Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Leukemia ; 31(9): 1872-1881, 2017 09.
Article in English | MEDLINE | ID: mdl-28017967

ABSTRACT

Duvelisib, an oral dual inhibitor of PI3K-δ and PI3K-γ, is in phase III trials for the treatment of chronic lymphocytic leukemia (CLL) and indolent non-Hodgkin's lymphoma. In CLL, duvelisib monotherapy is associated with high iwCLL (International Workshop on Chronic Lymphocytic Leukemia) and nodal response rates, but complete remissions are rare. To characterize the molecular effect of duvelisib, we obtained samples from CLL patients on the duvelisib phase I trial. Gene expression studies (RNAseq, Nanostring, Affymetrix array and real-time RT-PCR) demonstrated increased expression of BCL2 along with several BH3-only pro-apoptotic genes. In concert with induction of transcript levels, reverse phase protein arrays and immunoblots confirmed increase at the protein level. The BCL2 inhibitor venetoclax induced greater apoptosis in ex vivo-cultured CLL cells obtained from patients on duvelisib compared with pre-treatment CLL cells from the same patients. In vitro combination of duvelisib and venetoclax resulted in enhanced apoptosis even in CLL cells cultured under conditions that simulate the tumor microenvironment. These data provide a mechanistic rationale for testing the combination of duvelisib and venetoclax in the clinic. Such combination regimen (NCT02640833) is being evaluated for patients with B-cell malignancies including CLL.


Subject(s)
Apoptosis Regulatory Proteins/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Isoquinolines/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Purines/pharmacology , Sulfonamides/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Drug Synergism , Humans , Isoquinolines/therapeutic use , Purines/therapeutic use , Sulfonamides/therapeutic use , Tumor Cells, Cultured
3.
Pharmacogenomics J ; 16(6): 530-535, 2016 11.
Article in English | MEDLINE | ID: mdl-26345518

ABSTRACT

Anthracyclines are efficient chemotherapy agents. However, their use is limited by anthracycline-induced cardiotoxicity (CT). We investigated the influence of polymorphisms in doxorubicin metabolic and functional pathways on late-onset CT as estimated by echocardiography in 251 childhood acute lymphoblastic leukemia (cALL) patients. Association analyses revealed a modulating effect of two variants: A-1629 T in ABCC5, an ATP-binding cassette transporter, and G894T in the NOS3 endothelial nitric oxide synthase gene. Individuals with the ABCC5 TT-1629 genotype had an average of 8-12% reduction of ejection (EF) and shortening fractions (SF; EF: P<0.0001, and SF: P=0.001, respectively). A protective effect of the NOS3 TT894 genotype on EF was seen in high-risk patients (P=0.02), especially in those who did not receive dexrazoxane (P=0.002). Analysis of an additional cohort of 44 cALL patients replicated the ABCC5 association but was underpowered for NOS3. In summary, we identified two biomarkers that may contribute to cALL anthracycline CT risk stratification.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Doxorubicin/adverse effects , Heart Diseases/genetics , Multidrug Resistance-Associated Proteins/genetics , Nitric Oxide Synthase Type III/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Cardiotonic Agents/therapeutic use , Cardiotoxicity , Child , Child, Preschool , Dexrazoxane/therapeutic use , Female , Genetic Predisposition to Disease , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/prevention & control , Heterozygote , Homozygote , Humans , Infant , Male , Multidrug Resistance-Associated Proteins/metabolism , Myocardial Contraction , Nitric Oxide Synthase Type III/metabolism , Pharmacogenetics , Phenotype , Protective Factors , Risk Assessment , Risk Factors , Stroke Volume , Time Factors , Treatment Outcome , Ventricular Function, Left , Young Adult
4.
Leukemia ; 29(9): 1811-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25917267

ABSTRACT

The functional relevance of the B-cell receptor (BCR) and the evolution of protein kinases as therapeutic targets have recently shifted the paradigm for treatment of B-cell malignancies. Inhibition of p110δ with idelalisib has shown clinical activity in chronic lymphocytic leukemia (CLL). The dynamic interplay of isoforms p110δ and p110γ in leukocytes support the hypothesis that dual blockade may provide a therapeutic benefit. IPI-145, an oral inhibitor of p110δ and p110γ isoforms, sensitizes BCR-stimulated and/or stromal co-cultured primary CLL cells to apoptosis (median 20%, n=57; P<0.0001) including samples with poor prognostic markers, unmutated IgVH (n=28) and prior treatment (n=15; P<0.0001). IPI-145 potently inhibits the CD40L/IL-2/IL-10 induced proliferation of CLL cells with an IC50 in sub-nanomolar range. A corresponding dose-responsive inhibition of pAKT(Ser473) is observed with an IC50 of 0.36 nM. IPI-145 diminishes the BCR-induced chemokines CCL3 and CCL4 secretion to 17% and 37%, respectively. Pre-treatment with 1 µM IPI-145 inhibits the chemotaxis toward CXCL12; reduces pseudoemperipolesis to median 50%, inferring its ability to interfere with homing capabilities of CLL cells. BCR-activated signaling proteins AKT(Ser473), BAD(Ser112), ERK(Thr202/Tyr204) and S6(Ser235/236) are mitigated by IPI-145. Importantly, for clinical development in hematological malignancies, IPI-145 is selective to CLL B cells, sparing normal B- and T-lymphocytes.


