Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Front Mol Biosci ; 10: 1148933, 2023.
Article in English | MEDLINE | ID: mdl-37091863

ABSTRACT

14-3-3 proteins play critical roles in controlling multiple aspects of the cellular response to stress and DNA damage including regulation of metabolism, cell cycle progression, cell migration, and apoptotic cell death by binding to protein substrates of basophilic protein kinases following their phosphorylation on specific serine/threonine residues. Although over 200 mammalian proteins that bind to 14-3-3 have been identified, largely through proteomic studies, in many cases the relevant protein kinase responsible for conferring 14-3-3-binding to these proteins is not known. To facilitate the identification of kinase-specific 14-3-3 clients, we developed a biochemical approach using high-density protein filter arrays and identified the translational regulatory molecule PABPC1 as a substrate for Chk1 and MAPKAP Kinase-2 (MK2) in vitro, and for MK2 in vivo, whose phosphorylation results in 14-3-3-binding. We identify Ser-470 on PABPC1 within the linker region connecting the RRM domains to the PABC domain as the critical 14-3-3-binding site, and demonstrate that loss of PABPC1 binding to 14-3-3 results in increased cell proliferation and decreased cell death in response to UV-induced DNA damage.

2.
Virulence ; 13(1): 890-902, 2022 12.
Article in English | MEDLINE | ID: mdl-35587156

ABSTRACT

Antibodies to SARS-CoV-2 are central to recovery and immunity from COVID-19. However, the relationship between disease severity and the repertoire of antibodies against specific SARS-CoV-2 epitopes an individual develops following exposure remains incompletely understood. Here, we studied seroprevalence of antibodies to specific SARS-CoV-2 and other betacoronavirus antigens in a well-annotated, community sample of convalescent and never-infected individuals obtained in August 2020. One hundred and twenty-four participants were classified into five groups: previously exposed but without evidence of infection, having no known exposure or evidence of infection, seroconverted without symptoms, previously diagnosed with symptomatic COVID-19, and recovered after hospitalization with COVID-19. Prevalence of IgGs specific to the following antigens was compared between the five groups: recombinant SARS-CoV-2 and betacoronavirus spike and nucleocapsid protein domains, peptides from a tiled array of 22-mers corresponding to the entire spike and nucleocapsid proteins, and peptides corresponding to predicted immunogenic regions from other proteins of SARS-CoV-2. Antibody abundance generally correlated positively with severity of prior illness. A number of specific immunogenic peptides and some that may be associated with milder illness or protection from symptomatic infection were identified. No convincing association was observed between antibodies to Receptor Binding Domain(s) (RBDs) of less pathogenic betacoronaviruses HKU1 or OC43 and COVID-19 severity. However, apparent cross-reaction with SARS-CoV RBD was evident and some predominantly asymptomatic individuals had antibodies to both MERS-CoV and SARS-CoV RBDs. Findings from this pilot study may inform development of diagnostics, vaccines, and therapeutic antibodies, and provide insight into viral pathogenic mechanisms.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Humans , Pilot Projects , Seroepidemiologic Studies , Spike Glycoprotein, Coronavirus
3.
J Histotechnol ; 43(4): 182-195, 2020 12.
Article in English | MEDLINE | ID: mdl-33245266

ABSTRACT

The ability to reproduce scientific findings is foundational in research; yet, it is compromised in part by poorly characterized reagents, including antibodies. In this report, we describe the application of complementary validation strategies tailored for use in immunohistochemical assays in the characterization of rabbit monoclonal antibodies against YAP and TAZ, homologous and sequentially similar transcriptional effectors of the Hippo signaling pathway. A lack of antibody reagents rigorously validated for immunohistochemistry has limited the Hippo signaling research community's ability to interrogate YAP and TAZ independently in tissue. In a series of normal and diseased human tissues, we were able to demonstrate differential expression patterns of YAP and TAZ, suggesting the potential for functional differences of these proteins. These differences can now be studied in greater detail with these highly validated tools.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antibodies, Monoclonal/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Humans , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism
4.
Sci Rep ; 6: 31740, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27545199

ABSTRACT

MUC1 is a shared tumor antigen expressed on >80% of human cancers. We completed the first prophylactic cancer vaccine clinical trial based on a non-viral antigen, MUC1, in healthy individuals at-risk for colon cancer. This trial provided a unique source of potentially effective and safe immunotherapeutic drugs, fully-human antibodies affinity-matured in a healthy host to a tumor antigen. We purified, cloned, and characterized 13 IgGs specific for several tumor-associated MUC1 epitopes with a wide range of binding affinities. These antibodies bind hypoglycosylated MUC1 on human cancer cell lines and tumor tissues but show no reactivity against fully-glycosylated MUC1 on normal cells and tissues. We found that several antibodies activate complement-mediated cytotoxicity and that T cells carrying chimeric antigen receptors with the antibody variable regions kill MUC1(+) target cells, express activation markers, and produce interferon gamma. Fully-human and tumor-specific, these antibodies are candidates for further testing and development as immunotherapeutic drugs.


