Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Blood Cancer J ; 5: e316, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26024286

ABSTRACT

Although several new therapeutic approaches have improved outcomes in the treatment of hematologic malignancies, unmet need persists in acute myeloid leukemia (AML), multiple myeloma (MM) and non-Hodgkin's lymphoma. Here we describe the proteomic identification of a novel cancer target, SAIL (Surface Antigen In Leukemia), whose expression is observed in AML, MM, chronic lymphocytic leukemia (CLL), diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL). While SAIL is widely expressed in CLL, AML, MM, DLBCL and FL patient samples, expression in cancer cell lines is mostly limited to cells of AML origin. We evaluated the antitumor activity of anti-SAIL monoclonal antibodies, 7-1C and 67-7A, conjugated to monomethyl auristatin F. Following internalization, anti-SAIL antibody-drug conjugates (ADCs) exhibited subnanomolar IC50 values against AML cell lines in vitro. In pharmacology studies employing AML cell line xenografts, anti-SAIL ADCs resulted in significant tumor growth inhibition. The restricted expression profile of this target in normal tissues, the high prevalence in different types of hematologic cancers and the observed preclinical activity support the clinical development of SAIL-targeted ADCs.


Subject(s)
Aminobenzoates/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/administration & dosage , Hematologic Neoplasms/drug therapy , Immunotherapy/methods , Oligopeptides/administration & dosage , Animals , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , In Situ Hybridization , Mice , Mice, SCID , Tandem Mass Spectrometry , Tissue Array Analysis , Xenograft Model Antitumor Assays
2.
Am J Transplant ; 12(10): 2754-62, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22883025

ABSTRACT

There are lines of evidence that B cells may play a role in transplantation. B cell activating factor, BAFF, is a homotrimer that has been shown to play a role in B cell survival, maturation and activation. To date, little is known of the role of BAFF and its receptors in transplantation. We analyzed the level of BAFF mRNA and its soluble protein, as well as transcripts coding for its receptors, BAFF-R, TACI and BCMA, in the blood of 143 patients with stable kidney transplant function 5 years or more posttransplantation. Three endpoints were analyzed: the time to renal dysfunction, the time to appearance of anti-HLA antibodies and the time to development of donor-specific antibodies. We established threshold values for BAFF and BAFF-R and showed that (1) stable patients with high BAFF-R levels had a higher risk of developing graft dysfunction, (2) patients with lower levels of BAFF transcripts or a higher level of soluble BAFF had a significantly higher risk of developing donor-specific antibodies. These data suggest that BAFF constitutes a risk factor for renal graft dysfunction and development of donor-specific antibodies. They also suggest that agents targeting BAFF-R interactions may offer new therapeutic opportunities in transplantation.


Subject(s)
Antibody Formation , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/metabolism , Kidney Transplantation , Tissue Donors , Female , Humans , Male , Risk Factors
3.
Mucosal Immunol ; 4(2): 172-85, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20980995

ABSTRACT

The tumor necrosis factor (TNF)-family cytokine TL1A (TNFSF15) costimulates T cells through its receptor DR3 (TNFRSF25) and is required for autoimmune pathology driven by diverse T-cell subsets. TL1A has been linked to human inflammatory bowel disease (IBD), but its pathogenic role is not known. We generated transgenic mice that constitutively express TL1A in T cells or dendritic cells. These mice spontaneously develop IL-13-dependent inflammatory small bowel pathology that strikingly resembles the intestinal response to nematode infections. These changes were dependent on the presence of a polyclonal T-cell receptor (TCR) repertoire, suggesting that they are driven by components in the intestinal flora. Forkhead box P3 (FoxP3)-positive regulatory T cells (Tregs) were present in increased numbers despite the fact that TL1A suppresses the generation of inducible Tregs. Finally, blocking TL1A-DR3 interactions abrogates 2,4,6 trinitrobenzenesulfonic acid (TNBS) colitis, indicating that these interactions influence other causes of intestinal inflammation as well. These results establish a novel link between TL1A and interleukin 13 (IL-13) responses that results in small intestinal inflammation, and also establish that TL1A-DR3 interactions are necessary and sufficient for T cell-dependent IBD.