Subject(s)
Apoptosis/drug effects , Isoquinolines/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Purines/pharmacology , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Adenine/analogs & derivatives , Aged , Aged, 80 and over , Bone Marrow Cells/metabolism , Case-Control Studies , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Chemotaxis/drug effects , Chemotaxis/immunology , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Phosphorylation , Piperidines , Prognosis , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinazolinones/pharmacology , Receptors, Antigen, B-Cell/metabolism , Stromal Cells/metabolism
5.
Ann Oncol ; 23(9): 2245-2252, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22269178

ABSTRACT

BACKGROUND: Most non-small-cell lung cancer (NSCLC) patients receive cisplatin-based chemotherapy though clinical response is restricted to a subset of patients. DNA repair protein levels are possible surrogates for cisplatin-induced DNA adduct (and subsequent cell death) repair efficiency and thus molecular determinants of therapeutic efficacy. The International Adjuvant Lung Trial (IALT)-Bio study previously suggested ERCC1 and MSH2 as predictive of cisplatin-based therapeutic benefit. PATIENTS AND METHODS: DNA repair protein expression (XPF, BRCA1, ERCC1, MSH2, p53, PARP1, and ATM) was assessed by immunohistochemistry on a large subset of patients (N = 769) from the IALT trial. Tissue Microarray slides were digitally scanned and signal quantified by user-defined macros. Statistical analyses (univariate and multivariate) of 5-year disease-free survival (DFS) and 5-year overall survival used binary cut-offs (H score low/high expression). RESULTS: In patients with squamous cell carcinoma (SCC), ATM, p53, PARP1, ERCC1, and MSH2 displayed significant (borderline) predictive values, mainly on DFS with chemotherapy efficacy limited to low marker levels. Adenocarcinoma (ADC) results were not significant. BRCA1 and XPF were not significant for predictive modeling in either SCC or ADCs. CONCLUSION: Here predictive utility of DNA repair enzymes co-segregates with SCC histology, focusing their predictive value to this histological subclass of NSCLC. Distinct mechanisms of chemotherapeutic response or resistance might exist among histological subclasses of solid tumors.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/metabolism , Cisplatin/pharmacology , DNA-Binding Proteins/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Adenocarcinoma of Lung , Aged , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Chemotherapy, Adjuvant , Cisplatin/therapeutic use , DNA Repair , DNA-Binding Proteins/genetics , Disease-Free Survival , Female , Gene Expression , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Multivariate Analysis , Prognosis , Randomized Controlled Trials as Topic , Tissue Array Analysis , Treatment Outcome
6.
Oncogene ; 27(15): 2237-42, 2008 Apr 03.
Article in English | MEDLINE | ID: mdl-17934517

ABSTRACT

An unusual group of carcinomas, here termed nuclear protein in testis (NUT) midline carcinomas (NMC), are characterized by translocations that involve NUT, a novel gene on chromosome 15. In about 2/3rds of cases, NUT is fused to BRD4 on chromosome 19. Using a candidate gene approach, we identified two NMCs harboring novel rearrangements that result in the fusion of NUT to BRD3 on chromosome 9. The BRD3-NUT fusion gene encodes a protein composed of two tandem chromatin-binding bromodomains, an extra-terminal domain, a bipartite nuclear localization sequence, and almost the entirety of NUT that is highly homologous to BRD4-NUT. The function of NUT is unknown, but here we show that NUT contains nuclear localization and export sequences that promote nuclear-cytoplasmic shuttling via a leptomycin-sensitive pathway. In contrast, BRD3-NUT and BRD4-NUT are strictly nuclear, implying that the BRD moiety retains NUT in the nucleus via interactions with chromatin. Consistent with this idea, FRAP studies show that BRD4, BRD4-NUT and BRD3-NUT have significantly slower rates of lateral nuclear diffusion than that of NUT. To investigate the functional role of BRD-NUT fusion proteins in NMCs, we investigated the effects of siRNA-induced BRD3-NUT and BRD4-NUT withdrawal. Silencing of these proteins in NMC cell lines resulted in squamous differentiation and cell cycle arrest. Together, these data suggest that BRD-NUT fusion proteins contribute to carcinogenesis by associating with chromatin and interfering with epithelial differentiation.


