Matches in SemOpenAlex for { <https://semopenalex.org/work/W1978322477> ?p ?o ?g. }
Showing items 1 to 99 of
99
with 100 items per page.
- W1978322477 endingPage "1574" @default.
- W1978322477 startingPage "1570" @default.
- W1978322477 abstract "Engagement of the T-cell receptor (TCR) initiates a signaling cascade that ultimately results in activation of the transcription factor NF-κB, which regulates many T-cell functions including proliferation, differentiation and cytokine production. Herein we demonstrate that Rip2, a caspase recruitment domain (CARD)-containing serine/threonine kinase, plays an important role in this cascade and is required for optimal TCR signaling and NF-κB activation. Following TCR engagement, Rip2 associated with Bcl10, a CARD-containing signaling component of the TCR-induced NF-κB pathway, and induced its phosphorylation. Rip2-deficient mice were defective in TCR-induced NF-κB activation, interleukin-2 production, and proliferation in vitro and exhibited defective T-cell-dependent responses in vivo. The defect in Rip2-/- T-cells correlated with a lack of TCR-induced Bcl10 phosphorylation. Furthermore, deficiency in Bcl10-dependent NF-κB activation could be rescued in Rip2-/- embryonic fibroblasts by exogenous wild-type Rip2 but not a kinase-dead mutant. Together these data define an important role for Rip2 in TCR-induced NF-κB activation and T-cell function and highlight the significance of post-translational modification of Bcl10 by Rip2 in T-cell signaling. Engagement of the T-cell receptor (TCR) initiates a signaling cascade that ultimately results in activation of the transcription factor NF-κB, which regulates many T-cell functions including proliferation, differentiation and cytokine production. Herein we demonstrate that Rip2, a caspase recruitment domain (CARD)-containing serine/threonine kinase, plays an important role in this cascade and is required for optimal TCR signaling and NF-κB activation. Following TCR engagement, Rip2 associated with Bcl10, a CARD-containing signaling component of the TCR-induced NF-κB pathway, and induced its phosphorylation. Rip2-deficient mice were defective in TCR-induced NF-κB activation, interleukin-2 production, and proliferation in vitro and exhibited defective T-cell-dependent responses in vivo. The defect in Rip2-/- T-cells correlated with a lack of TCR-induced Bcl10 phosphorylation. Furthermore, deficiency in Bcl10-dependent NF-κB activation could be rescued in Rip2-/- embryonic fibroblasts by exogenous wild-type Rip2 but not a kinase-dead mutant. Together these data define an important role for Rip2 in TCR-induced NF-κB activation and T-cell function and highlight the significance of post-translational modification of Bcl10 by Rip2 in T-cell signaling. Many diverse stimuli activate NF-κB by inducing the phosphorylation and destruction of inhibitory molecules known as the IκBs that retain NF-κB in the cytoplasm (1.Ghosh S. May M.J. Kopp E.B. Annu. Rev. Immunol. 1998; 16: 225-260Crossref PubMed Scopus (4607) Google Scholar). The IκB kinase (IKK) 1The abbreviations used are: IKK, IκB kinase; CARD, caspase recruitment domain; TCR, T-cell receptor; TLR, Toll-like receptor; LPS, lipopolysaccaride; HA, hemagglutinin; PMA, phorbol myristate acetate; IL-2, interleukin-2; MEF, mouse embryonic fibroblast; ERK, extracellular signal-regulated kinase; JNK, c-Jun NH2-terminal kinase. complex, composed of two kinase subunits, IKKα and IKKβ, and a non-catalytic subunit, NEMO/IKKγ, is responsible for the phosphorylation of the IκBs. The association of Bcl10 and CARMA1 (CARD11), two caspase-recruitment domain (CARD)-containing proteins, has been shown to be essential to the transduction of the signal from the T-cell receptor (TCR) to the IKK complex (2.Thome M. Tschopp J. Trends Immunol. 