8-i o3 peptide 29 . AND Rltpr and AND Rltpr Bas CD4 + T cells were then subjected to retroviral infection to express a EGFP-tagged CD28 reporter 9 . AND Rltpr CD28-EGFP and AND Rltpr Bas CD28-EGFP CD4 + T cells were allowed to settle on planar bilayer membranes containing glycophosphatidylinostitol (GPI) anchored I-E k molecules loaded with MCC 88 -io3, ICAM-1-GPI and CD80-GPI molecules. The movement of the AND TCR (tagged with a DyLight 649-labelled anti-TCR Fab) and of CD28-EGFP was imaged after T cell-planar bilayer contact by confocal microscopy. Rltpr Bas had no measurable effect on the generation of TCR and CD28 MCs and on their subsequent translocation to the cSMAC.

To visualize the dynamics of Rltpr and of Rltpr Bas following TCR-CD28 mediated T cell activation and determine whether such dynamics depended on CD28-CD80 engagement, full-length Rltpr and Rltpr Bas proteins were fused to a Yellow Fluorescent Protein for Energy Transfer (YPET) reporter 34 and separately expressed in CD4 + AND T cells. AND Rltpr- YPET and AND Rltpr Bas -YPET CD4 + T cells expressing similar reporter levels were allowed to settle on planar bilayers. As previously observed for TCR-CD3 35 ' 36 and CD28 9 , Rltpr- YPET and Rltpr Bas -YPET MCs were generated at the T cell-bilayer interface as soon as a T cell attached and they subsequently translocated to the cSMAC. Rltpr- YPET and Rltpr Bas - YPET MCs constantly colocalized with CD28 MCs. Upon translocation to the cSMAC they segregated away from TCR MCs to occupy the CD28 hi CD3 l0 signaling cSMAC. Therefore, both Rltpr and Rltpr Bas generated clusters when T cells attached to the planar bilayer. Importantly, both Rltpr and Rltpr Bas MCs colocalized with CD28 MCs, suggesting that the lack of CD28-mediated costimulatory signals observed in the presence of Rltpr Bas was not due to the inability of Rltpr Bas to colocalize with CD28 MCs.

Rltpr and Rltpr Bas MCs form in a CD80 dependent manner

CD28 MC formation is completely dependent on binding of CD28 to CD80 9 . Likewise, the formation of Rltpr- YPET and Rltpr Bas -YPET MCs only occurred when the planar bilayer membrane contained CD80 molecules. CD28, Rltpr and Rltpr Bas MC formation depended on the presence of CD80 and followed a similar spatiotemporal distribution at the IS. Rltpr and Rltpr Bas are thus likely part of the CD28 "signalosome". However, Rltpr and Rltpr Bas were not coprecipitated with CD28 in lysates from CD4 + T cells following stimulation with TCR plus CD28 antibodies or PMA, suggesting that the CD28- Rltpr colocalization observed at the IS resulted from an indirect association between CD28 and Rltpr. Rltpr is essential for CD28-induced PKC-Θ and CARMA1 translocation to the cSMAC Engagement of the TCR results in the generation of diacylglycerol (DAG) and suffices to promote transient and diffuse PKC-Θ recruitment at the T cell-APC interface 9 . However, the formation of discrete PKC-Θ MCs and their subsequent translocation to the cSMAC depends on CD28-CD80 binding 9 , and a similar trend has been observed for Carmal 12 ' 37 . Considering that CD28 colocalized with both Rltpr and Rltpr Bas and that CD28 costimulation was totally blocked in the presence of Rltpr Bas , we analyzed whether Rltpr Bas abrogated the capacity of CD28 to recruit PKC-Θ and Carmal at the IS. Accordingly, AND Rltpr and AND Rltpr Bas CD4 + T cells expressing PKC-9-EGFP or Carmal -EGFP were imaged using planar bilayers containing I-E k -MCC88 io3, ICAM-1 and CD80. In contrast to Rltpr, in the presence of Rltpr Bas there was no accumulation of either PKC-Θ or Carmal at the cSMAC. Therefore, although Rltpr and Rltpr Bas MCs were both capable of forming and of translocating to the cSMAC upon TCR stimulation and in a manner dependent on CD28-CD80 binding, the presence of Rltpr Bas prevented the accumulation of PKC-Θ and Carmal at the cSMAC. These results suggest that Rltpr plays an essential role in CD28-mediated costimulation based on its capacity to couple CD28 to PKC-Θ and Carmal .