Subject(s)
Carcinoma/pathology , Cell Differentiation/genetics , Cell Proliferation , Epithelial Cells/physiology , Nuclear Proteins/physiology , Oncogene Proteins, Fusion/physiology , Oncogene Proteins/physiology , Testicular Neoplasms/pathology , Base Sequence , Carcinoma/genetics , Cell Cycle Proteins , Cell Nucleus/metabolism , Chromatin/metabolism , Epithelial Cells/metabolism , HeLa Cells , Humans , Male , Molecular Sequence Data , Multigene Family , Neoplasm Proteins , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Testicular Neoplasms/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Tumor Cells, Cultured
7.
Breast Cancer Res Treat ; 104(2): 129-44, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17393302

ABSTRACT

Breast tumors with prominent plasma cell (PC) infiltrates often have a more favorable natural course that may plausibly be mediated by anti-tumor activity of the elicited antibodies. These breast tumor-associated PCs are typically IgG dominant in contrast to normal breast PCs, which are mainly IgA. It is our hypothesis that this PC infiltration represents a host immune response that is driven by one or more tumor antigens. Previously, we and others showed that medullary carcinoma (MC) had a focused repertoire and features suggestive of a protein antigen driven response. Infrequently, non-MC, not otherwise specified (NOS) breast tumors may exhibit heavy PC infiltrations, also of IgG isotype. In this first characterization of this favorable prognosis NOS subgroup, IgG heavy chain (Hc) and light chain (Lc) variable (V) regions from three PC-infiltrated NOS tumors were randomly cloned and sequenced. We found biased (V) gene usage by the infiltrating PCs and somatic hypermutation in the rearranged Ig Hc and Lc V regions that were compatible with antigenic selection of the progenitor B cells. The antibody response of NOS infiltrated breast cancer is repertoire-focused, with 13-68% of isolates being clonally reiterated in the samples. Each NOS patient used distinct Hc V-D-J and Lc V-J rearrangements, with her own immune response "footprint," but the overall pattern of gene usage followed that typical of exogenous antigen-induced immune responses. The data are consistent with the hypothesis that PC infiltrates infrequently arising in NOS tumors, as previously inferred for MC, are in response to one or more breast cancer-associated protein tumor antigens.


Subject(s)
Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Immunoglobulin G/immunology , Plasma Cells/immunology , Base Sequence , Breast Neoplasms/metabolism , Female , Humans , Immunoglobulin G/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Isotypes/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunophenotyping , Lymphocytes/immunology , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
8.
Ann Oncol ; 17(8): 1275-82, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16788002

ABSTRACT

Bone marrow (BM) mast cells (MC) are commonly found in association with lymphoplasmacytic cells (LPC) in patients with Waldenström's macroglobulinemia (WM). We therefore sought to clarify the role of MC in WM. Co-culture of sublethally irradiated HMC-1 MC, KU812 basophilic cells, or autologous BM MC along with BM LPC from WM patients resulted in MC dose-dependent tumor colony formation and/or proliferation as assessed by 3H-thymidine uptake studies. Furthermore, by immunohistochemistry, multicolor flow cytometry and/or RT-PCR analysis, CD40 ligand (CD154), a potent inducer of B-cell expansion, was expressed on BM MC from 32 of 34 (94%), 11 of 13 (85%), and 7 of 9 (78%) patients, respectively. In contrast, MC from five healthy donors did not express CD154. By multicolor flow cytometry, CD154 was expressed on BM LPC from 35 of 38 (92%) patients and functionality was confirmed by CD154 and CD40 agonistic antibody stimulation, which induced proliferation, support survival and/or pERK phosphorylation of LPC. Moreover, MC induced expansion of LPC from 3 of 5 patients was blocked in a dose dependent manner by use of a CD154 blocking protein. These studies demonstrate that in WM, MC may support tumor cell expansion through constitutive CD154-CD40 signaling and therefore provide the framework for therapeutic targeting of MC and MC-WM cell interactions in WM.


Subject(s)
Bone Marrow Cells/pathology , CD40 Ligand/metabolism , Mast Cells/pathology , Waldenstrom Macroglobulinemia/pathology , B-Lymphocytes/chemistry , B-Lymphocytes/pathology , Bone Marrow Cells/chemistry , CD40 Ligand/analysis , CD40 Ligand/antagonists & inhibitors , Cell Proliferation/drug effects , Coculture Techniques , Humans , Mast Cells/chemistry , Mast Cells/drug effects , Recombinant Fusion Proteins/pharmacology , Signal Transduction , Tumor Cells, Cultured , Waldenstrom Macroglobulinemia/metabolism
9.
Annu Rev Pathol ; 1: 375-404, 2006.
Article in English | MEDLINE | ID: mdl-18039120

ABSTRACT

The association between Epstein-Barr virus (EBV) and a large number of benign and malignant diseases is unique among DNA viruses. Within infected tissues, proteins that are expressed during the normal lytic and latent viral life cycle lead to cellular alterations that contribute to these EBV-associated diseases. Although the early events of EBV infection are poorly understood, increasing knowledge of the viral processes that govern viral latency has shed light upon the potential mechanisms by which EBV infection can lead to cellular transformation. Our current understanding of the role of EBV in the development of Burkitt lymphoma, Hodgkin lymphoma, nasopharyngeal carcinoma, and other EBV-associated diseases is discussed.