2003; 24: 419-424Abstract Full Text Full Text PDF PubMed Scopus (86) Google Scholar). Mice deficient for either Bcl10 or CARMA1 display profound defects in T-cell proliferation and cytokine production due to a lack of NF-κB activation (3.Ruland J. Duncan G.S. Elia A. del Barco Barrantes I. Nguyen L. Plyte S. Millar D.G. Bouchard D. Wakeham A. Ohashi P.S. Mak T.W. Cell. 2001; 104: 33-42Abstract Full Text Full Text PDF PubMed Scopus (481) Google Scholar, 4.Newton K. Dixit V.M. Curr. Biol. 2003; 13: 1247-1251Abstract Full Text Full Text PDF PubMed Scopus (131) Google Scholar, 5.Egawa T. Albrecht B. Favier B. Sunshine M.J. Mirchandani K. O'Brien W. Thome M. Littman D.R. Curr. Biol. 2003; 13: 1252-1258Abstract Full Text Full Text PDF PubMed Scopus (212) Google Scholar, 6.Hara H. Wada T. Bakal C. Kozieradzki I. Suzuki S. Suzuki N. Nghiem M. Griffiths E.K. Krawczyk C. Bauer B. D'Acquisto F. Ghosh S. Yeh W.C. Baier G. Rottapel R. Penninger J.M. Immunity. 2003; 18: 763-775Abstract Full Text Full Text PDF PubMed Scopus (280) Google Scholar, 7.Jun J.E. Wilson L.E. Vinuesa C.G. Lesage S. Blery M. Miosge L.A. Cook M.C. Kucharska E.M. Hara H. Penninger J.M. Domashenz H. Hong N.A. Glynne R.J. Nelms K.A. Goodnow C.C. Immunity. 2003; 18: 751-762Abstract Full Text Full Text PDF PubMed Scopus (259) Google Scholar); however, the mechanism by which the CARMA1/Bcl10 complex activates IKK remains unclear. In vitro experiments have indicated that Bcl10 undergoes phosphorylation when over expressed with its viral homologue, E10 or CARMA1 (8.Thome M. Gaide O. Micheau O. Martinon F. Bonnet D. Gonzalez M. Tschopp J. J. Cell Biol. 2001; 152: 1115-1122Crossref PubMed Scopus (17) Google Scholar, 9.Gaide O. Martinon F. Micheau O. Bonnet D. Thome M. Tschopp J. FEBS Lett. 2001; 496: 121-127Crossref PubMed Scopus (169) Google Scholar, 10.Bertin J. Wang L. Guo Y. Jacobson M.D. Poyet J.L. Srinivasula S.M. Merriam S. DiStefano P.S. Alnemri E.S. J. Biol. Chem. 2001; 276: 11877-11882Abstract Full Text Full Text PDF PubMed Scopus (278) Google Scholar). In these studies, Bcl10 phosphorylation correlated with its ability to activate NF-κB, suggesting that this modification was required for NF-κB activation. Indeed, the COOH-terminal domain of Bcl10 is rich in serine and threonine residues and has been proposed as the site of CARMA1-mediated phosphorylation (10.Bertin J. Wang L. Guo Y. Jacobson M.D. Poyet J.L. Srinivasula S.M. Merriam S. DiStefano P.S. Alnemri E.S. J. Biol. Chem. 2001; 276: 11877-11882Abstract Full Text Full Text PDF PubMed Scopus (278) Google Scholar). Since CARMA1 itself is not a kinase, the kinase responsible for Bcl10 phosphorylation has remained an open question. Rip2 is a serine/threonine kinase that contains a CARD domain at its carboxyl terminus and has been shown to induce NF-κB activation in over expression systems (11.McCarthy J.V. Ni J. Dixit V.M. J. Biol. Chem. 1998; 273: 16968-16975Abstract Full Text Full Text PDF PubMed Scopus (363) Google Scholar, 12.Thome M. Hofmann K. Burns K. Martinon F. Bodmer J.L. Mattmann C. Tschopp J. Curr. Biol. 1998; 8: 885-888Abstract Full Text Full Text PDF PubMed Google Scholar, 13.Inohara N. del Peso L. Koseki T. Chen S. Nunez G. J. Biol. Chem. 1998; 273: 12296-12300Abstract Full Text Full Text PDF PubMed Scopus (216) Google Scholar). Rip2 has also been shown to associate in vitro with members of the TRAF family, such as TRAF6, that plays an essential role in the innate immune response downstream of Toll-like receptors (TLRs) (14.Mak T.W. Yeh W.C. Nature. 2002; 418: 835-836Crossref PubMed Scopus (12) Google Scholar, 15.Lomaga M.A. Yeh W.C. Sarosi I. Duncan G.S. Furlonger C. Ho A. Morony S. Capparelli C. Van G. Kaufman S. van der Heiden A. Itie A. Wakeham A. Khoo W. Sasaki T. Cao Z. Penninger J.M. Paige C.J. Lacey D.L. Dunstan C.R. Boyle W.J. Goeddel D.V. Mak T.W. Genes Dev. 1999; 13: 1015-1024Crossref PubMed Scopus (1080) Google Scholar). In addition, Rip2 has been implicated in regulating both the innate and adaptive immune responses (16.Kobayashi K. Inohara N. Hernandez L.D. Galan J.E. Nunez G. Janeway C.A. Medzhitov R. Flavell R.A. Nature. 2002; 416: 194-199Crossref PubMed Scopus (744) Google Scholar, 17.Chin A.I. Dempsey P.W. Bruhn K. Miller J.F. Xu Y. Cheng G. Nature. 