Subject(s)
Enteritis/immunology , Interleukin-13/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , CD2 Antigens/genetics , CD2 Antigens/immunology , Colitis/immunology , Colitis/pathology , Dendritic Cells/immunology , Dose-Response Relationship, Immunologic , Enteritis/pathology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/immunology , Gene Order , HEK293 Cells , Humans , Immunologic Memory/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-13/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Receptors, Tumor Necrosis Factor, Member 25/metabolism , T-Lymphocytes
4.
Ann Rheum Dis ; 67(8): 1132-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-17967830

ABSTRACT

OBJECTIVE: To assess the effects of tumour necrosis factor (TNF) antagonist therapy on B lymphocyte stimulator (BLyS) expression in patients with rheumatoid arthritis (RA). METHODS: Blood from 38 patients with RA from a single centre was collected prior to and following initiation of TNF antagonist therapy. Plasma BLyS protein levels, blood leukocyte BLyS mRNA levels and disease activity were longitudinally monitored. Twelve patients with RA who either refused or were felt not to be candidates for TNF antagonist therapy and five normal healthy volunteers served as TNF antagonist-naïve controls. RESULTS: Baseline plasma BLyS protein levels, but not blood leukocyte BLyS mRNA levels, were elevated in patients with RA. Plasma BLyS protein levels declined following initiation of TNF antagonist therapy in good responders (GR) to TNF antagonist therapy but not in poor responders (PR). By contrast, the erythrocyte sedimentation rate (ESR) declined in response to TNF antagonist therapy in GR and PR. TNF antagonist therapy did not promote change in blood leukocyte BLyS mRNA levels in either GR or PR, suggesting that the TNF antagonist-associated changes in circulating BLyS protein levels reflected changes in local BLyS production in the affected joints rather than changes in systemic BLyS production. BLyS expression did not change over time in either the normal or RA control groups. CONCLUSIONS: A good clinical response to TNF antagonist therapy in patients with RA is associated with a decline in plasma BLyS protein levels. Increased BLyS expression in affected joints may contribute to ongoing disease activity, and reduction of such expression may help promote a favourable clinical response to TNF antagonist therapy.


Subject(s)
Arthritis, Rheumatoid/immunology , B-Cell Activating Factor/blood , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , B-Cell Activating Factor/genetics , Blood Sedimentation , Case-Control Studies , Chi-Square Distribution , Female , Gene Expression , Humans , Leukocytes/immunology , Male , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Treatment Outcome
5.
Ann Rheum Dis ; 67(7): 1011-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17962238

ABSTRACT

OBJECTIVE: To assess the relationships between serum B lymphocyte stimulator (BLyS) levels, autoantibody profile and clinical response in patients with systemic lupus erythematosus (SLE) following rituximab-based B cell depletion therapy (BCDT). METHODS: A total of 25 patients with active refractory SLE were followed for >or=1 year following BCDT. Disease activity was assessed using the British Isles Lupus Assessment Group (BILAG) system, and serum levels of BLyS and autoantibodies to dsDNA and extractable nuclear antigens (ENA) measured by ELISA. Serum immunoglobulins and anti-dsDNA antibodies were assessed for expression of the 9G4 idiotope (indicating VH4-34 germline gene origin). RESULTS: Following BCDT, all patients depleted in the peripheral blood and improved clinically for >or=3 months. Pre-BCDT BLyS levels were quantifiable (median 1.9 ng/ml) in 18/25 patients and rose in most patients at 3 months post-BCDT (median 4.15 ng/ml). Nine patients, all with quantifiable pre-BCDT serum BLyS, experienced a disease flare within 1 year. This group of patients was more likely to harbour anti-Ro/SSA antibodies (odds ratio 1.76; p = 0.06) with higher serum levels (p = 0.0027; Mann-Whitney U test). Serum levels of anti-ribonucleoprotein (RNP)/Sm were also higher in this group (p<0.05). Expression of VH4-34 by serum immunoglobulins and anti-dsDNA antibodies had no predictive value for the length of clinical response. CONCLUSIONS: Patients with SLE with an expanded autoantibody profile and raised BLyS levels at baseline had shorter clinical responses to BCDT. This may reflect a greater propensity to, and degree of, epitope spreading in such patients and suggests that treatment regimens beyond BCDT may be necessary to induce long-lasting clinical remissions in these individuals.


Subject(s)
Autoantibodies/blood , B-Cell Activating Factor/blood , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/drug therapy , Lymphocyte Depletion/methods , Antibodies, Antinuclear/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Antirheumatic Agents/therapeutic use , Follow-Up Studies , Genes, Immunoglobulin Heavy Chain/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Lupus Erythematosus, Systemic/immunology , Lymphocyte Count , Recurrence , Rituximab , Severity of Illness Index , Time Factors , Treatment Outcome
6.
Rheumatology (Oxford) ; 46(1): 37-43, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16735452