Subject(s)
Burkitt Lymphoma/pathology , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/physiology , Hodgkin Disease/pathology , Infectious Mononucleosis/pathology , Animals , B-Lymphocytes/pathology , B-Lymphocytes/virology , Burkitt Lymphoma/virology , Cell Transformation, Viral , Epstein-Barr Virus Infections/complications , Hodgkin Disease/virology , Humans , Infectious Mononucleosis/virology , Virus Latency , Virus Replication
10.
Blood ; 97(12): 3910-8, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11389034

ABSTRACT

The molecular cloning of the t(5;10)(q33;q22) associated with atypical chronic myeloid leukemia (CML) is reported. Fluorescence in situ hybridization (FISH), Southern blot, and reverse transcriptase- polymerase chain reaction analysis demonstrated that the translocation resulted in an H4/platelet-derived growth factor receptor betaR (PDGFbetaR) fusion transcript that incorporated 5' sequences from H4 fused in frame to 3' PDGFbetaR sequences encoding the transmembrane, WW-like, and tyrosine kinase domains. FISH combined with immunophenotype analysis showed that t(5;10)(q33;q22) was present in CD13(+) and CD14(+) cells but was not observed in CD3(+) or CD19(+) cells. H4 has previously been implicated in pathogenesis of papillary thyroid carcinoma as a fusion partner of RET. The H4/RET fusion incorporates 101 amino acids of H4, predicted to encode a leucine zipper dimerization domain, whereas the H4/PDGFbetaR fusion incorporated an additional 267 amino acids of H4. Retroviral transduction of H4/PDGFbetaR, but not a kinase-inactive mutant, conferred factor-independent growth to Ba/F3 cells and caused a T-cell lymphoblastic lymphoma in a murine bone marrow transplantation assay of transformation. Mutational analysis showed that the amino-terminal H4 leucine zipper domain (amino acids 55-93), as well as H4 amino acids 101 to 386, was required for efficient induction of factor-independent growth of Ba/F3 cells. Tryptophan-to-alanine substitutions in the PDGFbetaR WW-like domain at positions 566/593, or tyrosine-to-phenylalanine substitutions at PDGFbetaR positions 579/581 impaired factor-independent growth of Ba/F3 cells. H4/PDGFbetaR is an oncoprotein expressed in t(5;10)(q33;q22) atypical CML and requires dimerization motifs in the H4 moiety, as well as residues implicated in signal transduction by PDGFbetaR, for efficient induction of factor-independent growth of Ba/F3 cells. (Blood. 2001;97:3910-3918)


Subject(s)
Carcinoma, Papillary/genetics , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 5 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Proteins/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Thyroid Neoplasms/genetics , Translocation, Genetic , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 5/genetics , Cloning, Molecular , Cytogenetic Analysis , Cytoskeletal Proteins , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Gene Rearrangement , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mutagenesis , Myeloid Cells/metabolism , Myeloid Cells/pathology , Oncogene Proteins, Fusion , Protein Structure, Tertiary , Proteins/metabolism , Transfection
11.
Mol Cell Biol ; 21(10): 3547-57, 2001 May.
Article in English | MEDLINE | ID: mdl-11313480

ABSTRACT

TEL-JAK2 fusion proteins, which are a result of t(9;12)(p24;p13) translocations associated with human leukemia, activate Stat5 in vitro and in vivo and cause a myelo- and lymphoproliferative disease in a murine bone marrow transplant model. We report that Socs-1, a member of the SOCS family of endogenous inhibitors of JAKs and STATs, inhibits transformation of Ba/F3 cells by TEL-JAK2 but has no effect on Ba/F3 cells transformed by BCR-ABL, TEL-ABL, or TEL-platelet-derived growth factor receptor beta. TEL-JAK2, in addition to activating Stat5, associates with Shc and Grb2 and induces activation of Erk2, and expression of Socs-1 inhibits engagement of each of these signaling molecules. TEL-JAK2 kinase activity is inhibited by Socs-1, as assessed by in vitro kinase assays. In addition, Socs-1 induces proteasomal degradation of TEL-JAK2. Mutational analysis indicates that the SOCS box of Socs-1 is required for proteasomal degradation and for abrogation of growth of TEL-JAK2-transformed cells. Furthermore, murine bone marrow transplant assays demonstrate that expression of Socs-1 prolongs latency of TEL-JAK2-mediated disease in vivo. Collectively, these data indicate that Socs-1 inhibits TEL-JAK2 in vitro and in vivo through inhibition of kinase activity and induction of TEL-JAK2 protein degradation.