Subject(s)
Antibodies/immunology , Breast Neoplasms/immunology , Cancer Vaccines/immunology , Mucin-1/immunology , Adult , Amino Acid Sequence , Antibody Affinity/immunology , Base Sequence , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cancer Vaccines/therapeutic use , Cell Line , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Epitopes/immunology , Female , HEK293 Cells , Humans , Immunoglobulin G/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , MCF-7 Cells , Male , Middle Aged , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Young Adult
5.
eNeuro ; 2(3)2015.
Article in English | MEDLINE | ID: mdl-26464987

ABSTRACT

Injury to the CNS induces astrogliosis, an astrocyte-mediated response that has both beneficial and detrimental impacts on surrounding neural and non-neural cells. The precise signaling events underlying astrogliosis are not fully characterized. Here, we show that astrocyte activation was altered and proliferation was reduced in Semaphorin 4B (Sema4B)-deficient mice following injury. Proliferation of cultured Sema4B(-/-) astrocytes was also significantly reduced. In contrast to its expected role as a ligand, the Sema4B ectodomain was not able to rescue Sema4B(-/-) astrocyte proliferation but instead acted as an antagonist against Sema4B(+/-) astrocytes. Furthermore, the effects of Sema4B on astrocyte proliferation were dependent on phosphorylation of the intracellular domain at Ser825. Our results suggest that Sema4B functions as an astrocyte receptor, defining a novel signaling pathway that regulates astrogliosis after CNS injury.

8.
Nat Biotechnol ; 30(5): 447-52, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446692

ABSTRACT

We describe a proteomics approach that identifies antigen-specific antibody sequences directly from circulating polyclonal antibodies in the serum of an immunized animal. The approach involves affinity purification of antibodies with high specific activity and then analyzing digested antibody fractions by nano-flow liquid chromatography coupled to tandem mass spectrometry. High-confidence peptide spectral matches of antibody variable regions are obtained by searching a reference database created by next-generation DNA sequencing of the B-cell immunoglobulin repertoire of the immunized animal. Finally, heavy and light chain sequences are paired and expressed as recombinant monoclonal antibodies. Using this technology, we isolated monoclonal antibodies for five antigens from the sera of immunized rabbits and mice. The antigen-specific activities of the monoclonal antibodies recapitulate or surpass those of the original affinity-purified polyclonal antibodies. This technology may aid the discovery and development of vaccines and antibody therapeutics, and help us gain a deeper understanding of the humoral response.


Subject(s)
Antibodies, Monoclonal/chemistry , Blood Proteins/chemistry , Proteomics/methods , Animals , Antibodies/chemistry , Antibody Specificity/genetics , B-Lymphocytes/cytology , Chromatography, Liquid/methods , Immunoglobulin G/chemistry , Immunoglobulin Variable Region/chemistry , Mice , Rabbits , Receptors, Progesterone/chemistry , Serum/immunology , Tandem Mass Spectrometry/methods
9.
Mol Cell Proteomics ; 11(5): 187-201, 2012 May.
Article in English | MEDLINE | ID: mdl-22322096