2002; 416: 190-194Crossref PubMed Scopus (330) Google Scholar). Mice deficient in Rip2 mounted only an attenuated immune response against Toll-like receptor agonists such as lipopolysaccaride (LPS) (16.Kobayashi K. Inohara N. Hernandez L.D. Galan J.E. Nunez G. Janeway C.A. Medzhitov R. Flavell R.A. Nature. 2002; 416: 194-199Crossref PubMed Scopus (744) Google Scholar, 17.Chin A.I. Dempsey P.W. Bruhn K. Miller J.F. Xu Y. Cheng G. Nature. 2002; 416: 190-194Crossref PubMed Scopus (330) Google Scholar). Interestingly, CD4+ T-cells from Rip2-deficient mice were unable to proliferate efficiently in response to antigen-induced T-cell activation, but no mechanism was provided for this striking observation (16.Kobayashi K. Inohara N. Hernandez L.D. Galan J.E. Nunez G. Janeway C.A. Medzhitov R. Flavell R.A. Nature. 2002; 416: 194-199Crossref PubMed Scopus (744) Google Scholar, 17.Chin A.I. Dempsey P.W. Bruhn K. Miller J.F. Xu Y. Cheng G. Nature. 2002; 416: 190-194Crossref PubMed Scopus (330) Google Scholar). We sought to define the role of Rip2 in antigen-induced NF-κB activation and T-cell proliferation. Generation of Rip2-/- Mice—A targeting vector that removed exon I of Rip2 was electroporated into ES cells. Homologous recombinants were used to generate chimeric founder mice by microinjection into C57BL/6J blastocysts. Germ line transmission was confirmed by Southern blot analysis of genomic tail DNA. Two independent ES clone lines resulted in mice with identical phenotypes. All mice used in experiments were backcrossed onto C57BL/6 five to seven generations and were confirmed to be >95% C57BL/6 by PCR analysis of genomic tail DNA. Immunoprecipitations—293 T-cells were transfected with expression vectors for NH2-terminal HA- or Myc/His-tagged Rip2, Rip2 311-541(ΔKinase) and Rip2 1-454 (ΔCARD) (11.McCarthy J.V. Ni J. Dixit V.M. J. Biol. Chem. 1998; 273: 16968-16975Abstract Full Text Full Text PDF PubMed Scopus (363) Google Scholar), and V5-tagged Bcl10 (18.Yan M. Lee J. Schilbach S. Goddard A. Dixit V. J. Biol. Chem. 1999; 274: 10287-10292Abstract Full Text Full Text PDF PubMed Scopus (99) Google Scholar). Tagged proteins were immunoprecipitated using monoclonal antibodies to Rip2 (Genentech, Inc.), V5 (Invitrogen), HA and Myc (Zymed Laboratories Inc.), and actin (ICN). Jurkat cells (4 × 107) were stimulated with cross-linking antibodies to the Jurkat TCR clone C305 (a gift from A. Chan, Genentech, Inc.). Cell lysates were prepared as described previously (10.Bertin J. Wang L. Guo Y. Jacobson M.D. Poyet J.L. Srinivasula S.M. Merriam S. DiStefano P.S. Alnemri E.S. J. Biol. Chem. 2001; 276: 11877-11882Abstract Full Text Full Text PDF PubMed Scopus (278) Google Scholar). Endogenous proteins were immunoprecipitated using anti-Bcl10 clone 331.3 (Santa Cruz) or anti-Rip2 and protein A/G beads (Pierce) and analyzed by Western blots using antibodies to Bcl10 (H-197) or Bcl10 (331.3) (Santa Cruz) and phospho-Zap-70 (Cell Signaling Technology). In some cases, immunoprecipitates were treated with λ phosphatase (New England Biolabs). Proliferation Assays—Splenic B and T-cells and CD4+ T were purified by negative selection using magnetic beads (Miltenyi Biotech) to >95% purity. Purified T-cells were activated with plate bound anti-CD3 (0-10 μg/ml) (BD Biosciences) with or without irradiated CD4-depleted APCs or plate-bound anti-CD28 (0-10 μg/ml) (BD Biosciences), phorbol myristate acetate (PMA) (2 ng/ml) plus ionomycin (0.1 μg/ml) (Sigma) in the presence or absence of IL-2 (50 ng/ml) (R & D Systems). B-cells were stimulated with anti-IgM (20 μg/ml) (Jackson Laboratories), LPS (20 μg/ml) (Sigma), or PMA (2 ng/ml) plus ionomycin (0.1 μg/ml). Cells were harvested at 24, 48, 72, and 96 h after an 8-h pulse with [3H]thymidine (1 μCi/well), and incorporation of [3H]thymidine was measured using a Matrix 96 direct β counter system (Hewlett-Packard). Data represent triplicate samples and are representative of at least three separate experiments. Neonatal Heart Allograft—Neonatal hearts from BALB/c (H-2d) mice were surgically implanted behind