Subject(s)
Carrier Proteins/physiology , Cysteine Endopeptidases/physiology , Hematopoietic Stem Cells/physiology , Multienzyme Complexes/physiology , Oncogene Proteins, Fusion/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins , Repressor Proteins , Animals , Cell Line , Cell Transformation, Neoplastic , Enzyme Activation , Hematopoietic Stem Cells/pathology , Janus Kinase 2 , Mice , Proteasome Endopeptidase Complex , Signal Transduction/physiology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins
12.
Blood ; 97(5): 1435-41, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222391

ABSTRACT

Tyrosine kinase fusion oncogenes that occur as a result of chromosomal translocations have been shown to activate proliferative and antiapoptotic pathways in leukemic cells, but the importance of autocrine and paracrine expression of hematopoietic cytokines in leukemia pathogenesis is not understood. Evidence that leukemic transformation may be, at least in part, cytokine dependent includes data from primary human leukemia cells, cell culture experiments, and murine models of leukemia. This report demonstrates that interleukin (IL)-3 plasma levels are elevated in myeloproliferative disease (MPD) caused by the TEL/tyrosine kinase fusions TEL/platelet-derived growth factor beta receptor (PDGFbetaR), TEL/Janus kinase 2 (JAK2), and TEL/neurotrophin-3 receptor (TRKC). Plasma granulocyte-macrophage colony-stimulating factor (GM-CSF) levels were elevated by TEL/PDGFbetaR and TEL/JAK2. However, all of the fusions tested efficiently induced MPD in mice genetically deficient for both GM-CSF and IL-3, demonstrating that these cytokines are not necessary for the development of disease in this model system. Furthermore, in experiments using normal marrow transduced with TEL/PDGFbetaR retrovirus mixed with marrow transduced with an enhanced green fluorescent protein (EGFP) retrovirus, the MPD induced in these mice demonstrated minimal stimulation of normal myelopoiesis by the TEL/PDGFbetaR-expressing cells. In contrast, recipients of mixed GM-CSF-transduced and EGFP-transduced marrow exhibited significant paracrine expansion of EGFP-expressing cells. Collectively, these data demonstrate that, although cytokine levels are elevated in murine bone marrow transplant models of leukemia using tyrosine kinase fusion oncogenes, GM-CSF and IL-3 are not required for myeloproliferation by any of the oncogenes tested.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Myeloproliferative Disorders/etiology , Oncogene Proteins, Fusion/pharmacology , Animals , Bone Marrow Transplantation , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-3/genetics , Interleukin-3/pharmacology , Mice , Mice, Knockout , Myeloproliferative Disorders/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/pharmacology , Oncogene Proteins, Fusion/genetics , Paracrine Communication , Transduction, Genetic
13.
Hum Pathol ; 32(12): 1382-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11774173

ABSTRACT

Inflammatory pseudotumors (IPTs) of the lymph node and spleen are an uncommon, benign cause of lymphadenopathy and/or splenomegaly that often bear striking clinicopathologic similarities to the inflammatory myofibroblastic tumors (IMTs) found in soft tissues. These tumors have classically been grouped together under the umbrella category of "inflammatory pseudotumor." Recent evidence shows that IMTs are in fact neoplastic processes that often harbor balanced chromosomal translocations involving the ALK kinase gene. These translocations result in expression of ALK kinase in IMTs as assessed by immunohistochemical studies. However, the relationship between IMT and IPT of the lymph node and spleen is uncertain. To determine if ALK tyrosine kinase expression is also present in IPT, 13 cases of IPT (9 involving lymph nodes, 4 splenic lesions) were examined for the presence of ALK tyrosine kinase by immunohistochemical staining on paraffin-embedded tissue. In addition, in situ hybridization studies for Epstein-Barr virus--encoded RNAs (EBER) and immunoperoxidase studies for human herpesvirus-8 (HHV8)--specific proteins were performed. All cases had clinical, morphologic, and immunophenotypic findings typical of IPT and had varying proportions of fibroblastic and inflammatory components. Age ranged from 11 to 75 (median, 40) years; 8 subjects were male, and 5 were female. None of the cases (0 of 13) had positive staining for ALK kinase or HHV8, and in 1 a lymph node (1 of 13) was focally positive for EBV (EBER) by in situ hybridization. The absence of ALK kinase as detected by immunohistochemical studies in IPT of the lymph node and spleen suggests that this entity is biologically distinct from the histologically similar IMT.