ABSTRACT

Proteomic studies of post-translational modifications by metal affinity or antibody-based methods often employ data-dependent analysis, providing rich data sets that consist of randomly sampled identified peptides because of the dynamic response of the mass spectrometer. This can complicate the primary goal of programs for drug development, mutational analysis, and kinase profiling studies, which is to monitor how multiple nodes of known, critical signaling pathways are affected by a variety of treatment conditions. Cell Signaling Technology has developed an immunoaffinity-based LC-MS/MS method called PTMScan Direct for multiplexed analysis of these important signaling proteins. PTMScan Direct enables the identification and quantification of hundreds of peptides derived from specific proteins in signaling pathways or specific protein types. Cell lines, tissues, or xenografts can be used as starting material. PTMScan Direct is compatible with both SILAC and label-free quantification. Current PTMScan Direct reagents target key nodes of many signaling pathways (PTMScan Direct: Multipathway), serine/threonine kinases, tyrosine kinases, and the Akt/PI3K pathway. Validation of each reagent includes score filtering of MS/MS assignments, filtering by identification of peptides derived from expected targets, identification of peptides homologous to expected targets, minimum signal intensity of peptide ions, and dependence upon the presence of the reagent itself compared with a negative control. The Multipathway reagent was used to study sensitivity of human cancer cell lines to receptor tyrosine kinase inhibitors and showed consistent results with previously published studies. The Ser/Thr kinase reagent was used to compare relative levels of kinase-derived phosphopeptides in mouse liver, brain, and embryo, showing tissue-specific activity of many kinases including Akt and PKC family members. PTMScan Direct will be a powerful quantitative method for elucidation of changes in signaling in a wide array of experimental systems, combining the specificity of traditional biochemical methods with the high number of data points and dynamic range of proteomic methods.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Peptide Fragments/chemistry , Protein Processing, Post-Translational , Animals , Brain/metabolism , Cell Line , Chromatography, Affinity , Chromatography, Liquid , Embryo, Mammalian/metabolism , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/isolation & purification , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Mice , Mice, Inbred BALB C , Peptide Fragments/isolation & purification , Peptide Mapping/methods , Phosphoproteins/chemistry , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Phosphorylation , Protein Interaction Maps , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/isolation & purification , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tandem Mass Spectrometry
10.
Int J Mol Sci ; 14(1): 286-307, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23344034

ABSTRACT

Traditional methods for analysis of peptides using liquid chromatography and tandem mass spectrometry (LC-MS/MS) lack the specificity to comprehensively monitor specific biological processes due to the inherent duty cycle limitations of the MS instrument and the stochastic nature of the analytical platform. PTMScan Direct is a novel, antibody-based method that allows quantitative LC-MS/MS profiling of specific peptides from proteins that reside in the same signaling pathway. New PTMScan Direct reagents have been produced that target peptides from proteins involved in DNA Damage/Cell Cycle and Apoptosis/Autophagy pathways. Together, the reagents provide access to 438 sites on 237 proteins in these signaling cascades. These reagents have been used to profile the response to UV damage of DNA in human cell lines. UV damage was shown to activate canonical DNA damage response pathways through ATM/ATR-dependent signaling, stress response pathways and induce the initiation of apoptosis, as assessed by an increase in the abundance of peptides corresponding to cleaved, activated caspases. These data demonstrate the utility of PTMScan Direct as a multiplexed assay for profiling specific cellular responses to various stimuli, such as UV damage of DNA.


Subject(s)
Apoptosis/radiation effects , Chromatography, Liquid/methods , DNA Damage , Protein Processing, Post-Translational/radiation effects , Signal Transduction/radiation effects , Tandem Mass Spectrometry/methods , Ultraviolet Rays , Amino Acid Sequence , Autophagy/radiation effects , Cell Cycle/radiation effects , Cell Line, Tumor , Humans , Indicators and Reagents , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Protein Interaction Mapping
11.
Mol Cell ; 44(6): 864-77, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22195962

ABSTRACT

Many tumor cells rely on aerobic glycolysis instead of oxidative phosphorylation for their continued proliferation and survival. Myc and HIF-1 are believed to promote such a metabolic switch by, in part, upregulating gene expression of pyruvate dehydrogenase (PDH) kinase 1 (PDHK1), which phosphorylates and inactivates mitochondrial PDH and consequently pyruvate dehydrogenase complex (PDC). Here we report that tyrosine phosphorylation enhances PDHK1 kinase activity by promoting ATP and PDC binding. Functional PDC can form in mitochondria outside of the matrix in some cancer cells and PDHK1 is commonly tyrosine phosphorylated in human cancers by diverse oncogenic tyrosine kinases localized to different mitochondrial compartments. Expression of phosphorylation-deficient, catalytic hypomorph PDHK1 mutants in cancer cells leads to decreased cell proliferation under hypoxia and increased oxidative phosphorylation with enhanced mitochondrial utilization of pyruvate and reduced tumor growth in xenograft nude mice. Together, tyrosine phosphorylation activates PDHK1 to promote the Warburg effect and tumor growth.