ABSTRACT

OBJECTIVES: To investigate the role of B-Lymphocyte stimulator (BLyS) in mixed cryoglobulinaemia syndrome (MCsn), a systemic vasculitis associated with a high risk to develop lymphoma, since BLyS up-regulation may favour both autoimmunity and lymphoproliferation. METHODS: BLyS serum levels were analysed by enzyme-linked immunosorbent assay (positive when >0.85 ng/ml) in 66 patients with MCsn, 54 (81.8%) of whom were positive for hepatitis-C virus (HCV) infection. Thirty-three HCV-positive patients without MCsn were also studied. Patients were compared with 48 healthy blood donors (HBDs). BLyS modifications after antiviral therapy were also studied. RESULTS: A significantly higher frequency of BLyS serum positivity was detected both in MCsn patients and in HCV-positive patients without MCsn (37.9 and 30.3%, respectively) when compared with HBDs (4.2%) (P < 0.0001 vs MCsn and P = 0.0026 vs HCV-positive patients without MCsn, respectively). BLyS appeared significantly higher in MCsn (3.70 +/- 2.97 ng/ml) than in HCV-positive patients without MCsn (1.56 +/- 0.63 ng/ml; P = 0.0044). BLyS expression did not correlate with rheumatoid factor levels, cryoglobulin levels or definite MCsn-related systemic features. High BLyS levels were significantly associated only with MCsn-related overt lymphoproliferative disorder. Finally, antiviral treatment significantly increased BLyS levels, independently from HCV-RNA negativization. However, BLyS normalization was noticed after both HCV-RNA negativization and suspension of antiviral therapy by preliminary data. CONCLUSIONS: BLyS is up-regulated and may play a pathogenetic role in a fraction of patients with MCsn, similarly to other autoimmune diseases. HCV infection likely represents the early event leading to BLyS up-regulation in this setting. BLyS is up-regulated during antiviral treatment. Overall, these data provide new insights for BLyS and virus-related autoimmunity, lymphoproliferation and possible treatment strategies.


Subject(s)
Autoimmune Diseases/immunology , B-Cell Activating Factor/blood , Cryoglobulinemia/immunology , Hepatitis C/immunology , Up-Regulation , Adult , Aged , Aged, 80 and over , Antiviral Agents/therapeutic use , Autoimmune Diseases/virology , Cryoglobulinemia/virology , Enzyme-Linked Immunosorbent Assay/methods , Female , Hepatitis C/complications , Hepatitis C/drug therapy , Humans , Male , Middle Aged , Syndrome , Up-Regulation/drug effects
7.
Blood ; 98(6): 1935-41, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11535532

ABSTRACT

Cytokines, such as interleukin-2 (IL-2), activate intracellular signaling pathways via rapid tyrosine phosphorylation of their receptors, resulting in the activation of many genes involved in cell growth and survival. The deubiquitinating enzyme DUB-2 is induced in response to IL-2 but as yet its function has not been determined. The results of this study show that DUB-2 is expressed in human T-cell lymphotropic virus-I (HTLV-1)-transformed T cells that exhibit constitutive activation of the IL-2 JAK/STAT (signal transducers and activators of transcription) pathway, and when expressed in Ba/F3 cells DUB-2 markedly prolonged IL-2-induced STAT5 phosphorylation. Although DUB-2 did not enhance IL-2-mediated proliferation, when withdrawn from growth factor, cells expressing DUB-2 had sustained STAT5 phosphorylation and enhanced expression of IL-2-induced genes cis and c-myc. Moreover, DUB-2 expression markedly inhibited apoptosis induced by cytokine withdrawal allowing cells to survive. Taken together these data suggest that DUB-2 can enhance signaling through the JAK/STAT pathway, prolong lymphocyte survival, and, when constitutively expressed, may contribute to the activation of the JAK/STAT pathway observed in some transformed cells.


Subject(s)
Apoptosis , Cell Transformation, Viral , Endopeptidases , Immediate-Early Proteins/physiology , Interleukin-2/pharmacology , Leukemia-Lymphoma, Adult T-Cell/metabolism , Milk Proteins , Signal Transduction , Cell Line , Cell Line, Transformed , Cysteine Endopeptidases , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Human T-lymphotropic virus 1/pathogenicity , Humans , Immediate-Early Proteins/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Leupeptins/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Phosphorylation , Proteasome Endopeptidase Complex , STAT5 Transcription Factor , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Trans-Activators/metabolism , Trans-Activators/physiology , Transcriptional Activation , Transfection , Ubiquitins/metabolism
8.
Cytokine ; 13(1): 25-31, 2001 Jan 07.
Article in English | MEDLINE | ID: mdl-11145839