Subject(s)
Fibromatosis, Abdominal/pathology , Granuloma, Plasma Cell/pathology , Lymph Nodes/pathology , Lymphatic Diseases/pathology , Ribosomal Proteins , Splenic Diseases/pathology , Adult , Aged , Anaplastic Lymphoma Kinase , Biomarkers, Tumor/analysis , Child , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/pathology , Female , Fibromatosis, Abdominal/enzymology , Granuloma, Plasma Cell/enzymology , Granuloma, Plasma Cell/virology , Herpesviridae Infections/complications , Herpesviridae Infections/pathology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/isolation & purification , Humans , Immunoenzyme Techniques , Immunophenotyping , In Situ Hybridization , Lymph Nodes/enzymology , Lymph Nodes/virology , Lymphatic Diseases/enzymology , Lymphatic Diseases/virology , Male , Middle Aged , Protein-Tyrosine Kinases/metabolism , RNA-Binding Proteins/analysis , Receptor Protein-Tyrosine Kinases , Splenic Diseases/enzymology , Splenic Diseases/virology
14.
EMBO J ; 19(8): 1827-38, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10775267

ABSTRACT

The TEL-TRKC fusion is expressed as a consequence of t(12;15)(p13;q25), and is associated with two human cancers: congenital fibrosarcoma and acute myelogenous leukemia (AML). We report that the T/T(F) and T/T(L) fusion variants associated with congenital fibrosarcoma and AML, respectively, are constitutively tyrosine phosphorylated, and confer factor-independent growth to the murine hematopoietic cell line Ba/F3. Retroviral transduction of T/T(L) causes a rapidly fatal myeloproliferative disease in a murine bone marrow transplant (BMT) model, whereas T/T(F) causes a long-latency, pre-B-cell lymphoblastic lymphoma. TEL-TRKC variants are potent activators of the MAP kinase pathway, but neither variant activates Stat5 or other Stat family members. T/T(L), but not T/T(F), induces tyrosine phosphorylation of phospholipase Cgamma (PLCgamma), phosphoinositol-3 kinase and SHC. However, mutation analysis demonstrates that PLCgamma tyrosine phos phorylation by T/T(L) is dispensable for induction of the myeloproliferative phenotype by T/T(L). Collectively, these data demonstrate that the TEL-TRKC fusion variants are oncoproteins that activate the MAP kinase pathway, and do not require activation of either PLCgamma or Stat5 for efficient induction of a myeloproliferative phenotype in the murine BMT model.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 15 , DNA-Binding Proteins/metabolism , Fibrosarcoma/genetics , Leukemia, Myeloid, Acute/genetics , Milk Proteins , Oncogene Proteins, Fusion/metabolism , Receptor, trkC/metabolism , Repressor Proteins , Transcription Factors/metabolism , Translocation, Genetic , 3T3 Cells , Animals , Bone Marrow/metabolism , Cell Transformation, Neoplastic , Fibrosarcoma/congenital , Hematopoietic Stem Cells/metabolism , Humans , Isoenzymes/metabolism , MAP Kinase Signaling System , Mice , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma , Phosphorylation , Proto-Oncogene Proteins c-ets , STAT5 Transcription Factor , Signal Transduction , Tissue Distribution , Trans-Activators/metabolism , Tumor Cells, Cultured , Type C Phospholipases/metabolism , Tyrosine/metabolism , ETS Translocation Variant 6 Protein
15.
Am J Clin Pathol ; 112(3): 343-8, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10478139

ABSTRACT

This report describes a novel, reliable, and simplified approach for determination of intranuclear terminal deoxynucleotidyl transferase (TdT) expression. This approach utilizes standard permeabilization/fixation solutions to reliably detect intracellular antigens with minimal alterations in the light scatter properties of the analyzed cells. In contrast to other described methods, fluorescein isothiocyanate-conjugated anti-TdT antibody is added to previously analyzed and permeabilized cells after a leukemic cell population has been identified using characteristic surface staining patterns. The method eliminates the need for additional sample preparation or cumbersome permeablization steps and can easily be incorporated into any clinical laboratory's existing flow cytometry panels. Sixty-eight cases were analyzed with this method, including 31 acute myelogenous leukemias, 30 acute lymphoblastic leukemias, and 7 chronic lymphoproliferative disorders. To confirm the validity of the method, parallel immunoperoxidase staining and microscopic evaluation of cytocentrifuge test sample preparations were performed. Statistical analysis of the results reveals the method to be highly sensitive and specific, demonstrating exact correlation to the microscopic method. The ease and expeditiousness of this new procedure allows TdT testing to be routinely incorporated into the immunophenotyping repertoire of a busy clinical laboratory. In addition, the method should be readily adaptable to analyze a variety of other clinically relevant intranuclear and intracytoplasmic antigens.


Subject(s)
Cell Nucleus/enzymology , DNA Nucleotidylexotransferase/analysis , Flow Cytometry , Leukemia, Myeloid/enzymology , Lymphoproliferative Disorders/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Acute Disease , Chronic Disease , Fluorescein-5-isothiocyanate , Humans , Immunoenzyme Techniques , Predictive Value of Tests , Reproducibility of Results , Sensitivity and Specificity , Statistics as Topic/methods , Time Factors
16.
Am J Clin Pathol ; 110(4): 465-70, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9763032

ABSTRACT

The clinical usefulness of monitoring CD4+ T-lymphocyte counts in patients infected with HIV is now well established. The need for accurate, rapid, and cost-effective methods for making these determinations is evident. Recent technologic advances have allowed for the development of a system for the determination of CD4+ T-lymphocyte counts by simultaneous 4-color flow cytometry. A new 4-color 2-tube flow cytometric method for analyzing CD4+ T-lymphocyte subsets in whole blood was compared with a standard 2-color 5-tube method. The new method provides results almost identical to those of the well-established 2-color method used in our clinical laboratory. Statistical analyses indicate very low variability in CD4+ counts between the 2 methods, strongly supporting the usefulness of this new procedure. In addition, the 4-color procedure provides a 15% reduction in the materials cost per test compared with the 2-color method, as well as a marked reduction in the time expenditure of flow cytometry technologists.