Subject(s)
Mitochondria/enzymology , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Tyrosine/metabolism , Animals , Female , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Transplantation , Neoplasms/pathology , Phosphorylation , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Transplantation, Heterologous
12.
Mol Cell Biol ; 31(24): 4938-50, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21969607

ABSTRACT

The Warburg effect describes an increase in aerobic glycolysis and enhanced lactate production in cancer cells. Lactate dehydrogenase A (LDH-A) regulates the last step of glycolysis that generates lactate and permits the regeneration of NAD(+). LDH-A gene expression is believed to be upregulated by both HIF and Myc in cancer cells to achieve increased lactate production. However, how oncogenic signals activate LDH-A to regulate cancer cell metabolism remains unclear. We found that the oncogenic receptor tyrosine kinase FGFR1 directly phosphorylates LDH-A. Phosphorylation at Y10 and Y83 enhances LDH-A activity by enhancing the formation of active, tetrameric LDH-A and the binding of LDH-A substrate NADH, respectively. Moreover, Y10 phosphorylation of LDH-A is common in diverse human cancer cells, which correlates with activation of multiple oncogenic tyrosine kinases. Interestingly, cancer cells with stable knockdown of endogenous LDH-A and rescue expression of a catalytic hypomorph LDH-A mutant, Y10F, demonstrate increased respiration through mitochondrial complex I to sustain glycolysis by providing NAD(+). However, such a compensatory increase in mitochondrial respiration in Y10F cells is insufficient to fully sustain glycolysis. Y10 rescue cells show decreased cell proliferation and ATP levels under hypoxia and reduced tumor growth in xenograft nude mice. Our findings suggest that tyrosine phosphorylation enhances LDH-A enzyme activity to promote the Warburg effect and tumor growth by regulating the NADH/NAD(+) redox homeostasis, representing an acute molecular mechanism underlying the enhanced lactate production in cancer cells.


Subject(s)
Homeostasis , L-Lactate Dehydrogenase/metabolism , NAD/metabolism , Tyrosine/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Respiration , Glycolysis , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , L-Lactate Dehydrogenase/genetics , Lactate Dehydrogenase 5 , Lactic Acid/biosynthesis , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
13.
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
14.
J Clin Invest ; 120(4): 1165-77, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20234090

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is one of the most common types of human cancer and frequently metastasizes to LNs. Identifying metastasis-promoting factors is of immense clinical interest, as the prognosis for patients with even a single unilateral LN metastasis is extremely poor. Here, we report that p90 ribosomal S6 kinase 2 (RSK2) promotes human HNSCC cell invasion and metastasis. We determined that RSK2 was overexpressed and activated in highly invasive HNSCC cell lines compared with poorly invasive cell lines. Expression of RSK2 also correlated with metastatic progression in patients with HNSCC. Ectopic expression of RSK2 substantially enhanced the invasive capacity of HNSCC cells, while inhibition of RSK2 activity led to marked attenuation of invasion in vitro. Additionally, shRNA knockdown of RSK2 substantially reduced the invasive and metastatic potential of HNSCC cells in vitro and in vivo in a xenograft mouse model, respectively. Mechanistically, we determined that cAMP-responsive element-binding protein (CREB) and Hsp27 are phosphorylated and activated by RSK2 and are important for the RSK2-mediated invasive ability of HNSCC cells. Our findings suggest that RSK2 is involved in the prometastatic programming of HNSCC cells, through phosphorylation of proteins in a putative signaling network. Moreover, targeting RSK2 markedly attenuates in vitro invasion and in vivo metastasis of HNSCC cells, suggesting that RSK2 may represent a therapeutic target in the treatment of metastatic HNSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Actins/chemistry , Animals , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Progression , HSP27 Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Lymphatic Metastasis , Mice , Neoplasm Invasiveness , Phosphorylation , RNA, Small Interfering/genetics , Ribosomal Protein S6 Kinases, 90-kDa/analysis , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors
15.
Blood ; 115(5): 1037-48, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-19996410

ABSTRACT

Constitutively active JAK2V617F and thrombopoietin receptor (TpoR) W515L/K mutants are major determinants of human myeloproliferative neoplasms (MPNs). We show that a TpoRW515 mutation (W515A), which we detected in 2 myelofibrosis patients, and the Delta5TpoR active mutant, where the juxtamembrane R/KW(515)QFP motif is deleted, induce a myeloproliferative phenotype in mouse bone marrow reconstitution experiments. This phenotype required cytosolic Y112 of the TpoR. Phosphotyrosine immunoprofiling detected phosphorylated cytosolic TpoR Y78 and Y112 in cells expressing TpoRW515A. Mutation of cytosolic Y112 to phenylalanine prevented establishment of the in vivo phenotype and decreased constitutive active signaling by Delta5TpoR and TpoRW515A, especially via the mitogen-activated protein (MAP)-kinase pathway, without decreasing Janus kinase 2 (JAK2) activation. In contrast, mutation of cytosolic Y78 to phenylalanine enhanced the myeloproliferative syndrome induced by the TpoRW515 mutants, by enhancing receptor-induced JAK2 activation. We propose that TpoR cytosolic phosphorylated Y112 and flanking sequences could become targets for pharmacologic inhibition in MPNs.


Subject(s)
Mutation , Myeloproliferative Disorders/genetics , Primary Myelofibrosis/genetics , Receptors, Thrombopoietin/genetics , Tyrosine/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Line , Cell Proliferation , Humans , Immunoblotting , Janus Kinase 2/metabolism , Mice , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Phosphoproteins/metabolism , Phosphorylation , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Receptors, Thrombopoietin/metabolism , Transfection , Tyrosine/metabolism
16.
Sci Signal ; 2(97): ra73, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19920251

ABSTRACT

The Warburg effect describes a pro-oncogenic metabolism switch such that cancer cells take up more glucose than normal tissue and favor incomplete oxidation of glucose even in the presence of oxygen. To better understand how tyrosine kinase signaling, which is commonly increased in tumors, regulates the Warburg effect, we performed phosphoproteomic studies. We found that oncogenic forms of fibroblast growth factor receptor type 1 inhibit the pyruvate kinase M2 (PKM2) isoform by direct phosphorylation of PKM2 tyrosine residue 105 (Y(105)). This inhibits the formation of active, tetrameric PKM2 by disrupting binding of the PKM2 cofactor fructose-1,6-bisphosphate. Furthermore, we found that phosphorylation of PKM2 Y(105) is common in human cancers. The presence of a PKM2 mutant in which phenylalanine is substituted for Y(105) (Y105F) in cancer cells leads to decreased cell proliferation under hypoxic conditions, increased oxidative phosphorylation with reduced lactate production, and reduced tumor growth in xenografts in nude mice. Our findings suggest that tyrosine phosphorylation regulates PKM2 to provide a metabolic advantage to tumor cells, thereby promoting tumor growth.


Subject(s)
Glycolysis/physiology , Neoplasms/metabolism , Pyruvate Kinase/metabolism , Tyrosine/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation , Fructosediphosphates/metabolism , Glucose/metabolism , Humans , Immunoblotting , K562 Cells , Male , Mice , Mice, Nude , Mutation , Neoplasms/genetics , Neoplasms/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oxygen Consumption , Phosphorylation , Proteomics/methods , Pyruvate Kinase/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Transplantation, Heterologous , Tumor Burden
17.
Am J Surg Pathol ; 33(7): 984-91, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19363441

ABSTRACT

NUT midline carcinoma (NMC) is a uniformly lethal malignancy that is defined by rearrangement of the nuclear protein in testis (NUT) gene on chromosome 15q14. NMCs are morphologically indistinguishable from other poorly differentiated carcinomas, and the diagnosis is usually made currently by fluorescence in situ hybridization (FISH). As normal NUT expression is confined to testis and ovary, we reasoned that an immunohistochemical (IHC) stain for NUT would be useful in diagnosing NMC. To this end, we raised a highly specific rabbit monoclonal antibody, C52, against a recombinant NUT polypeptide, and developed an IHC staining protocol. The sensitivity and specificity of C52 staining was evaluated in a panel of 1068 tissues, predominantly diverse types of carcinomas (n=906), including 30 NMCs. Split-apart FISH for NUT rearrangement was used as a "gold standard" diagnostic test for NMC. C52 immunoreactivity among carcinomas was confined to NMCs. IHC staining had a sensitivity of 87%, a specificity of 100%, a negative predictive value of 99%, and a positive predictive value of 100%. Two new cases of NMC containing BRD4-NUT fusions were detected by C52 IHC, but missed by conventional FISH. In both instances, these tumors contained cryptic BRD4-NUT rearrangements, as confirmed by FISH using a refined set of probes. Some germ cell tumors, including 64% of dysgerminomas, showed weak NUT immunoreactivity, consistent with the expression of NUT in normal germ cells. We conclude that IHC staining with the C52 monoclonal antibody is a highly sensitive and specific test that reliably distinguishes NMC from other forms of carcinoma. The NUT antibody is being prepared for commercial release and will be available in the near future.


Subject(s)
Antibodies, Monoclonal , Carcinoma/diagnosis , Animals , Antibody Specificity , Carcinoma/genetics , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Neoplasm Proteins , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Oncogene Proteins/genetics , Oncogene Proteins/immunology , Oncogene Proteins, Fusion , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
18.
Clin Cancer Res ; 15(9): 3023-8, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19366827

ABSTRACT

PURPOSE: Activating mutations within the tyrosine kinase domain of epidermal growth factor receptor (EGFR) are found in approximately 10% to 20% of non-small-cell lung cancer (NSCLC) patients and are associated with response to EGFR inhibitors. The most common NSCLC-associated EGFR mutations are deletions in exon 19 and L858R mutation in exon 21, together accounting for 90% of EGFR mutations. To develop a simple, sensitive, and reliable clinical assay for the identification of EGFR mutations in NSCLC patients, we generated mutation-specific rabbit monoclonal antibodies against each of these two most common EGFR mutations and aimed to evaluate the detection of EGFR mutations in NSCLC patients by immunohistochemistry. EXPERIMENTAL DESIGN: We tested mutation-specific antibodies by Western blot, immunofluorescence, and immunohistochemistry. In addition, we stained 40 EGFR genotyped NSCLC tumor samples by immunohistochemistry with these antibodies. Finally, with a panel of four antibodies, we screened a large set of NSCLC patient samples with unknown genotype and confirmed the immunohistochemistry results by DNA sequencing. RESULTS: These two antibodies specifically detect the corresponding mutant form of EGFR by Western blotting, immunofluorescence, and immunohistochemistry. Screening a panel of 340 paraffin-embedded NSCLC tumor samples with these antibodies showed that the sensitivity of the immunohistochemistry assay is 92%, with a specificity of 99% as compared with direct and mass spectrometry-based DNA sequencing. CONCLUSIONS: This simple assay for detection of EGFR mutations in diagnostic human tissues provides a rapid, sensitive, specific, and cost-effective method to identify lung cancer patients responsive to EGFR-based therapies.


Subject(s)
Antibodies, Monoclonal , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation/immunology , Animals , Biological Assay , Blotting, Western , Carcinoma, Non-Small-Cell Lung/secondary , DNA Mutational Analysis , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoglobulin G/immunology , Lung Neoplasms/pathology , Mice , Mice, Nude , Rabbits , Sensitivity and Specificity , Sequence Deletion , Transplantation, Heterologous , Tumor Cells, Cultured
19.
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
20.
RNA ; 14(7): 1318-27, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18515545

ABSTRACT

Eukaryotic initiation factor (eIF) 4E, the mRNA 5'-cap-binding protein, mediates the association of eIF4F with the mRNA 5'-cap structure to stimulate cap-dependent translation initiation in the cytoplasm. The assembly of eIF4E into the eIF4F complex is negatively regulated through a family of repressor proteins, called the eIF4E-binding proteins (4E-BPs). eIF4E is also present in the nucleus, where it is thought to stimulate nuclear-cytoplasmic transport of certain mRNAs. eIF4E is transported to the nucleus via its interaction with 4E-T (4E-transporter), but it is unclear how it is retained in the nucleus. Here we show that a sizable fraction (approximately 30%) of 4E-BP1 is localized to the nucleus, where it binds eIF4E. In mouse embryo fibroblasts (MEFs) subjected to serum starvation and/or rapamycin treatment, nuclear 4E-BPs sequester eIF4E in the nucleus. A dramatic loss of nuclear 4E-BP1 occurs in c-Ha-Ras-expressing MEFs, which fail to show starvation-induced nuclear accumulation of eIF4E. Therefore, 4E-BP1 is a regulator of eIF4E cellular localization.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/analysis , Cell Cycle Proteins , Cell Line , Cell Nucleus/chemistry , Cells, Cultured , Embryo, Mammalian/cytology , Eukaryotic Initiation Factor-4E/analysis , Eukaryotic Initiation Factors , Fibroblasts/chemistry , Fibroblasts/cytology , Mice , Phosphoproteins/analysis , Phosphorylation , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...