the dorsum of the ear pinna of 12-week-old male Rip2-/- and wild-type mice (Both H-2b). Heart grafts were examined with a stereomicroscope at 10-20-fold magnification every other day until rejection. Western Blots—Purified T-cells (2 × 106) were stimulated with 10 μg/ml plate-bound anti-CD3 for 0-30 min. Western blots were performed using phospho-specific antibodies for IκBα, ERK1/2, and JNK (Cell Signaling Technology). Blots were stripped and re-probed with antibodies to IκBα and p44 (Cell Signaling Technology). NF-κB Luciferase Assays—Mouse embryonic fibroblasts (MEFs) were transfected using Superfect (Qiagen) with 2-μg expression vectors (V5-Bcl10, HA-Rip2, Myc/His-K47ARip2 (11.McCarthy J.V. Ni J. Dixit V.M. J. Biol. Chem. 1998; 273: 16968-16975Abstract Full Text Full Text PDF PubMed Scopus (363) Google Scholar), or HA-pcDNA3), 0.25 μg pLam3 (NF-κB-responsive luciferase), and 0.025 μg of pR-TK (control luciferase). Luciferase activity was measured 48 h post-transfection using the dual luciferase reporter assay system (Promega). Data represent triplicate samples and are representative of at least three separate experiments. Isolation of Phosphorylated Proteins—Splenic T-cells from Rip2-/- and wild-type mice (4 × 107) cells/ml) were stimulated with 10 μg/ml plate-bound anti-CD3 for 0, 15, and 30 min. Cells were lysed under denaturing conditions to disrupt protein-protein interactions and diluted to 0.1 mg/ml in phospho-lysis buffer (Qiagen). Phosphorylated proteins were separated using the phospho-protein purification kit (Qiagen) according to the manufacturer's instructions. Rip2 Associates with Bcl10 and Induces Its Phosphorylation—We investigated whether Rip2 could associate with molecules known to play essential roles in the TCR-induced signaling cascade. Initially, we tested whether Rip2 and Bcl10 could associate by overexpressing tagged versions of both proteins in 293 T-cells. V5-tagged Bcl10 could be co-immunoprecipitated with HA-tagged Rip2 (Fig. 1A). Interestingly, two bands representing Bcl10 were observed. The upper band was determined to be a hyperphosphorylated form of Bcl10, since it could be collapsed to the lower band by λ phosphatase treatment (Fig. 1A). Hyperphosphorylation of Bcl10 was also apparent by mobility shift in whole cell lysates from 293T-cells co-transfected with Rip2 and Bcl10, compared with the very low levels of phosphorylation seen with Bcl10 alone (Fig. 1B). To establish the domains of Rip2 responsible for hyperphosphorylation of Bcl10, mutants with deletions of either the kinase domain or the CARD domain were used in co-expression studies. Bcl10 hyperphosphorylation required both a functional kinase domain and CARD domain of Rip2 as neither mutant induced phosphorylation of Bcl10 (Fig. 1B). Moreover, phosphorylation of Bcl10 was specific for Rip2, as overexpression of RIP or Rip3 did not induce Bcl10 phosphorylation (Fig. 1C). To determine whether Rip2 was involved in Bcl10-dependent signaling pathways, we studied the interaction of endogenous proteins in Jurkat cells stimulated with cross-linking antibodies to CD3-TCR. Rip2 and Bcl10 consistently associated in a transient and time-dependent manner after TCR engagement (Fig. 1D, bottom panel). Induction of phosphorylated Zap-70 confirmed TCR activation (Fig. 1E). We next examined the phosphorylation status of Bcl10 using phosphoserine-specific antibodies. Lysates from anti-CD3 treated and untreated Jurkats were immunoprecipitated using antibodies to Bcl10 and Western blots were performed using antibodies for phosphoserine and Bcl10. Serine phosphorylated Bcl10 was detected after 15-min treatment with anti-CD3 (Fig. 1F, top panel) and treatment of immunoprecipitates with λ phosphatase (λPPase) significantly diminished levels of serine-phosphorylated Bcl10. Phosphorylation of endogenous Bcl10 was also apparent after treatment with anti-CD3, as evidenced by a slower migrating band that could be collapsed by treatment with λ phosphatase (Fig. 1F, bottom panel). Taken together, these results were