Subject(s)
CD4 Lymphocyte Count/methods , Flow Cytometry/economics , Flow Cytometry/methods , Immunophenotyping/methods , Adult , Aged , Cell Separation/methods , Color , Cost-Benefit Analysis , Female , Humans , Male , Middle Aged , Regression Analysis
17.
Blood ; 84(12): 4195-202, 1994 Dec 15.
Article in English | MEDLINE | ID: mdl-7994033

ABSTRACT

Thrombin receptor (TR) activation by alpha-thrombin requires proteolytic cleavage, although synthetic peptides modeled after the new N-terminus directly effect receptor activation without cleavage, presumably by interacting with an unidentified region of the receptor. To further define critical residues responsible for receptor activation, we performed epitope mapping of anti-TR1-160, a previously described polyclonal antibody that inhibits peptide ligand-induced receptor activation in various cell types expressing a functional TR. An enzyme-linked immunosorbent assay (ELISA) using overlapping decapeptides derived from the TR extracellular domains identified four immunodominant peaks within the long N-terminal extension centered between amino acids 34-44, 48-67, 65-79, and 87-94. Soluble peptides derived from regions 83-94, but not those from other regions of the receptor, neutralized the ability of anti-TR1-160 to inhibit peptide ligand-induced platelet aggregation, suggesting that antibodies directed against this region of the TR are important in ligand-mediated activation. Thrombin receptor mutants lacking discrete regions of the TR were subsequently evaluated using microinjected Xenopus oocytes. Whereas a TR mutant lacking amino acid residues Thr67-Lys82 (TR delta 67-82) showed normal to exaggerated responses to either alpha-thrombin or synthetic peptide ligands, only TR mutants with limited deletions spanning the residues Gln83-Ser93 exhibited dysfunctional responses to either agonist (200 nmol/L alpha-thrombin or 200 mumol/L TR42-47). These data provide a model for receptor activation that implicates a discrete and previously uncharacterized sequence within the TR N-terminal extension that is necessary for initiation of signal transduction events independent of the initiating agonist.


Subject(s)
Receptors, Thrombin/genetics , Signal Transduction , Thrombin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Humans , Immunodominant Epitopes/immunology , Microinjections , Molecular Sequence Data , Oocytes , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Peptide Fragments/metabolism , Receptors, Thrombin/immunology , Sequence Alignment , Xenopus
18.
Thromb Haemost ; 72(6): 964-72, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7537915

ABSTRACT

We produced a murine monoclonal antibody, 7H2, and localized its epitope to one or more small regions on platelet glycoprotein (GP) IIIa. 7H2-IgG and 7H2-F(ab')2 completely inhibit platelet aggregation and fibrinogen binding at low agonist concentrations, but only partially inhibit aggregation and fibrinogen binding at high agonist concentrations; 7H2-Fab has no effect on aggregation or fibrinogen binding at any agonist concentration. 7H2-IgG binds to the entire platelet population as judged by flow cytometry. At near saturating concentrations, approximately 40,000 7H2-IgG antibody molecules bind per platelet. In contrast, approximately 80,000 7H2 Fab molecules bind per platelet, suggesting that 7H2-IgG binding is bivalent. 7H2 was unable to inhibit fibrinogen binding to purified, immobilized GPIIb/IIIa. These data indicate that the bivalent binding of 7H2 to GPIIIa is required for its partial inhibition of fibrinogen binding to platelets, perhaps through dimerization of GPIIb/IIIa surface receptors (or more complex GPIIb/IIIa redistribution triggered by 7H2 binding) resulting in limited accessibility of fibrinogen to its binding site(s).


Subject(s)
Binding Sites, Antibody , Fibrinogen/immunology , Immunoglobulin G/immunology , Platelet Aggregation Inhibitors/immunology , Platelet Membrane Glycoproteins/immunology , Animals , Antibodies, Monoclonal , Antigen-Antibody Reactions , Epitopes/blood , Humans , Immunoglobulin Fab Fragments/immunology , Iodine Radioisotopes , Mice , Protein Binding
19.
J Clin Invest ; 92(6): 2796-806, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8254034