Spatiotemporal distribution of Rltpr and Rltpr as at T cell-APC interface

Subcellular proximity during intracellular signaling is often indicative of functional connections. Therefore, we analyzed the spatiotemporal distribution of Rltpr and Rltpr Bas in live T cell-APC conjugates and compared it to that of 53 transmembrane and cytoplasmic molecules involved in T cell activation 38 . Rltpr- YPET and Rltpr Bas -YPET CD4 + T cells were separately expressed in primary 5C.C7 CD4 + T cells that have a TCR specific for a moth cytochrome c peptide (MCC82 103) and H-2 I-E k . Upon tight conjugate-formation with MCC- pulsed mouse CH27 B lymphoma cells areas of Rltpr- YPET and Rltpr Bas -YPET enrichment were determined in three dimensions up to 7 minutes after conjugate formation and classified at 12 time points according to six subcellular spatial patterns (38).

Rltpr and Rltpr Bas were unusual among the molecules analyzed to date in that they straddled four different subcellular distributions, suggestive of a protein linking various subcellular processes. First, in a sizable fraction of T cell-APC couples, Rltpr accumulated at the center of the T cell-APC interface in a manner similar to CD28. This accumulation was rapid and well sustained. In contrast, the central accumulation of Rltpr Bas was often delayed and more transient. Second, in a substantial fraction of conjugated T cells, both Rltpr and Rltpr Bas accumulated according to a 'lamellal' pattern, a F-actin-based structure that supports the activity of proteins linking proximal signaling and actin dynamics (K.T. Roybal and C.W., submitted). Third, most of the central accumulation of Rltpr Bas - but not of Rltpr - occurred in a large invagination forming at the center of the interface within 1 minute of contact and thought to recycle proximal TCR signaling components through receptor internalization 39 . Fourth, Rltpr and Rltpr Bas often displayed an internal punctuate accumulation, a pattern that is probably associated with intracellular vesicles and has only been observed equally prominently with TCR complexes in which the clusters of arginine and lysine residues found in the CD3^ subunit were mutated 40 . This internal punctuate accumulation is suggestive of a role of Rltpr in CD28 internalization. Such internal punctuate structures occurred in 93 ± 5 % of cell couples involving T cells expressing Rltpr Bas - YPET, as compared to 58 ± 10 % of those involving T cell expressing Rltpr- YPET (p< 0.001).

The proteins that showed the closest spatiotemporal distribution to Rltpr were Rpre, a sensor for the negatively charged polyphosphoinositides found at the cytosolic surface of the plasma membrane and of intracellular vesicles, and the adaptors Nek, Grb2, and Cin85. Both Nek and Cin85 have been shown to link antigen receptors to actin regulation and receptor internalization 41 . Therefore, these studies suggest that, in addition to its capacity to link CD28 to PKC9 and Carmal, Rltpr additionally controls CD28 internalization by coupling it to actin polymerization. CD28 has been shown to be rapidly internalized after ligation 42 ' 43 . Intriguingly, T cells expressing Rltpr Bas showed an increase in antibody-induced CD28 internalization as compared with T cells expressing Rltpr. Therefore, by preserving the actin- uncapping activity but breaking the linkage of CD28 to PKC9 and Carmal, the Rltpr Bas mutation might reduce the "drag" through the cytosol and thus increase CD28 internalization over residency at the cell surface.

Discussion:

Using a sensitized ENU mutagenesis screen we demonstrated that the poorly characterized Rltpr gene product is essential for CD28 costimulation. Consistent with its involvement in CD28 function, Rltpr was primarily expressed in developing and mature T cells. We showed that Rltpr Bas did not affect the expression of CD28 at the T-cell surface and that TCR-CD28 engagement at the IS resulted in the spatial co localization of CD28 with both Rltpr and Rltpr Bas molecules. However, Rltpr Bas abrogated the connection between CD28 and PKC- Θ and Carmal . Consistent with that mechanistic defect, comparative analysis of antigen- induced CD4 + and CD8 + T cell activation and of nTreg and T FH cell development in Rltpr Bas and Cd28 ~ ' ~ mice showed that the Rltpr Bas mutation constituted a functional phenocopy of a CD28 deficiency. These last findings reinforce in a unbiased manner the importance of CD28- PKC-9-Carmal signaling for the differentiation of nTreg and T FH cells 44 ' 45 .

Many LRR-containing domains provide a structural framework for protein-protein interactions 24 , and the non-conservative Rltpr Bas mutation that occurred in the eighth LRR likely prevented Rltpr from playing its role of intermediate between CD28 and PKC- Θ/Carmal . Owing to its large size, Rltpr is likely a multitask protein composed of distinct functional domains. Rltpr Bas was still capable of constitutively associating with CP, a finding consistent with the large distance that separate the Rltpr Ba mutation from the CPI motif. Actin polymerization in vivo occurs primarily at the barbed end of actin filament and mechanisms that control the number of free barbed ends are crucial for the regulation of actin filament assembly within cells. The presence of a CP at the barbed end of an actin filament inhibits the addition of actin subunits at that end. Therefore, by preventing the action of CP, actin- uncapping proteins such as Rltpr generate free barbed ends and enhance actin polymerization. pi 16, a Dictyostelium Lrcc-like protein, localizes in dynamic actin-rich cellular extensions including macropynocytic crown and the leading edge of locomoting cells 46 . Dictyostelium cells lacking pi 16 showed a reduction in the formation of these structures. On that basis, it has been proposed that most Lrcc-family proteins act to promote actin polymerization and enhance processes such as cell movement and endocytosis 21-23 ' 46 ' 47 . By demonstrating that the actin-uncapping protein Rltpr is essential for CD28 costimulation, the present study supports the view that actin dynamic regulation plays an important role in CD28-mediated costimulation 42 ' 43 .

Using a system-scale approach 38 , we showed that the pattern of distribution of Rltpr at the T cell-APC interface had the greatest similarity with that of the Nek and Cin85 adaptors. Nek and Cin85 have been shown to link antigen receptors to actin regulation and receptor internalization 41 . Interestingly, Cin85 primarily down-regulates the activity of receptor protein-tyrosine kinase via endocytosis and possesses actin-uncapping activity 21 . Reminiscent of our observations with Rltpr, CD2 is also prominently found in the large T cell invagination that forms at T cell-APC interface 39 , and it associates with CP via Cin85 48 . The exacerbated occurrence of patterns associated with internalization (invagination and internal accumulation) in T cells expressing Rltpr Bas as compared to WT T cells is consistent with the view that the L432P mutation relieved Rltpr from long-lasting interactions with effectors such as PKC-Θ and Carmal that reside at the inner face of the T cell membrane. Congruent with hat view, T cells expressing Rltpr Bas showed a higher rate of antibody-mediated CD28 endocytosis as compared to WT T cells. Moreover, the central accumulation of Rltpr Bas -YPET was shorter than that of Rltpr- YPET. Therefore, the extent to which the CD28-Rltpr "nucleation center" resides at the T cell surface prior to endocytosis appears to depend on its ability to interact with PKC-Θ and Carmal . Interestingly, gene-expression profiling of whole skin samples from psoriatic patients showed that the Rltpr gene was downregulated 49 , and a recent study demonstrated that polymorphisms with the human Rltpr gene were associated with ankylosing spondylitis 50 , pointing to a possible role of Rltpr in T-cell dependent autoimmunity.

In conclusion, these genetic, functional and imaging studies have uncovered a role for Rltpr, a lymphoid lineage-specific actin-uncapping protein, as an essential player of the CD28 costimulatory pathway. They led us to modify the existing model of CD28 costimulation to a new more complete model in which Rltpr plays an essential role owing to its capacity to couple CD28 to PKC-Θ and Carmal . Whether CD28 engagement induces distinct signals or simply amplifies TCR signaling has been the object of controversy. It will be interesting to determine whether Rltpr constitutes the long-sought component specific for the CD28 signaling pathway.

REFERENCES:

Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.

1. Okkenhaug, K. et al. A point mutation in CD28 distinguishes proliferative signals from survival signals. Nature Immunology 2, 325-332 (2001). 2. Holdorf, A.D., Lee, K.H., Burack, W.R., Allen, P.M. & Shaw, A.S. Regulation of Lck activity by CD4 and CD28 in the immunological synapse. Nature Immunology 3, 259- 264 (2002).

3. Isakov, N. & Altman, A. PKC-theta-mediated signal delivery from the TCR/CD28 surface receptors. Front Immunol 3, 273 (2012).

4. Raab, M., Pfister, S. & Rudd, C.E. CD28 signaling via VAV/SLP-76 adaptors: regulation of cytokine transcription independent of TCR ligation. Immunity 15, 921-933 (2001).

5. Pagan, A. J., Pepper, M., Chu, H.H., Green, J.M. & Jenkins, M.K. CD28 Promotes CD4+ T cell clonal expansion during infection independently of its YMNM and PYAP motifs. Journal of immunology 189, 2909-2917 (2012).

6. Thome, M., Charton, J.E., Pelzer, C. & Hailfmger, S. Antigen receptor signaling to NF-kappaB via CARMA1, BCL10, and MALT1. Cold Spring Harbor perspectives in biology 2, a003004 (2010).

7. Wang, X., Chuang, H.C., Li, J.P. & Tan, T.H. Regulation of PKC-theta function by phosphorylation in T cell receptor signaling. Front Immunol 3, 197 (2012).

8. Jiang, C. & Lin, X. Regulation of NF-kappaB by the CARD proteins. Immunological reviews 246, 141-153 (2012).

9. Yokosuka, T. et al. Spatiotemporal regulation of T cell costimulation by TCR-CD28 microclusters and protein kinase C theta translocation. Immunity 29, 589-601

(2008) .

10. Dustin, M.L. The cellular context of T cell signaling. Immunity 30, 482-492

(2009) .

11. Vardhana, S., Choudhuri, K., Varma, R. & Dustin, M.L. Essential role of ubiquitin and TSG101 protein in formation and function of the central supramolecular activation cluster. Immunity 32, 531-540 (2010).

12. Yokosuka, T. et al. Spatiotemporal basis of CTLA-4 costimulatory molecule- mediated negative regulation of T cell activation. Immunity 33, 326-339 (2010).

13. Aguado, E. et al. Induction of T Helper Type 2 Immunity by a Point Mutation in the LAT Adaptor. Science 296, 2036-2040 (2002).

14. Sommers, C.L. et al. A LAT mutation that inhibits T cell development yet induces lymphoproliferation. Science 296, 2040-2043 (2002). 15. Wang, Y. et al. Th2 lymphoproliferative disorder of LatY136F mutant mice unfolds independently of TCR-MHC engagement and is insensitive to the action of Foxp3+ regulatory T cells. J Immunol 180, 1565-1575 (2008).

16. Mingueneau, M. et al. Loss of the LAT adaptor converts antigen-responsive T cells into pathogenic effectors that function independently of the T cell receptor. Immunity

31, 197-208 (2009).

17. Chevrier, S., Genton, C, Malissen, B., Malissen, M. & Acha-Orbea, H. Dominant Role of CD80-CD86 Over CD40 and ICOSL in the Massive Polyclonal B Cell Activation Mediated by LAT(Y136F) CD4(+) T Cells. Front Immunol 3, 27 (2012).

18. Beutler, B. et al. Genetic analysis of resistance to viral infection. Nat Rev

Immunol 7, 753-766 (2007).

19. Cook, M.C., Vinuesa, C.G. & Goodnow, C.C. ENU-mutagenesis: insight into immune function and pathology. Current opinion in immunology 18, 627-633 (2006).

20. Liang, Y., Niederstrasser, FL, Edwards, M., Jackson, C.E. & Cooper, J.A. Distinct roles for CARMIL iso forms in cell migration. Mol Biol Cell 20, 5290-5305 (2009).

21. Hernandez- Valladares, M. et al. Structural characterization of a capping protein interaction motif defines a family of actin filament regulators. Nat Struct Mol Biol 17, 497- 503 (2010).

22. Fujiwara, I., Remmert, K. & Hammer, J.A., 3rd Direct observation of the uncapping of capping protein-capped actin filaments by CARMIL homology domain 3. J Biol

Chem 285, 2707-2720 (2010).

23. Yang, C. et al. Mammalian CARMIL inhibits actin filament capping by capping protein. Dev Cell 9, 209-221 (2005).

24. Ng, A.C. et al. Human leucine-rich repeat proteins: a genome-wide bioinformatic categorization and functional analysis in innate immunity. Proceedings of the

National Academy of Sciences of the United States of America 108 Suppl 1, 4631-4638 (2011).

25. Junttila, M.R., Saarinen, S., Schmidt, T., Kast, J. & Westermarck, J. Single- step Strep- tag purification for the isolation and identification of protein complexes from mammalian cells. Proteomics 5, 1 199-1203 (2005).

26. Bachmann, M.F. et al. T cell responses are governed by avidity and co- stimulatory thresholds. European journal of immunology 26, 2017-2022 (1996).

27. Shahinian, A. et al. Differential T cell costimulatory requirements in CD28- deficient mice. Science 261, 609-612 (1993). 28. Hogquist, K.A. et al. T cell receptor antagonist peptides induce positive selection. Cell 76, 17-27 (1994).

29. Kaye, J. et al. Selective development of CD4+ T cells in transgenic mice expressing a class II MHC-restricted antigen receptor. Nature 341, 746-749 (1989).

30. Tai, X., Cowan, M., Feigenbaum, L. & Singer, A. CD28 costimulation of developing thymocytes induces Foxp3 expression and regulatory T cell differentiation independently of interleukin 2. Nature Immunology 6, 152-162 (2005).

31. Roman, E., Shino, H., Qin, F.X. & Liu, Y.J. Cutting edge: Hematopoietic- derived APCs select regulatory T cells in thymus. Journal of immunology 185, 3819-3823

(2010).

32. Vang, K.B. et al. Cutting edge: CD28 and c-Rel-dependent pathways initiate regulatory T cell development. Journal of immunology 184, 4074-4077 (2010).

33. Salek-Ardakani, S. et al. B cell-specific expression of B7-2 is required for follicular Th cell function in response to vaccinia virus. Journal of immunology 186, 5294-

5303 (2011).

34. Nguyen, A.W. & Daugherty, P.S. Evolutionary optimization of fluorescent proteins for intracellular FRET. Nat Biotechnol 23, 355-360 (2005).

35. Campi, G., Varma, R. & Dustin, M.L. Actin and agonist MHC-peptide complex- dependent T cell receptor microclusters as scaffolds for signaling. The Journal of experimental medicine 202, 1031-1036 (2005).

36. Yokosuka, T. et al. Newly generated T cell receptor microclusters initiate and sustain T cell activation by recruitment of Zap70 and SLP-76. Nat Immunol 6, 1253-1262 (2005).

37. Zanin-Zhorov, A. et al. Protein kinase C-theta mediates negative feedback on regulatory T cell function. Science 328, 372-376 (2010).

38. Singleton, K.L. et al. Spatiotemporal patterning during T cell activation is highly diverse. Sci Signal 2, ral5 (2009).

39. Singleton, K. et al. A large T cell invagination with CD2 enrichment resets receptor engagement in the immunological synapse. Journal of immunology 177, 4402-

4413 (2006).

40. DeFord- Watts, L.M. et al. The CD3 zeta subunit contains a phosphoinositide- binding motif that is required for the stable accumulation of TCR-CD3 complex at the immunological synapse. Journal of immunology 186, 6839-6847 (2011). 41. Burkhardt, J.K., Carrizosa, E. & Shaffer, M.H. The actin cytoskeleton in T cell activation. Annu Rev Immunol 26, 233-259 (2008).

42. Cefai, D. et al. CD28 receptor endocytosis is targeted by mutations that disrupt phosphatidylinositol 3-kinase binding and costimulation. Journal of immunology 160, 2223-2230 (1998).

43. Badour, K. et al. Interaction of the Wiskott-Aldrich syndrome protein with sorting nexin 9 is required for CD28 endocytosis and cosignaling in T cells. Proceedings of the National Academy of Sciences of the United States of America 104, 1593-1598 (2007).

44. Barnes, M.J. et al. Commitment to the regulatory T cell lineage requires CARMA1 in the thymus but not in the periphery. PLoS Biol 7, e51 (2009).

45. Molinero, L.L. et al. CARMA1 controls an early checkpoint in the thymic development of FoxP3+ regulatory T cells. Journal of immunology 182, 6736-6743 (2009).

46. Jung, G., Remmert, K., Wu, X., Volosky, J.M. & Hammer, J.A., 3rd The Dictyostelium CARMIL protein links capping protein and the Arp2/3 complex to type I myosins through their SH3 domains. J Cell Biol 153, 1479-1497 (2001).

47. Takeda, S. et al. Actin capping protein and its inhibitor CARMIL: how intrinsically disordered regions function. Phys Biol 8, 035005 (2011).

48. Hutchings, N.J., Clarkson, N., Chalkley, R., Barclay, A.N. & Brown, M.H. Linking the T cell surface protein CD2 to the actin-capping protein CAPZ via CMS and CIN85. J Biol Chem 278, 22396-22403 (2003).

49. Matsuzaka, Y. et al. Identification, expression analysis and polymorphism of a novel RLTPR gene encoding a RGD motif, tropomodulin domain and proline/leucine- rich regions. Gene 343, 291-304 (2004).

50. Pointon, J.J. et al. The chromosome 16q region associated with ankylosing spondylitis includes the candidate gene tumour necrosis factor receptor type 1 -associated death domain (TRADD). Ann Rheum Dis 69, 1243-1246 (2010).

51. Seder, R.A., Paul, W.E., Davis, M.M. & Fazekas de St Groth, B. The presence of interleukin 4 during in vitro priming determines the lymphokine-producing potential of CD4+ T cells from T cell receptor transgenic mice. The Journal of experimental medicine 176, 1091-1098 (1992).

52. Malissen, M. et al. Altered T cell development in mice with a targeted mutation of the CD3-epsilon gene. Embo J X , 4641-4653 (1995). 53. Ordonez-Rueda, D. et al. A hypomorphic mutation in the Gfil transcriptional repressor results in a novel form of neutropenia. European journal of immunology 42, 2395- 2408 (2012).

54. Harakalova, M. et al. Multiplexed array-based and in-solution genomic enrichment for flexible and cost-effective targeted next-generation sequencing. Nat Protoc

6, 1870-1886 (2011).

55. Kitamura, T. et al. Retro virus-mediated gene transfer and expression cloning: powerful tools in functional genomics. Exp Hematol 31, 1007-1014 (2003).

56. Heavey, B., Charalambous, C, Cobaleda, C. & Busslinger, M. Myeloid lineage switch of Pax5 mutant but not wild-type B cell progenitors by C/EBPalpha and GATA factors. The EMBO journal 22, 3887-3897 (2003).

57. Morita, S., Kojima, T. & Kitamura, T. Plat-E: an efficient and stable system for transient packaging of retroviruses. Gene Ther 7, 1063-1066 (2000).

58. Reske-Kunz, A.B. & Rude, E. Insulin-specific T cell hybridomas derived from (H-2b x H-2k)Fl mice preferably employ Fl -unique restriction elements for antigen recognition. European journal of immunology 15, 1048-1054 (1985).

59. Tokunaga, M., Kitamura, K., Saito, K., Iwane, A.H. & Yanagida, T. Single molecule imaging of fluorophores and enzymatic reactions achieved by objective-type total internal reflection fluorescence microscopy. Biochem Biophys Res Commun 235, 47-53 (1997).