consistent with Rip2 binding Bcl10 upon TCR engagement and inducing its phosphorylation. Defective T-cell Proliferation and Function in Rip2-/- Mice—To examine the effects of Rip2 on T cell activation in an in vivo setting, we generated Rip2-deficient mice by homologous recombination. Rip2-/- T-cells were deficient in anti-CD3 induced proliferation (Fig. 2, A and B). This defect could not be rescued by co-stimulation with anti-CD28 or activation using PMA in combination with calcium ionophore (ion) (Fig. 2C). The levels of IL-2 produced after treatment with anti-CD3 alone, anti-CD3 with anti-CD28, or with PMA and ionomycin were drastically reduced compared with wild-type T-cells (data not shown). Moreover, addition of exogenous IL-2 was not able to rescue the defect in proliferation in Rip2-/- T-cells after stimulation (Fig. 2D). Consistent with previous reports, B-cell proliferation in response to PMA/ionomycin, IgM, and LPS was comparable between Rip2-deficient and wild-type B-cells (data not shown) (16.Kobayashi K. Inohara N. Hernandez L.D. Galan J.E. Nunez G. Janeway C.A. Medzhitov R. Flavell R.A. Nature. 2002; 416: 194-199Crossref PubMed Scopus (744) Google Scholar). Taken together, these results suggested that the defect in proliferation in Rip2-/- mice was confined to T-cells and likely due to impairment upstream of IL-2 gene transcription and NF-κB activation. Previous in vivo experiments on Rip2-/- mice tested T-cell responsiveness using models such as Listeria challenge and T-cell-dependent antibody responses (16.Kobayashi K. Inohara N. Hernandez L.D. Galan J.E. Nunez G. Janeway C.A. Medzhitov R. Flavell R.A. Nature. 2002; 416: 194-199Crossref PubMed Scopus (744) Google Scholar, 17.Chin A.I. Dempsey P.W. Bruhn K. Miller J.F. Xu Y. Cheng G. Nature. 2002; 416: 190-194Crossref PubMed Scopus (330) Google Scholar), which also involve participation of TLRs and other innate signaling cascades through adjuvant and bacterial components. Therefore, to test T-cell responsiveness in vivo, we designed a high bar functional test, the ability to participate in a graft rejection response, which does not require major driver co-signals from pathways of the innate immune system. Hearts from allogeneic neonate BALB/c (H-2d) mice were transplanted into the ear pinna of wild-type and Rip2-deficient mice (H-2b) and allograft survival monitored. While all hearts were rejected by wild-type mice by day 15, over 50% of the neonate hearts were still beating in Rip2-deficient mice and continued to function for an additional 5 days (Fig. 2E). Therefore, Rip2-deficient mice rejected heart allografts much less readily than wild-type mice, consistent with our in vitro data and a defect in normal T-cell activation and function. Defective NF-κB Activation in Rip2-deficient Cells—To determine the molecular basis of the impairment in T-cell receptor signaling in the absence of Rip2, we analyzed pathways activated by TCR engagement in wild-type and Rip2-/- T-cells. T-cells from wild-type and Rip2-/- mice were treated with plate-bound anti-CD3 or TNFα, and lysates were assessed by Western blot using phospho-specific antibodies to IκBα. IκBα was rapidly phosphorylated and degraded in wild-type T-cells but not in Rip2-/- cells (Fig. 3A). In contrast, treatment of both wild-type and Rip2-/- T-cells with TNFα promoted equivalent phosphorylation and degradation of IκBα (Fig. 3B). Hence, NF-κB signaling downstream of other surface receptors remained intact in Rip2-/- mice. TCR engagement also elicits activation of the RAS/MAPK (mitogen-activated protein kinase) pathway. Western blotting using phospho-specific anti-ERK1/2 antibodies demonstrated that ERK-1 and ERK-2 were phosphorylated with similar kinetics in wild-type and Rip2-/- T-cells after TCR engagement (Fig. 3C, upper panel). Similarly, activation of the JNK signaling pathway post-TCR engagement was equivalent in both wild-type and Rip2-deficient T-cells (Fig. 3C, lower panel). These results confirmed that the defect was specific for NF-κB signaling downstream of the TCR and that parallel pathways activated by TCR engagement remained intact. To address the role of Rip2 kinase activity in Bcl10-dependent NF-κB activation, we transfected MEFs from wild-type and Rip2-/- mice with Bcl10 and a luciferase reporter for NF-κB. While Bcl10 could induce NF-κB activation in wild-type MEFs, NF-κB activation by Bcl10 was significantly decreased in Rip2-/- MEFS (Fig. 3D). Transfection of exogenous wild-type Rip2, but not a kinase-dead mutant, K47A, could rescue Bcl10-induced NF-κB reporter activity in Rip2-/- MEFs (Fig. 3D). Therefore, the kinase activity of Rip2 is required for optimal Bcl10-induced NF-κB activation. Bcl10 Is Phosphorylated after TCR Engagement in Wild-type but Not Rip2-/- Mice—Taken together, our data suggested that Rip2 functions to regulate T-cell activation by phosphorylating Bcl10. Therefore, we wished to establish whether the lack of NF-κB activation observed in Rip2-/- T-cells correlated with a lack of Bcl10 phosphorylation after TCR engagement. Wild-type and Rip2-deficient T-cells were treated with α-CD3, and cell lysates were fractionated using a phosphoserine/threonine column. Under these lysis conditions, all protein-protein interactions are disrupted, and only phosphorylated proteins bind the column, while unphosphorylated proteins flow through. Western blotting of the phosphorylated protein fractions using antibodies to phospho-ERK and Bcl10 revealed that while phosphorylated ERK1/2 could easily be detected in the purified phosphorylated fractions of both wild-type and Rip2-/- T-cells (Fig. 3E, middle panel), Bcl10 was only present in the purified phosphorylated fractions of α-CD3 treated wild-type T-cells (Fig. 3E, top panel). By contrast, similar levels of Bcl10 were detected in the non-phosphorylated fractions from wild-type and knock-out T-cells (Fig. 3E, bottom panel). To confirm that no unphosphorylated proteins contaminated the phosphorylated protein fraction, lysates from both fractions were Western blotted using antibodies for Hsp60. Hsp60 was abundant in the non-phosphorylated fraction but undetectable in the phosphorylated protein fraction (Fig. 3F). These data demonstrate that Bcl10 is phosphorylated in mouse primary T cells after TCR stimulation, and deficiency of Rip2 precludes phosphorylation of Bcl10. Herein we provide evidence for the importance of Rip2 in TCR-mediated NF-κB activation and Bcl10-dependent signaling. Phosphorylation of Bcl10 occurs after TCR engagement, and lack of phosphorylation correlates with a defect in NF-κB activation and T-cell proliferation. Earlier studies have shown that Bcl10 is phosphorylated upon over expression of CARMA1; however, the importance of phosphorylation in T-cell signaling was unclear. Our data suggest that phosphorylation of Bcl10 by Rip2 plays a key role in signaling between the TCR and the IKK complex. Recent reports (4.Newton K. Dixit V.M. Curr. Biol. 2003; 13: 1247-1251Abstract Full Text Full Text PDF PubMed Scopus (131) Google Scholar, 5.Egawa T. Albrecht B. Favier B. Sunshine M.J. Mirchandani K. O'Brien W. Thome M. Littman D.R. Curr. Biol. 2003; 13: 1252-1258Abstract Full Text Full Text PDF PubMed Scopus (212) Google Scholar, 6.Hara H. Wada T. Bakal C. Kozieradzki I. Suzuki S. Suzuki N. Nghiem M. Griffiths E.K. Krawczyk C. Bauer B. D'Acquisto F. Ghosh S. Yeh W.C. Baier G. Rottapel R. Penninger J.M. Immunity. 2003; 18: 763-775Abstract Full Text Full Text PDF PubMed Scopus (280) Google Scholar, 7.Jun J.E. Wilson L.E. Vinuesa C.G. Lesage S. Blery M. Miosge L.A. Cook M.C. Kucharska E.M. Hara H. Penninger J.M. Domashenz H. Hong N.A. Glynne R.J. Nelms K.A. Goodnow C.C. Immunity. 2003; 18: 751-762Abstract Full Text Full Text PDF PubMed Scopus (259) Google Scholar, 19.Gaide O. Favier B. Legler D.F. Bonnet D. Brissoni B. Valitutti S. Bron C. Tschopp J. Thome M. Nat. Immunol. 2002; 3: 836-843Crossref PubMed Scopus (292) Google Scholar, 20.Wang D. You Y. Case S.M. McAllister-Lucas L.M. Wang L. DiStefano P.S. Nunez G. Bertin J. Lin X. Nat. Immunol. 2002; 3: 830-835Crossref PubMed Scopus (253) Google Scholar, 21.Pomerantz J.L. Denny E.M. Baltimore D. EMBO J. 2002; 21: 5184-5194Crossref PubMed Scopus (173) Google Scholar) have demonstrated that CARMA1 is critically involved in TCR-induced NF-κB activation. It remains unclear whether Bcl10 phosphorylation is required for its association with CARMA1. The kinetics of the association between Rip2 and Bcl10 and subsequent Bcl10 phosphorylation in Jurkat cells correlates with the kinetics of the published interaction between CARMA1 and Bcl10 in Jurkat T-cells (19.Gaide O. Favier B. Legler D.F. Bonnet D. Brissoni B. Valitutti S. Bron C. Tschopp J. Thome M. Nat. Immunol. 2002; 3: 836-843Crossref PubMed Scopus (292) Google Scholar). Phosphorylation of Bcl10 may either facilitate its recruitment to lipid rafts or serve to activate other key molecules in downstream signaling events that ultimately activate the IKK complex. For example, MALT1/paracaspase, a death domain-containing caspase-like molecule, has also been shown to associate with Bcl10 and enhance NF-κB activation (22.Uren A.G. O'Rourke K. Aravind L.A. Pisabarro M.T. Seshagiri S. Koonin E.V. Dixit V.M. Mol. Cell. 2000; 6: 961-967Abstract Full Text Full Text PDF PubMed Google Scholar, 23.Lucas P.C. Yonezumi M. Inohara N. McAllister-Lucas L.M. Abazeed M.E. Chen F.F. Yamaoka S. Seto M. Nunez G. J. Biol. Chem. 2001; 276: 19012-19019Abstract Full Text Full Text PDF PubMed Scopus (379) Google Scholar) and is also required for TCR-induced proliferation, cytokine production, and NF-κB activation (24.Ruefli-Brasse A.A. French D.M. Dixit V.M. Science. 2003; 302: 1581-1584Crossref PubMed Scopus (304) Google Scholar). Our data are consistent with previous reports that Rip2-deficient mice suffer from defects in the adaptive immune response due to lack of antigen-induced T-cell proliferation and NF-κB activation (16.Kobayashi K. Inohara N. Hernandez L.D. Galan J.E. Nunez G. Janeway C.A. Medzhitov R. Flavell R.A. Nature. 2002; 416: 194-199Crossref PubMed Scopus (744) Google Scholar, 17.Chin A.I. Dempsey P.W. Bruhn K. Miller J.F. Xu Y. Cheng G. Nature. 2002; 416: 190-194Crossref PubMed Scopus (330) Google Scholar). Similar to published results, we also observed a defect in cytokine production in macrophages stimulated with LPS and other Toll-like receptors, demonstrating an additional defect in innate immunity (data not shown) (16.Kobayashi K. Inohara N. Hernandez L.D. Galan J.E. Nunez G. Janeway C.A. Medzhitov R. Flavell R.A. Nature. 2002; 416: 194-199Crossref PubMed Scopus (744) Google Scholar, 17.Chin A.I. Dempsey P.W. Bruhn K. Miller J.F. Xu Y. Cheng G. Nature. 2002; 416: 190-194Crossref PubMed Scopus (330) Google Scholar). Since Rip2 associates with key signaling molecules in both the adaptive and innate immune responses, such as Bcl10 and TRAF6 respectively, it is reasonable that the absence of this promiscuous kinase would impinge on multiple signaling pathways and result in broad ranging deficits in immune system function. We thank F. Martin, D. Dornan, N. Kayagaki, and the Dixit laboratory members for helpful discussions and review of this manuscript and S. Erickson, X. Sun, and the transgenic laboratory for the generation and care of animals used in this study." @default.
- W1978322477 created "2016-06-24" @default.
- W1978322477 creator A5043737419 @default.
- W1978322477 creator A5051300168 @default.
- W1978322477 creator A5055581115 @default.
- W1978322477 creator A5084210146 @default.
- W1978322477 date "2004-01-01" @default.
- W1978322477 modified "2023-09-27" @default.
- W1978322477 title "Rip2 Participates in Bcl10 Signaling and T-cell Receptor-mediated NF-κB Activation" @default.
- W1978322477 cites W1621883807 @default.
- W1978322477 cites W1975293480 @default.
- W1978322477 cites W1975870295 @default.
- W1978322477 cites W1977642281 @default.
- W1978322477 cites W1977830503 @default.
- W1978322477 cites W1988886903 @default.
- W1978322477 cites W1990735654 @default.
- W1978322477 cites W2016116811 @default.
- W1978322477 cites W2029295983 @default.
- W1978322477 cites W2033403762 @default.
- W1978322477 cites W2039253450 @default.
- W1978322477 cites W2041170922 @default.
- W1978322477 cites W2041928346 @default.
- W1978322477 cites W2043102057 @default.
- W1978322477 cites W2059685005 @default.
- W1978322477 cites W2066177415 @default.
- W1978322477 cites W2075626786 @default.
- W1978322477 cites W2085685430 @default.
- W1978322477 cites W2096977835 @default.
- W1978322477 cites W2106877484 @default.
- W1978322477 cites W2137823685 @default.
- W1978322477 cites W2159460252 @default.
- W1978322477 cites W2170552500 @default.
- W1978322477 doi "https://doi.org/10.1074/jbc.c300460200" @default.
- W1978322477 hasPubMedId "https://pubmed.ncbi.nlm.nih.gov/14638696" @default.
- W1978322477 hasPublicationYear "2004" @default.
- W1978322477 type Work @default.
- W1978322477 sameAs 1978322477 @default.
- W1978322477 citedByCount "88" @default.
- W1978322477 countsByYear W19783224772012 @default.
- W1978322477 countsByYear W19783224772013 @default.
- W1978322477 countsByYear W19783224772016 @default.
- W1978322477 countsByYear W19783224772017 @default.
- W1978322477 countsByYear W19783224772018 @default.
- W1978322477 countsByYear W19783224772019 @default.
- W1978322477 countsByYear W19783224772020 @default.
- W1978322477 countsByYear W19783224772021 @default.
- W1978322477 countsByYear W19783224772022 @default.
- W1978322477 countsByYear W19783224772023 @default.
- W1978322477 crossrefType "journal-article" @default.
- W1978322477 hasAuthorship W1978322477A5043737419 @default.
- W1978322477 hasAuthorship W1978322477A5051300168 @default.
- W1978322477 hasAuthorship W1978322477A5055581115 @default.
- W1978322477 hasAuthorship W1978322477A5084210146 @default.
- W1978322477 hasBestOaLocation W19783224771 @default.
- W1978322477 hasConcept C104317684 @default.
- W1978322477 hasConcept C107006841 @default.
- W1978322477 hasConcept C123298856 @default.
- W1978322477 hasConcept C170493617 @default.
- W1978322477 hasConcept C185592680 @default.
- W1978322477 hasConcept C2777730290 @default.
- W1978322477 hasConcept C502942594 @default.
- W1978322477 hasConcept C55493867 @default.
- W1978322477 hasConcept C62478195 @default.
- W1978322477 hasConcept C86339819 @default.
- W1978322477 hasConcept C86803240 @default.
- W1978322477 hasConcept C95444343 @default.
- W1978322477 hasConceptScore W1978322477C104317684 @default.
- W1978322477 hasConceptScore W1978322477C107006841 @default.
- W1978322477 hasConceptScore W1978322477C123298856 @default.
- W1978322477 hasConceptScore W1978322477C170493617 @default.
- W1978322477 hasConceptScore W1978322477C185592680 @default.
- W1978322477 hasConceptScore W1978322477C2777730290 @default.
- W1978322477 hasConceptScore W1978322477C502942594 @default.
- W1978322477 hasConceptScore W1978322477C55493867 @default.
- W1978322477 hasConceptScore W1978322477C62478195 @default.
- W1978322477 hasConceptScore W1978322477C86339819 @default.
- W1978322477 hasConceptScore W1978322477C86803240 @default.
- W1978322477 hasConceptScore W1978322477C95444343 @default.
- W1978322477 hasIssue "2" @default.
- W1978322477 hasLocation W19783224771 @default.
- W1978322477 hasOpenAccess W1978322477 @default.
- W1978322477 hasPrimaryLocation W19783224771 @default.
- W1978322477 hasRelatedWork W1523175586 @default.
- W1978322477 hasRelatedWork W1860642179 @default.
- W1978322477 hasRelatedWork W2005482505 @default.
- W1978322477 hasRelatedWork W2066868766 @default.
- W1978322477 hasRelatedWork W2166247944 @default.
- W1978322477 hasRelatedWork W2314353299 @default.
- W1978322477 hasRelatedWork W2365541820 @default.
- W1978322477 hasRelatedWork W2750891004 @default.
- W1978322477 hasRelatedWork W27859750 @default.
- W1978322477 hasRelatedWork W3029746177 @default.
- W1978322477 hasVolume "279" @default.
- W1978322477 isParatext "false" @default.
- W1978322477 isRetracted "false" @default.
- W1978322477 magId "1978322477" @default.
- W1978322477 workType "article" @default.