ABSTRACT

The accessibility of activated GPIIb/IIIa receptors on the luminal surface of platelets adherent to damaged blood vessels or atherosclerotic plaques is likely to play a crucial role in subsequent platelet recruitment. To define better the factors involved in this process, we developed a functional assay to assess the presence of activated, luminal GPIIb/IIIa receptors, based on their ability to bind erythrocytes containing a high density of covalently coupled RGD-containing peptides (thromboerythrocytes). Platelets readily adhered to wells coated with purified type I rat skin collagen and the adherent platelets bound a dense lawn of thromboerythrocytes. With fibrinogen-coated wells, platelet adhesion increased as the fibrinogen-coating concentration increased, reaching a plateau at about 11 micrograms/ml. Thromboerythrocyte binding to the platelets adherent to fibrinogen showed a paradoxical response, increasing at fibrinogen coating concentrations up to approximately 4-6 micrograms/ml and then dramatically decreasing at higher fibrinogen-coating concentrations. Scanning electron microscopy demonstrated that the morphology of platelets adherent to collagen was similar to that of platelets adherent to low density fibrinogen, with extensive filopodia formation and ruffling. In contrast, platelets adherent to high density fibrinogen showed a bland, flattened appearance. Immunogold staining of GPIIb/IIIa receptors demonstrated concentration of the receptors on the filopodia, and depletion of receptors on the flattened portion of the platelets. Thus, there is a paradoxical loss of accessible, activated GPIIb/IIIa receptors on the luminal surface of platelets adherent to high density fibrinogen. Two factors may contribute to this result: engagement of GPIIb/IIIa receptors with fibrinogen on the abluminal surface leading to the loss of luminal receptors, and loss of luminal filopodia that interact with thromboerythrocytes. These data provide insight into the differences in thrombogenicity between surfaces, and may provide a mechanism for purposefully passivating platelet-reactive artificial surfaces.


Subject(s)
Blood Platelets/physiology , Platelet Adhesiveness , Platelet Membrane Glycoproteins/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/pharmacology , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cross-Linking Reagents , Erythrocytes/metabolism , Fibrinogen/metabolism , Humans , In Vitro Techniques , Kinetics , Microscopy, Electron, Scanning , Molecular Sequence Data , Oligopeptides , Platelet Adhesiveness/drug effects
20.
Blood ; 82(7): 2125-36, 1993 Oct 01.
Article in English | MEDLINE | ID: mdl-8400262

ABSTRACT

The recently cloned functional thrombin receptor is thought to be activated by thrombin cleavage of the bond between R41 and S42, followed by the insertion of the new N-terminal region ("tethered ligand") into an unknown site in the receptor. Antibodies to peptides at or near the cleavage site have been reported to inhibit thrombin-induced platelet activation to varying extents, but the precise mechanism(s) of their inhibition is unknown. We have produced: (1) a polyclonal antibody in rabbits to a peptide containing amino acids 34 to 52 (anti-TR34-52); enzyme-linked immunosorbent assays (ELISA) indicate that anti-TR34-52 contains antibodies to regions on both sides of the thrombin cleavage site; (2) two murine monoclonal antibodies (MoAbs) to a peptide containing amino acids 29 to 68; one antibody reacts primarily with residues N-terminal to the thrombin cleavage site, and the other reacts primarily with residues C-terminal to the cleavage site; and (3) a polyclonal rabbit antibody to a peptide containing amino acids 83 to 94 (anti-TR83-94). Anti-TR34-52 binds to platelets as judged by flow cytometry, and pretreating platelets with a thrombin receptor peptide ligand does not lead to loss of antibody reactivity, suggesting that platelet activation does not initiate redistribution or internalization of surface thrombin receptors. In contrast, pretreating platelets with thrombin leads to complete loss of anti-TR34-52 binding. Similarly, the binding of both MoAbs to platelets is dramatically reduced by pretreatment with thrombin. However, the binding of anti-TR83-94 is not decreased by thrombin activation, confirming that the receptor is not internalized. Anti-TR34-52 profoundly inhibits low dose thrombin-induced platelet shape change and aggregation, but the inhibition can be overcome with higher thrombin doses. However, anti-TR34-52 does not inhibit platelet aggregation induced by tethered ligand peptides. The TR34-52 peptide is a thrombin substrate, with cleavage occurring at the R41-S42 bond as judged by high performance liquid chromatography (HPLC) and platelet aggregation analysis. Anti-TR34-52 prevented cleavage of the TR34-52 peptide, suggesting that the antibody prevents platelet activation, at least in part, by preventing cleavage of the thrombin receptor. These data, although indirect, provide additional support for a thrombin activation mechanism involving thrombin cleavage of the receptor; in addition, they provide new evidence indicating that receptor cleavage is followed by loss of the N-terminal peptide, and insertion of the tethered ligand into a protected domain.


Subject(s)
Blood Platelets/metabolism , Receptors, Thrombin/metabolism , Amino Acid Sequence , Animals , Antibodies , Antibodies, Monoclonal , Blood Platelets/drug effects , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Humans , Mice/immunology , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/immunology , Rabbits/immunology , Receptors, Thrombin/analysis , Receptors, Thrombin/immunology , Recombinant Proteins/analysis , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL