Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1106537, 2023.
Article in English | MEDLINE | ID: mdl-36845162

ABSTRACT

Autoimmune diseases vary in the magnitude and diversity of autoantibody profiles, and these differences may be a consequence of different types of breaks in tolerance. Here, we compared the disparate autoimmune diseases autoimmune polyendocrinopathy-candidiasis-ecto-dermal dystrophy (APECED), systemic lupus erythematosus (SLE), and Sjogren's syndrome (SjS) to gain insight into the etiology of breaks in tolerance triggering autoimmunity. APECED was chosen as a prototypical monogenic disease with organ-specific pathology while SjS and SLE represent polygenic autoimmunity with focal or systemic disease. Using protein microarrays for autoantibody profiling, we found that APECED patients develop a focused but highly reactive set of shared mostly anti-cytokine antibodies, while SLE patients develop broad and less expanded autoantibody repertoires against mostly intracellular autoantigens. SjS patients had few autoantibody specificities with the highest shared reactivities observed against Ro-52 and La. RNA-seq B-cell receptor analysis revealed that APECED samples have fewer, but highly expanded, clonotypes compared with SLE samples containing a diverse, but less clonally expanded, B-cell receptor repertoire. Based on these data, we propose a model whereby the presence of autoreactive T-cells in APECED allows T-dependent B-cell responses against autoantigens, while SLE is driven by breaks in peripheral B-cell tolerance and extrafollicular B-cell activation. These results highlight differences in the autoimmunity observed in several monogenic and polygenic disorders and may be generalizable to other autoimmune diseases.


Subject(s)
Autoimmune Diseases , Lupus Erythematosus, Systemic , Polyendocrinopathies, Autoimmune , Sjogren's Syndrome , Humans , Autoantibodies , Autoimmune Diseases/genetics , Autoimmune Diseases/complications , Autoantigens , Receptors, Antigen, B-Cell
2.
Clin Cancer Res ; 21(11): 2591-600, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25733599

ABSTRACT

PURPOSE: Prostate cancer aggressiveness and appropriate therapy are routinely determined following biopsy sampling. Current clinical and pathologic parameters are insufficient for accurate risk prediction leading primarily to overtreatment and also missed opportunities for curative therapy. EXPERIMENTAL DESIGN: An 8-biomarker proteomic assay for intact tissue biopsies predictive of prostate pathology was defined in a study of 381 patient biopsies with matched prostatectomy specimens. A second blinded study of 276 cases validated this assay's ability to distinguish "favorable" versus "nonfavorable" pathology independently and relative to current risk classification systems National Comprehensive Cancer Network (NCCN and D'Amico). RESULTS: A favorable biomarker risk score of ≤0.33, and a nonfavorable risk score of >0.80 (possible range between 0 and 1) were defined on "false-negative" and "false-positive" rates of 10% and 5%, respectively. At a risk score ≤0.33, predictive values for favorable pathology in very low-risk and low-risk NCCN and low-risk D'Amico groups were 95%, 81.5%, and 87.2%, respectively, higher than for these current risk classification groups themselves (80.3%, 63.8%, and 70.6%, respectively). The predictive value for nonfavorable pathology was 76.9% at biomarker risk scores >0.8 across all risk groups. Increased biomarker risk scores correlated with decreased frequency of favorable cases across all risk groups. The validation study met its two coprimary endpoints, separating favorable from nonfavorable pathology (AUC, 0.68; P < 0.0001; OR, 20.9) and GS-6 versus non-GS-6 pathology (AUC, 0.65; P < 0.0001; OR, 12.95). CONCLUSIONS: The 8-biomarker assay provided individualized, independent prognostic information relative to current risk stratification systems, and may improve the precision of clinical decision making following prostate biopsy.


Subject(s)
Biomarkers, Tumor/biosynthesis , Neoplasm Recurrence, Local/genetics , Prognosis , Prostatic Neoplasms/genetics , Aged , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Proteomics , Risk Assessment
3.
PLoS One ; 6(4): e19169, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21552520

ABSTRACT

Activating mutations of FMS-like tyrosine kinase-3 (FLT3) are found in approximately 30% of patients with acute myeloid leukemia (AML). FLT3 is therefore an attractive drug target. However, the molecular mechanisms by which FLT3 mutations lead to cell transformation in AML remain unclear. To develop a better understanding of FLT3 signaling as well as its downstream effectors, we performed detailed phosphoproteomic analysis of FLT3 signaling in human leukemia cells. We identified over 1000 tyrosine phosphorylation sites from about 750 proteins in both AML (wild type and mutant FLT3) and B cell acute lymphoblastic leukemia (normal and amplification of FLT3) cell lines. Furthermore, using stable isotope labeling by amino acids in cell culture (SILAC), we were able to quantified over 400 phosphorylation sites (pTyr, pSer, and pThr) that were responsive to FLT3 inhibition in FLT3 driven human leukemia cell lines. We also extended this phosphoproteomic analysis on bone marrow from primary AML patient samples, and identify over 200 tyrosine and 800 serine/threonine phosphorylation sites in vivo. This study showed that oncogenic FLT3 regulates proteins involving diverse cellular processes and affects multiple signaling pathways in human leukemia that we previously appreciated, such as Fc epsilon RI-mediated signaling, BCR, and CD40 signaling pathways. It provides a valuable resource for investigation of oncogenic FLT3 signaling in human leukemia.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction , fms-Like Tyrosine Kinase 3/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Line, Tumor , Humans , Mutation , Phosphoamino Acids/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Quinazolines/pharmacology , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/genetics
4.
Cell Stem Cell ; 8(2): 200-13, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21295276

ABSTRACT

TET family enzymes convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in DNA. Here, we show that Tet1 and Tet2 are Oct4-regulated enzymes that together sustain 5hmC in mouse embryonic stem cells (ESCs) and are induced concomitantly with 5hmC during reprogramming of fibroblasts to induced pluripotent stem cells. ESCs depleted of Tet1 by RNAi show diminished expression of the Nodal antagonist Lefty1 and display hyperactive Nodal signaling and skewed differentiation into the endoderm-mesoderm lineage in embryoid bodies in vitro. In Fgf4- and heparin-supplemented culture conditions, Tet1-depleted ESCs activate the trophoblast stem cell lineage determinant Elf5 and can colonize the placenta in midgestation embryo chimeras. Consistent with these findings, Tet1-depleted ESCs form aggressive hemorrhagic teratomas with increased endoderm, reduced neuroectoderm, and ectopic appearance of trophoblastic giant cells. Thus, 5hmC is an epigenetic modification associated with the pluripotent state, and Tet1 functions to regulate the lineage differentiation potential of ESCs.


Subject(s)
Cytosine/analogs & derivatives , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Binding Sites/genetics , Binding Sites/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage , Chromatin Immunoprecipitation , Computational Biology , Cytosine/metabolism , DNA-Binding Proteins/genetics , Dioxygenases , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins/genetics , Teratoma/genetics , Teratoma/metabolism
5.
PLoS One ; 6(1): e15640, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21253578

ABSTRACT

Cholangiocarcinoma, also known as bile duct cancer, is the second most common primary hepatic carcinoma with a median survival of less than 2 years. The molecular mechanisms underlying the development of this disease are not clear. To survey activated tyrosine kinases signaling in cholangiocarcinoma, we employed immunoaffinity profiling coupled to mass spectrometry and identified DDR1, EPHA2, EGFR, and ROS tyrosine kinases, along with over 1,000 tyrosine phosphorylation sites from about 750 different proteins in primary cholangiocarcinoma patients. Furthermore, we confirmed the presence of ROS kinase fusions in 8.7% (2 out of 23) of cholangiocarcinoma patients. Expression of the ROS fusions in 3T3 cells confers transforming ability both in vitro and in vivo, and is responsive to its kinase inhibitor. Our data demonstrate that ROS kinase is a promising candidate for a therapeutic target and for a diagnostic molecular marker in cholangiocarcinoma. The identification of ROS tyrosine kinase fusions in cholangiocarcinoma, along with the presence of other ROS kinase fusions in lung cancer and glioblastoma, suggests that a more broadly based screen for activated ROS kinase in cancer is warranted.


Subject(s)
Bile Duct Neoplasms/enzymology , Bile Ducts, Intrahepatic , Cholangiocarcinoma/enzymology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Signal Transduction , Animals , Cell Line, Tumor , Humans , Immunoassay , Mice , Mice, Nude , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphorylation , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/metabolism
6.
Blood ; 113(12): 2776-90, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-18845790

ABSTRACT

Anaplastic large cell lymphoma represents a subset of neoplasms caused by translocations that juxtapose the anaplastic lymphoma kinase (ALK) to dimerization partners. The constitutive activation of ALK fusion proteins leads to cellular transformation through a complex signaling network. To elucidate the ALK pathways sustaining lymphomagenesis and tumor maintenance, we analyzed the tyrosine-kinase protein profiles of ALK-positive cell lines using 2 complementary proteomic-based approaches, taking advantage of a specific ALK RNA interference (RNAi) or cell-permeable inhibitors. A well-defined set of ALK-associated tyrosine phosphopeptides, including metabolic enzymes, kinases, ribosomal and cytoskeletal proteins, was identified. Validation studies confirmed that vasodilator-stimulated phosphoprotein and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase (ATIC) associated with nucleophosmin (NPM)-ALK, and their phosphorylation required ALK activity. ATIC phosphorylation was documented in cell lines and primary tumors carrying ALK proteins and other tyrosine kinases, including TPR-Met and wild type c-Met. Functional analyses revealed that ALK-mediated ATIC phosphorylation enhanced its enzymatic activity, dampening the methotrexate-mediated transformylase activity inhibition. These findings demonstrate that proteomic approaches in well-controlled experimental settings allow the definition of informative proteomic profiles and the discovery of novel ALK downstream players that contribute to the maintenance of the neoplastic phenotype. Prediction of tumor responses to methotrexate may justify specific molecular-based chemotherapy.


Subject(s)
Hydroxymethyl and Formyl Transferases/metabolism , Lymphoma, Large-Cell, Anaplastic/enzymology , Multienzyme Complexes/metabolism , Neoplasm Proteins/metabolism , Nucleotide Deaminases/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Antimetabolites, Antineoplastic/pharmacology , Carbazoles/pharmacology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Humans , Hydroxymethyl and Formyl Transferases/antagonists & inhibitors , Indazoles/pharmacology , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/pathology , Methotrexate/pharmacology , Microfilament Proteins/metabolism , Molecular Sequence Data , Multienzyme Complexes/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Nucleotide Deaminases/antagonists & inhibitors , Phenylurea Compounds/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/analysis , Protein Interaction Mapping , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/antagonists & inhibitors , Transcription, Genetic
7.
Proc Natl Acad Sci U S A ; 105(2): 692-7, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18180459

ABSTRACT

A major question regarding the sensitivity of solid tumors to targeted kinase inhibitors is why some tumors respond and others do not. The observation that many tumors express EGF receptor (EGFR), yet only a small subset with EGFR-activating mutations respond clinically to EGFR inhibitors (EGFRIs), suggests that responsive tumors uniquely depend on EGFR signaling for their survival. The nature of this dependence is not understood. Here, we investigate dependence on EGFR signaling by comparing non-small-cell lung cancer cell lines driven by EGFR-activating mutations and genomic amplifications using a global proteomic analysis of phospho-tyrosine signaling. We identify an extensive receptor tyrosine kinase signaling network established in cells expressing mutated and activated EGFR or expressing amplified c-Met. We show that in drug sensitive cells the targeted tyrosine kinase drives other RTKs and an extensive network of downstream signaling that collapse with drug treatment. Comparison of the signaling networks in EGFR and c-Met-dependent cells identify a "core network" of approximately 50 proteins that participate in pathways mediating drug response.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-met/metabolism , Cell Line, Tumor , Gefitinib , Humans , Models, Biological , Neoplasm Metastasis , Phosphotyrosine/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Quinazolines/pharmacology , Signal Transduction
8.
Cell ; 131(6): 1190-203, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-18083107

ABSTRACT

Despite the success of tyrosine kinase-based cancer therapeutics, for most solid tumors the tyrosine kinases that drive disease remain unknown, limiting our ability to identify drug targets and predict response. Here we present the first large-scale survey of tyrosine kinase activity in lung cancer. Using a phosphoproteomic approach, we characterize tyrosine kinase signaling across 41 non-small cell lung cancer (NSCLC) cell lines and over 150 NSCLC tumors. Profiles of phosphotyrosine signaling are generated and analyzed to identify known oncogenic kinases such as EGFR and c-Met as well as novel ALK and ROS fusion proteins. Other activated tyrosine kinases such as PDGFRalpha and DDR1 not previously implicated in the genesis of NSCLC are also identified. By focusing on activated cell circuitry, the approach outlined here provides insight into cancer biology not available at the chromosomal and transcriptional levels and can be applied broadly across all human cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/genetics , Anaplastic Lymphoma Kinase , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Enzyme Activation , Gene Fusion , Humans , Lung Neoplasms/genetics , Models, Biological , Molecular Sequence Data , Phosphorylation , Phosphotyrosine/genetics , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases , Receptor, Platelet-Derived Growth Factor alpha/metabolism
9.
Proc Natl Acad Sci U S A ; 104(50): 19855-60, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18077418

ABSTRACT

To ensure survival in the face of genomic insult, cells have evolved complex mechanisms to respond to DNA damage, termed the DNA damage checkpoint. The serine/threonine kinases ataxia telangiectasia-mutated (ATM) and ATM and Rad3-related (ATR) activate checkpoint signaling by phosphorylating substrate proteins at SQ/TQ motifs. Although some ATM/ATR substrates (Chk1, p53) have been identified, the lack of a more complete list of substrates limits current understanding of checkpoint pathways. Here, we use immunoaffinity phosphopeptide isolation coupled with mass spectrometry to identify 570 sites phosphorylated in UV-damaged cells, 498 of which are previously undescribed. Semiquantitative analysis yielded 24 known and 192 previously uncharacterized sites differentially phosphorylated upon UV damage, some of which were confirmed by SILAC, Western blotting, and immunoprecipitation/Western blotting. ATR-specific phosphorylation was investigated by using a Seckel syndrome (ATR mutant) cell line. Together, these results provide a rich resource for further deciphering ATM/ATR signaling and the pathways mediating the DNA damage response.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/radiation effects , Tumor Suppressor Proteins/physiology , Ultraviolet Rays , Amino Acid Motifs/genetics , Amino Acid Motifs/physiology , Amino Acid Motifs/radiation effects , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , DNA Damage/physiology , DNA Damage/radiation effects , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Humans , Immunoprecipitation , Phosphopeptides/immunology , Phosphopeptides/isolation & purification , Phosphopeptides/physiology , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Substrate Specificity/genetics , Substrate Specificity/radiation effects , Tumor Suppressor Proteins/immunology , Tumor Suppressor Proteins/metabolism
10.
Blood ; 108(13): 4202-4, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16946300

ABSTRACT

The 8p11 myeloproliferative syndrome (EMS) is associated with translocations that disrupt the FGFR1 gene. To date, 8 fusion partners of FGFR1 have been identified. However, no primary leukemia cell lines were identified that contain any of these fusions. Here, we screened more than 40 acute myeloid leukemia cell lines for constitutive phosphorylation of STAT5 and applied an immunoaffinity profiling strategy to identify tyrosine-phosphorylated proteins in the KG-1 cell line. Mass spectrometry analysis of KG-1 cells revealed aberrant tyrosine phosphorylation of FGFR1. Subsequent analysis led to the identification of a fusion of the FGFR1OP2 gene to the FGFR1 gene. Small interfering RNA (siRNA) against FGFR1 specifically inhibited the growth and induced apoptosis of KG-1 cells. Thus, the KG-1 cell line provides an in vitro model for the study of FGFR1 fusions associated with leukemia and for the analysis of small molecule inhibitors against FGFR1 fusions.


Subject(s)
Apoptosis/genetics , Leukemia, Myeloid, Acute/genetics , Models, Biological , Oncogene Proteins, Fusion/biosynthesis , Proto-Oncogene Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/biosynthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , STAT5 Transcription Factor/biosynthesis , STAT5 Transcription Factor/genetics
11.
Nat Immunol ; 7(8): 819-26, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16819554

ABSTRACT

Forkhead transcription factors are key participants in development and immune regulation. Here we demonstrate that absence of the gene encoding the forkhead transcription factor Foxp1 resulted in a profound defect in early B cell development. Foxp1 deficiency was associated with decreased expression of all B lineage genes in B220+ fetal liver cells as well as with a block in the transition from pro-B cell to pre-B cell involving diminished expression of recombination-activating genes 1 and 2. Foxp1 bound to the Erag enhancer and was involved in controlling variable-(diversity)-joining recombination of the gene encoding immunoglobulin heavy chain in a B cell lineage-specific way. Our results identify Foxp1 as an essential participant in the transcriptional regulatory network of B lymphopoiesis.


Subject(s)
B-Lymphocytes/cytology , Cell Lineage/immunology , Forkhead Transcription Factors/immunology , Lymphopoiesis/physiology , Repressor Proteins/immunology , Transcription, Genetic/immunology , Animals , B-Lymphocytes/immunology , Base Sequence , Blotting, Southern , Cell Differentiation/immunology , Chimera , Electrophoretic Mobility Shift Assay , Flow Cytometry , Gene Expression , Genes, RAG-1/immunology , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
12.
Cancer Cell ; 10(1): 65-75, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16843266

ABSTRACT

Tyrosine kinases are aberrantly activated in numerous malignancies, including acute myeloid leukemia (AML). To identify tyrosine kinases activated in AML, we developed a screening strategy that rapidly identifies tyrosine-phosphorylated proteins using mass spectrometry. This allowed the identification of an activating mutation (A572V) in the JAK3 pseudokinase domain in the acute megakaryoblastic leukemia (AMKL) cell line CMK. Subsequent analysis identified two additional JAK3 alleles, V722I and P132T, in AMKL patients. JAK3(A572V), JAK3(V722I), and JAK3(P132T) each transform Ba/F3 cells to factor-independent growth, and JAK3(A572V) confers features of megakaryoblastic leukemia in a murine model. These findings illustrate the biological importance of gain-of-function JAK3 mutations in leukemogenesis and demonstrate the utility of proteomic approaches to identifying clinically relevant mutations.


Subject(s)
Leukemia, Experimental/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Protein-Tyrosine Kinases/genetics , Alleles , Animals , Apoptosis/drug effects , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Imatinib Mesylate , Janus Kinase 2 , Janus Kinase 3 , K562 Cells , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Leukemia, Megakaryoblastic, Acute/metabolism , Leukemia, Megakaryoblastic, Acute/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , TYK2 Kinase
13.
Nature ; 441(7093): 646-50, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16511445

ABSTRACT

Precise regulation of the NFAT (nuclear factor of activated T cells) family of transcription factors (NFAT1-4) is essential for vertebrate development and function. In resting cells, NFAT proteins are heavily phosphorylated and reside in the cytoplasm; in cells exposed to stimuli that raise intracellular free Ca2+ levels, they are dephosphorylated by the calmodulin-dependent phosphatase calcineurin and translocate to the nucleus. NFAT dephosphorylation by calcineurin is countered by distinct NFAT kinases, among them casein kinase 1 (CK1) and glycogen synthase kinase 3 (GSK3). Here we have used a genome-wide RNA interference (RNAi) screen in Drosophila to identify additional regulators of the signalling pathway leading from Ca2+-calcineurin to NFAT. This screen was successful because the pathways regulating NFAT subcellular localization (Ca2+ influx, Ca2+-calmodulin-calcineurin signalling and NFAT kinases) are conserved across species, even though Ca2+-regulated NFAT proteins are not themselves represented in invertebrates. Using the screen, we have identified DYRKs (dual-specificity tyrosine-phosphorylation regulated kinases) as novel regulators of NFAT. DYRK1A and DYRK2 counter calcineurin-mediated dephosphorylation of NFAT1 by directly phosphorylating the conserved serine-proline repeat 3 (SP-3) motif of the NFAT regulatory domain, thus priming further phosphorylation of the SP-2 and serine-rich region 1 (SRR-1) motifs by GSK3 and CK1, respectively. Thus, genetic screening in Drosophila can be successfully applied to cross evolutionary boundaries and identify new regulators of a transcription factor that is expressed only in vertebrates.


Subject(s)
Drosophila/enzymology , Drosophila/genetics , Genome, Insect/genetics , NFATC Transcription Factors/metabolism , Protein Serine-Threonine Kinases/classification , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/classification , Protein-Tyrosine Kinases/metabolism , RNA Interference , Animals , Casein Kinase I/metabolism , Drosophila/metabolism , Genomics , Glycogen Synthase Kinase 3/metabolism , Interleukin-2/genetics , NFATC Transcription Factors/chemistry , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Transcription, Genetic/genetics , Dyrk Kinases
14.
Blood ; 107(12): 4888-97, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16497976

ABSTRACT

The Bcr-Abl fusion kinase drives oncogenesis in chronic myeloid leukemia (CML). CML patients are currently treated with the Abl tyrosine kinase inhibitor imatinib, which is effective in early stages of the disease. However, resistance to imatinib arises in later disease stages primarily because of a Bcr-Abl mutation. To gain deeper insight into Bcr-Abl signaling pathways, we generated phosphotyrosine profiles for 6 cell lines that represent 3 Bcr-Abl fusion types by using immunoaffinity purification of tyrosine phosphopeptides followed by tandem mass spectrometry. We identified 188 nonredundant tyrosine-phosphorylated sites, 77 of which are novel. By comparing the profiles, we found a number of phosphotyrosine sites common to the 6 cell lines regardless of cellular background and fusion type, several of which are decreased by imatinib treatment. Comparison of this Bcr-Abl signature with the profile of cells expressing an alternative imatinib-sensitive fusion kinase, FIP1L1-PDGFRalpha, revealed that these kinases signal through different pathways. This phosphoproteomic study of the Bcr-Abl fusion kinase highlights novel disease markers and potential drug-responsive biomarkers and adds novel insight into the oncogenic signals driven by the Bcr-Abl kinase.


Subject(s)
Biomarkers, Tumor/metabolism , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Phosphotyrosine/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Benzamides , Biomarkers, Tumor/analysis , Drug Resistance, Neoplasm/drug effects , Humans , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Oncogene Proteins, Fusion/metabolism , Phosphotyrosine/analysis , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proteomics , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , mRNA Cleavage and Polyadenylation Factors/metabolism
15.
Leuk Res ; 30(9): 1097-104, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16464493

ABSTRACT

STAT5 is constitutively phosphorylated in leukemic cells in approximately 70% of acute myeloid leukemia (AML) patients. To identify kinase candidates potentially responsible for STAT5 phosphorylation, we used liquid chromatography-tandem mass spectrometry (LC-MS/MS) mass spectrometry to detect phosphoproteins in AML cell lines. We established TEL-ARG and BCR-ABL fusion proteins as the mechanism underlying STAT5 phosphorylation in HT-93 and KBM-3 cells, respectively. In addition, we identified a JAK2 pseudokinase domain mutation in HEL cells and using siRNA downregulation, established JAK2 as the kinase responsible for phosphorylating STAT5. This study illustrates the benefit of LC-MS/MS mass spectrometry and siRNA for the identification of novel targets and mutations.


Subject(s)
Carcinogens , Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/genetics , Carcinogens/metabolism , Cell Line, Tumor , Chromatography, Liquid , Humans , Leukemia, Myeloid, Acute/metabolism , Mass Spectrometry , Mutation , Neoplasm Proteins/metabolism , Phosphorylation , Protein Processing, Post-Translational/genetics , Proteomics
16.
Int Immunol ; 17(11): 1513-24, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16199489

ABSTRACT

Th2 cells and mast cells are major sources of IL4, IL5 and IL13, cytokines that mediate immunity against parasites and are also central players in the pathophysiology of asthma, allergy and atopic disease. We asked whether Th2 cells and mast cells, which belong to the lymphoid and myeloid lineages, respectively, use different cis-acting regulatory regions to transcribe the cytokine genes. Comparison of DNase I hypersensitivity patterns at the RAD50/IL4/IL13 locus revealed that most hypersensitive sites (HSs) are common to Th2 and mast cells, but two regions [conserved non-coding sequence (CNS) 1 and mast cell HSs] show cell type-specific differences. CNS-1, one of the most highly conserved CNS regions in the RAD50/IL13/IL4 locus, displays two strong DNase I HSs in Th2 cells but is not DNase I hypersensitive in mast cells, explaining a previous finding that deletion of CNS-1 impairs cytokine expression in Th2 cells but not in mast cells. Conversely, two constitutive HSs (mast cell HSs) in the first intron of the IL13 gene are present in mast cells but not in Th2 cells; these sites develop early during mast cell differentiation and may have a role in maintaining accessibility of the IL13 locus to high-level transcription in stimulated cells.


Subject(s)
Cell Differentiation/immunology , Cytokines/biosynthesis , Gene Expression Regulation/immunology , Mast Cells/immunology , Th2 Cells/immunology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Acid Anhydride Hydrolases , Animals , Cell Differentiation/genetics , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , DNA-Binding Proteins , Gene Expression Regulation/genetics , Mice , Mice, Inbred BALB C , Quantitative Trait Loci/genetics , Quantitative Trait Loci/immunology , Regulatory Sequences, Nucleic Acid/genetics , Regulatory Sequences, Nucleic Acid/immunology , Transcription, Genetic/genetics , Transcription, Genetic/immunology
17.
Nat Immunol ; 5(8): 768-74, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15282556

ABSTRACT

The combination of bioinformatic and biological approaches constitutes a powerful method for identifying gene regulatory elements. High-quality genome sequences are available in public databases for several vertebrate species. Comparative cross-species sequence analysis of these genomes shows considerable conservation of noncoding sequences in DNA. Biological analyses show that an unexpectedly high number of the conserved sequences correspond to functional cis-regulatory regions that influence gene transcription. Because research biologists are often unfamiliar with the bioinformatic resources at their disposal, this commentary discusses how to integrate biological and bioinformatic methods in the discovery of gene regulatory regions and includes a tutorial on widely available comparative genomics programs.


Subject(s)
Computational Biology , DNA/genetics , Genomics , Regulatory Sequences, Nucleic Acid/genetics , Software , Animals , Conserved Sequence , Genome , Genomic Library , Humans , Sequence Homology, Nucleic Acid , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...