ABSTRACT

B lymphocyte stimulator (BLyS) is a novel member of the TNF family of proteins expressed by myeloid cells as membrane-bound and soluble forms. BLyS was shown to act specifically on B cells, inducing proliferation and immunoglobulin production both in vitro and in vivo. The present study was undertaken to characterize binding of radiolabeled BLyS to its cognate receptor on human B lymphocytes and examine intracellular events initiated by BLyS binding. Similar to other TNF family members, BLyS is present in solution as a homotrimer as determined by gel filtration chromatography and light scattering analysis. BLyS binding to B cells is specific as other TNF family members tested did not compete for(125)I-BLyS binding. Analysis of equilibrium binding of(125)I-labeled BLyS to purified human tonsillar B cells demonstrated saturable binding. Scatchard analysis of the binding data revealed a single class of high-affinity binding on human B cells with approximately 2600 binding sites per cell and an apparent dissociation constant (K(D)) of about 0.1 nM. In addition we report that BLyS binding to B cells results in the activation of NF-kappaB and the Ets family transcription factor, ELF-1, and in the induction of mRNA for Polo-like kinase (PLK).


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , NF-kappa B/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , B-Cell Activating Factor , Binding Sites , Cross-Linking Reagents , Humans , In Vitro Techniques , Iodine Radioisotopes , Kinetics , Membrane Proteins/chemistry , Protein Binding , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/chemistry
9.
Blood ; 97(1): 198-204, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11133761

ABSTRACT

B-lymphocyte stimulator (BLyS) is a recently identified novel member of the tumor necrosis factor ligand superfamily shown to exist in a membrane-bound and soluble form. BLyS was found to be specifically expressed on cells of myeloid lineage and to selectively stimulate B-lymphocyte proliferation and immunoglobulin production. The expression of a cytokine involved in potentiation of humoral immune responses, such as BLyS, is expected to be strictly controlled. The goal of the present study was to examine regulation of BLyS levels in monocytic cells in response to cytokines and during their differentiation to macrophages and dendritic cells. The presence of BLyS on the cell surface and in the culture medium of both normal blood monocytes and on tumor cells of myelomonocytic origin was demonstrated. BLyS gene expression and levels of membrane-associated and soluble BLyS were found to be regulated by cytokines, in particular interferon (IFN)-gamma and to a lesser extent interleukin-10 (IL-10). The expression of BLyS on monocyte membranes was retained following differentiation into macrophages, but detection on the surface of monocyte-derived dendritic cells required stimulation with IFN-gamma. Both IFN-gamma and IL-10 enhanced the release of soluble BLyS that was active in B-cell proliferation assays. Cells transfected with BLyS complementary DNA mutated in a predicted cleavage site failed to release BLyS into the culture medium, thereby suggesting that soluble BLyS was derived from the membrane form. These results provide further support for an important role for BLyS expressed in myeloid cells in B-cell expansion and antibody responses.


Subject(s)
Membrane Proteins/genetics , Myeloid Cells/metabolism , Tumor Necrosis Factor-alpha/genetics , Antibodies/metabolism , B-Cell Activating Factor , B-Lymphocytes/cytology , Cell Division/drug effects , Cytokines/pharmacology , Dendritic Cells/chemistry , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Macrophages/chemistry , Membrane Proteins/immunology , Membrane Proteins/metabolism , Monocytes/metabolism , Peptide Hydrolases/metabolism , RNA, Messenger/metabolism , Solubility , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
10.
J Biol Chem ; 275(45): 35478-85, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-10956646

ABSTRACT

An expression cloning approach was employed to identify the receptor for B-lymphocyte stimulator (BLyS) and identified the tumor necrosis factor receptor superfamily member TACI as a BLyS-binding protein. Expression of TACI in HEK293T cells confers on the cells the ability to bind BLyS with subnanomolar affinity. Furthermore, a TACI-Fc fusion protein recognizes both the cleaved, soluble form of BLyS as well as the membrane BLyS present on the cell surface of a recombinant cell line. TACI mRNA is found predominantly in B-cells and correlates with BLyS binding in a panel of B-cell lines. We also demonstrate that TACI interacts with nanomolar affinity with the BLyS-related tumor necrosis factor homologue APRIL for which no clear in vivo role has been described. BLyS and APRIL are capable of signaling through TACI to mediate NF-kappaB responses in HEK293 cells. We conclude that TACI is a receptor for BLyS and APRIL and discuss the implications for B-cell biology.


Subject(s)
B-Lymphocytes/physiology , Membrane Proteins , Neuropeptides/physiology , Nuclear Proteins/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/chemistry , B-Cell Activation Factor Receptor , B-Lymphocytes/metabolism , Cell Line , Cell Membrane/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Gene Library , Humans , Kinetics , Ligands , Polymerase Chain Reaction , Protein Binding , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transfection , Transmembrane Activator and CAML Interactor Protein
11.
J Biol Chem ; 274(42): 30266-72, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10514520

ABSTRACT

CIS is a cytokine-induced SH2-containing protein that was originally cloned as an interleukin (IL)-3-inducible gene. CIS is known to associate with the IL-3 receptor beta chain and erythropoietin receptor and to inhibit signaling mediated by IL-3 and erythropoietin. We now demonstrate that CIS also interacts with the IL-2 receptor beta chain (IL-2Rbeta). This interaction requires the A region of IL-2Rbeta (residues 313-382), which also mediates the association of IL-2Rbeta with Lck and Jak3. Correspondingly, CIS inhibits functions associated with both of these kinases: Lck-mediated phosphorylation of IL-2Rbeta and IL-2-mediated activation of Stat5. Thus, we demonstrate that CIS can negatively control at least two independent IL-2 signaling pathways. Although a functional SH2 binding domain of CIS was not required for its interaction with IL-2Rbeta in vitro, its phosphotyrosine binding capability was essential for the inhibitory action of CIS. On this basis, we have generated a mutant form of CIS protein with an altered SH2 domain that acts as a dominant negative and should prove useful in further understanding CIS action.


Subject(s)
Immediate-Early Proteins/metabolism , Interleukin-2/antagonists & inhibitors , Milk Proteins , Receptors, Interleukin-2/metabolism , Signal Transduction , Amino Acid Sequence , Cell Line , DNA-Binding Proteins/metabolism , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Interleukin-2/metabolism , Janus Kinase 1 , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , STAT5 Transcription Factor , Suppressor of Cytokine Signaling Proteins , Trans-Activators/metabolism , Tyrosine/metabolism , src Homology Domains
12.
Mol Cell Biol ; 19(7): 4980-8, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10373548

ABSTRACT

Members of the recently discovered SOCS/CIS/SSI family have been proposed as regulators of cytokine signaling, and while targets and mechanisms have been suggested for some family members, the precise role of these proteins remains to be defined. To date no SOCS proteins have been specifically implicated in interleukin-2 (IL-2) signaling in T cells. Here we report SOCS-3 expression in response to IL-2 in both T-cell lines and human peripheral blood lymphocytes. SOCS-3 protein was detectable as early as 30 min following IL-2 stimulation, while CIS was seen only at low levels after 2 h. Unlike CIS, SOCS-3 was rapidly tyrosine phosphorylated in response to IL-2. Tyrosine phosphorylation of SOCS-3 was observed upon coexpression with Jak1 and Jak2 but only weakly with Jak3. In these experiments, SOCS-3 associated with Jak1 and inhibited Jak1 phosphorylation, and this inhibition was markedly enhanced by the presence of IL-2 receptor beta chain (IL-2Rbeta). Moreover, following IL-2 stimulation of T cells, SOCS-3 was able to interact with the IL-2 receptor complex, and in particular tyrosine phosphorylated Jak1 and IL-2Rbeta. Additionally, in lymphocytes expressing SOCS-3 but not CIS, IL-2-induced tyrosine phosphorylation of STAT5b was markedly reduced, while there was only a weak effect on IL-3-mediated STAT5b tyrosine phosphorylation. Finally, proliferation induced by both IL-2- and IL-3 was significantly inhibited in the presence of SOCS-3. The findings suggest that when SOCS-3 is rapidly induced by IL-2 in T cells, it acts to inhibit IL-2 responses in a classical negative feedback loop.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-2/metabolism , Milk Proteins , Proteins/metabolism , Repressor Proteins , T-Lymphocytes/cytology , Trans-Activators/metabolism , Transcription Factors , Tyrosine/metabolism , Animals , Cell Division , Cell Line , Cell Line, Transformed , Humans , Interleukin-2/pharmacology , Interleukin-3/metabolism , Janus Kinase 1 , Janus Kinase 3 , Phosphorylation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proteins/genetics , Rabbits , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
13.
EMBO J ; 18(6): 1549-58, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10075926

ABSTRACT

Signaling through the hematopoietic receptors requires activation of receptor-associated Janus (Jak) kinases. For example, Jak1 and Jak3 bind specifically to the IL-2 receptor beta (IL-2Rbeta) and common gamma (gammac) chains, respectively, and initiate biochemical signals critical in controlling immune responses. The region of Jak responsible for receptor interactions, however, is not well characterized. Here we describe a naturally occurring Jak3 mutation from a patient with autosomal severe combined immunodeficiency (SCID), where a single amino acid substitution, Y100C, in Janus homology domain 7 (JH7) prevents kinase-receptor interaction. This mutation also results in a loss of IL-2-induced signaling in a B-cell line derived from this patient. Using mutational analysis we have identified a region of Jak3, including portions of JH6 and JH7, that is sufficient for kinase-receptor contact and show that this segment interacts with the proline-rich Box1 region of the receptor. Furthermore, a Jak3-Jak1 chimera containing only the JH6 and JH7 domains of Jak3 interacts with gammac and can reconstitute IL-2-dependent responses, including receptor phosphorylation and activation of signal transducer and activator of transcription (STAT) 5b. Our results suggest that the N-terminus of Jak kinases is critical for receptor binding, and is therefore likely to determine specificity of Jak kinase-receptor interactions.


Subject(s)
Point Mutation , Protein-Tyrosine Kinases/genetics , Severe Combined Immunodeficiency/genetics , Cell Line , Humans , Janus Kinase 1 , Janus Kinase 3 , Polymerase Chain Reaction , Protein Binding , Protein-Tyrosine Kinases/metabolism , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/physiology , Recombinant Fusion Proteins/metabolism , Severe Combined Immunodeficiency/enzymology , Transfection
14.
Mol Cell Biol ; 18(11): 6416-22, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9774657

ABSTRACT

Phosphatidylinositol 3-kinase (PI 3-K) plays an important role in signaling via a wide range of receptors such as those for antigen, growth factors, and a number of cytokines, including interleukin-2 (IL-2). PI 3-K has been implicated in both IL-2-induced proliferation and prevention of apoptosis. A number of potential mechanisms for the recruitment of PI 3-K to the IL-2 receptor have been proposed. We now have found that tyrosine residues in the IL-2 receptor beta chain (IL-2Rbeta) are unexpectedly not required for the recruitment of the p85 component of PI 3-K. Instead, we find that Jak1, which associates with membrane-proximal regions of the IL-2Rbeta cytoplasmic domain, is essential for efficient IL-2Rbeta-p85 interaction, although some IL-2Rbeta-p85 association can be seen in the absence of Jak1. We also found that Jak1 interacts with p85 in the absence of IL-2Rbeta and that IL-2Rbeta and Jak1 cooperate for the efficient recruitment and tyrosine phosphorylation of p85. This is the first report of a PI 3-K-Jak1 interaction, and it implicates Jak1 in an essential IL-2 signaling pathway distinct from the activation of STAT proteins.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Interleukin-2/chemistry , Cell Line , Humans , Interleukin-2/physiology , Janus Kinase 1 , Phosphorylation , Phosphotyrosine/analysis , Signal Transduction/physiology , src Homology Domains/physiology
15.
Proc Natl Acad Sci U S A ; 95(7): 3845-50, 1998 Mar 31.
Article in English | MEDLINE | ID: mdl-9520455

ABSTRACT

Interleukin 2 (IL-2) rapidly induces tyrosine phosphorylation of intracellular substrates, including the IL-2 receptor beta chain (IL-2Rbeta), Janus kinase 1 (Jak1), Jak3, signal transducer/activator of transcription proteins, and Shc, but the mechanism underlying dephosphorylation of these proteins is not known. The src homology 2 (SH2) containing tyrosine phosphatase 1 (SHP-1) is recruited by several hematopoietic surface receptors indicating that this phosphatase plays an important role as a regulator of signaling. We have found that IL-2 induces association of SHP-1 with the IL-2 receptor complex, and that once SHP-1 is recruited to the activated receptor it is able to decrease tyrosine phosphorylation of IL-2Rbeta and the associated tyrosine kinases Jak1 and Jak3. This dephosphorylation is specific as expression of a catalytically inactive form of SHP-1, or expression of the related phosphatase SHP-2 did not result in dephosphorylation of the IL-2 receptor components. Furthermore, we have found that SHP-1 expression is greatly decreased or undetectable in a number of IL-2 independent HTLV-I transformed T cell lines that exhibit constitutive Jak/signal transducer/activator of transcription activation. In HTLV-I infected T cells, down-regulation of SHP-1 expression was also found to correlate with the acquisition of IL-2 independence. These observations suggest that SHP-1 normally functions to antagonize the IL-2 signal transduction pathway and that HTLV-I infection and oncogenic transformation can lead to loss of SHP-1 expression resulting in constitutive activation of IL-2 regulated T cell responses.


Subject(s)
Cell Transformation, Viral/immunology , Gene Expression Regulation/immunology , Human T-lymphotropic virus 1 , Interleukin-2/immunology , Protein Tyrosine Phosphatases/immunology , Receptors, Interleukin-2/immunology , T-Lymphocytes/immunology , Down-Regulation , Humans , Intracellular Signaling Peptides and Proteins , Phosphorylation , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/biosynthesis , Receptors, Interleukin-2/biosynthesis , SH2 Domain-Containing Protein Tyrosine Phosphatases , Signal Transduction/immunology , T-Lymphocytes/virology , src Homology Domains
16.
J Virol ; 72(5): 4408-12, 1998 May.
Article in English | MEDLINE | ID: mdl-9557732

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) and HTLV-2 differ in pathogenicity in vivo. HTLV-1 causes leukemia and neurologic and inflammatory diseases, whereas HTLV-2 is less clearly associated with human disease. Both retroviruses transform human T cells in vitro, and transformation by HTLV-1 was found to be associated with the constitutive activation of the Jak/STAT pathway. To assess whether HTLV-2 transformation may also result in constitutive activation of the Jak/STAT pathway, six interleukin-2-independent, HTLV-2-transformed T-cell lines were analyzed for the presence of activated Jak and STAT proteins by electrophoretic mobility shift assay. In addition, the phosphorylation status of Jak and STAT proteins was assessed directly by immunoprecipitation and immunoblotting with an antiphosphotyrosine antibody. Jak/STAT proteins were not found to be constitutively activated in any of the T-cell lines infected by the type 2 human and nonhuman primate viruses, suggesting that HTLV-2 and the cognate virus simian T-lymphotropic virus type 2 from Pan paniscus transform T cells in vitro by mechanisms at least partially different from those used by HTLV-1.


Subject(s)
Cell Transformation, Viral , DNA-Binding Proteins/metabolism , Human T-lymphotropic virus 2/physiology , Milk Proteins , Protein-Tyrosine Kinases/metabolism , Simian T-lymphotropic virus 1/physiology , Trans-Activators/metabolism , Animals , Caseins/genetics , Cell Line, Transformed , Genes, fos , Haplorhini , Human T-lymphotropic virus 1/physiology , Humans , Janus Kinase 1 , Janus Kinase 3 , Phenotype , Promoter Regions, Genetic , Receptors, IgG/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor
17.
J Biol Chem ; 272(13): 8704-9, 1997 Mar 28.
Article in English | MEDLINE | ID: mdl-9079703

ABSTRACT

The interleukin-2 receptor alpha chain (IL-2Ralpha) is potently induced by antigens, mitogens, and certain cytokines that include IL-2 itself. This induction leads to the formation of high affinity IL-2 receptors when IL-2Ralpha is co-expressed with the beta (IL-2Rbeta) and gamma (gammac) chains of this receptor. We investigated the signaling pathways mediating IL-2-induced IL-2Ralpha mRNA expression using 32D myeloid progenitor cells stably transfected with either wild type IL-2Rbeta or mutants of IL-2Rbeta containing tyrosine to phenylalanine substitutions. Of the six cytoplasmic tyrosines in IL-2Rbeta, we have found that only the two tyrosines that mediate Stat5 activation (Tyr-392 and Tyr-510) contribute to IL-2-induced IL-2Ralpha gene expression and that either tyrosine alone is sufficient for this process. Interestingly, the IL-7 receptor contains a tyrosine (Tyr-429)-based sequence resembling the motifs encompassing Tyr-392 and Tyr-510 of IL-2Rbeta. Further paralleling the IL-2 system, IL-7 could activate Stat5 and drive expression of IL-2Ralpha mRNA in 32D cells transfected with the human IL-7R. However, IL-3 could not induce IL-2Ralpha mRNA in 32D cells, despite its ability to activate Stat5 via the endogenous IL-3 receptor. Moreover, the combination of IL-3 and IL-2 could not "rescue" IL-2Ralpha mRNA expression in cells containing an IL-2Rbeta mutant with phenylalanine substitutions at Tyr-392 and Tyr-510. These data suggest that Tyr-392 and Tyr-510 couple to an additional signaling pathway beyond STAT protein activation in IL-2-mediated induction of the IL-2Ralpha gene.


Subject(s)
DNA-Binding Proteins/metabolism , Milk Proteins , Receptors, Interleukin-2/genetics , Trans-Activators/metabolism , Tyrosine , Antigens, CD/chemistry , Antigens, CD/metabolism , DNA-Binding Proteins/biosynthesis , Humans , Interleukin-2/metabolism , Interleukin-3/metabolism , Interleukin-7/metabolism , RNA, Messenger/metabolism , Receptors, Interleukin/chemistry , Receptors, Interleukin/metabolism , Receptors, Interleukin-2/chemistry , Receptors, Interleukin-7 , STAT5 Transcription Factor , Trans-Activators/biosynthesis , Transcription, Genetic , Transfection
18.
Proc Natl Acad Sci U S A ; 94(25): 13897-902, 1997 Dec 09.
Article in English | MEDLINE | ID: mdl-9391124

ABSTRACT

Human T cell leukemia/lymphotropic virus type I (HTLV-I) induces adult T cell leukemia/lymphoma (ATLL). The mechanism of HTLV-I oncogenesis in T cells remains partly elusive. In vitro, HTLV-I induces ligand-independent transformation of human CD4+ T cells, an event that correlates with acquisition of constitutive phosphorylation of Janus kinases (JAK) and signal transducers and activators of transcription (STAT) proteins. However, it is unclear whether the in vitro model of HTLV-I transformation has relevance to viral leukemogenesis in vivo. Here we tested the status of JAK/STAT phosphorylation and DNA-binding activity of STAT proteins in cell extracts of uncultured leukemic cells from 12 patients with ATLL by either DNA-binding assays, using DNA oligonucleotides specific for STAT-1 and STAT-3, STAT-5 and STAT-6 or, more directly, by immunoprecipitation and immunoblotting with anti-phosphotyrosine antibody for JAK and STAT proteins. Leukemic cells from 8 of 12 patients studied displayed constitutive DNA-binding activity of one or more STAT proteins, and the constitutive activation of the JAK/STAT pathway was found to persist over time in the 2 patients followed longitudinally. Furthermore, an association between JAK3 and STAT-1, STAT-3, and STAT-5 activation and cell-cycle progression was demonstrated by both propidium iodide staining and bromodeoxyuridine incorporation in cells of four patients tested. These results imply that JAK/STAT activation is associated with replication of leukemic cells and that therapeutic approaches aimed at JAK/STAT inhibition may be considered to halt neoplastic growth.


Subject(s)
DNA-Binding Proteins/metabolism , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Milk Proteins , Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , Adult , Base Sequence , Cell Division , DNA Probes/genetics , Enzyme Activation , Human T-lymphotropic virus 1/pathogenicity , Humans , Janus Kinase 3 , Leukemia-Lymphoma, Adult T-Cell/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , Time Factors
19.
Immunity ; 5(1): 81-9, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8758897

ABSTRACT

The development of murine plasma cell tumors induced by raf/myc containing retroviruses is facilitated by T cells and completely dependent on IL-6. To determine whether kinases with differing specificities reflect alternative biochemical pathways in B cell tumorigenesis, we have employed an abl/myc containing retrovirus to assess neoplastic development. In contrast with raf/myc, abl/myc disease is T cell and IL-6 independent. An examination of the IL-6 signal transduction pathway reveals that this pathway, as defined by activation of Stat3, is inducible by IL-6 in raf/myc tumors but constitutively activated in abl/myc tumors. These findings provide a mechanism for the derivation of cytokine-independent plasma cell tumors and suggest that both IL-6-dependent and independent tumors may arise in vivo depending on the particular mutational events incurred during tumorigenesis.


Subject(s)
Genes, abl/immunology , Interleukin-6/immunology , Plasmacytoma/genetics , Retroviridae Infections/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Virus Infections/genetics , Animals , Base Sequence , Interleukin-6/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Nude , Molecular Sequence Data , Phenotype , Plasmacytoma/immunology , Retroviridae Infections/immunology , Tumor Virus Infections/immunology
20.
Proc Natl Acad Sci U S A ; 93(5): 2077-82, 1996 Mar 05.
Article in English | MEDLINE | ID: mdl-8700888

ABSTRACT

One of the earliest events induced by interleukin 2 (IL-2) is tyrosine phosphorylation of cellular proteins, including the IL-2 receptor beta chain (IL-2Rbeta). Simultaneous mutation of three tyrosines (Y338, Y392, and Y510) in the IL-2Rbeta cytoplasmic domain abrogated IL-2-induced proliferation, whereas mutation of only Y338 or of Y392 and Y510 inhibited proliferation only partially. While Y392 and Y510 were critical for IL-2-induced activation of signal transducers and activators of transcription (STAT proteins), Y338 was required for Shc-IL-2Rbeta association and for IL-2-induced tyrosine phosphorylation of Shc. Thus, activation of both Jak-STAT and Shc-coupled signaling pathways requires specific IL-2Rbeta tyrosines that together act in concert to mediate maximal proliferation. In COS-7 cells, overexpression of Jak1 augmented phosphorylation of Y338 as well as Y392 and Y510, suggesting that the role for this Jak kinase may extend beyond the Jak-STAT pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Lymphocyte Activation , Receptors, Interleukin-2/physiology , Signal Transduction , T-Lymphocytes/physiology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Humans , Janus Kinase 1 , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphotyrosine/chemistry , Protein Binding , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Receptor Aggregation , Receptors, Interleukin-2/chemistry , STAT1 Transcription Factor , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Trans-Activators/metabolism , Tyrosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...