Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
Cancer Res ; 60(14): 3909-15, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919668

ABSTRACT

Plasma cell neoplasia in humans generally occurs as multiple myeloma, an incurable form of cancer. Tumors with marked similarity can be induced in mice by a variety of agents, including chemicals, silicone, and oncogene-containing retroviruses, suggesting the use of murine tumors as an informative model to study plasma cell disease. Herein, we have focused on the role of insulin-like growth factor I receptor (IGF-IR) signaling in the development of plasma cell disease. The insulin receptor substrate 2/phosphatidylinositol 3'-kinase/p70S6K pathway was found to be either constitutively or IGF-I-dependently activated in all plasma cell tumors. Biological relevance was demonstrated in that plasma cell lines with up-regulated IGF-IR expression levels exhibited mitogenic responses to IGF-I. More importantly, expression of a dominant-negative mutant of IGF-IR in these lines strongly suppressed tumorigenesis in vivo. Taken together, these results demonstrate that up-regulation and activation of IGF-IR and the downstream signaling pathway involving insulin receptor substrate 2, phosphatidylinositol 3'-kinase, and p70S6K may play an important role in the development of a broad spectrum of plasma cell tumors.


Subject(s)
Plasmacytoma/metabolism , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/physiology , Animals , Culture Media, Serum-Free , Enzyme Activation , Female , Immunoblotting , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Plasmacytoma/chemically induced , Precipitin Tests , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Cells, Cultured , Up-Regulation
2.
J Immunol ; 164(9): 4678-88, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10779773

ABSTRACT

Wild-type (F/B), constitutively active (F/B*), and three kinase-inactive (F/Ba-, F/Bb-, F/Bc-) forms of Akt/protein kinase B (PKB) were permanently overexpressed in FDCP2 cells. In the absence of insulin-like growth factor-1 (IGF-1), activities of PKB, cyclic nucleotide phosphodiesterase 3B (PDE3B), and PDE4 were similar in nontransfected FDCP2 cells, mock-transfected (F/V) cells, and F/B and F/B- cells. In F/V cells, IGF-1 increased PKB, PDE3B, and PDE4 activities approximately 2-fold. In F/B cells, IGF-1, in a wortmannin-sensitive manner, increased PKB activity approximately 10-fold and PDE3B phosphorylation and activity ( approximately 4-fold), but increased PDE4 to the same extent as in F/V cells. In F/B* cells, in the absence of IGF-1, PKB activity was markedly increased ( approximately 10-fold) and PDE3B was phosphorylated and activated (3- to 4-fold); wortmannin inhibited these effects. In F/B* cells, IGF-1 had little further effect on PKB and activation/phosphorylation of PDE3B. In F/B- cells, IGF-1 activated PDE4, not PDE3B, suggesting that kinase-inactive PKB behaved as a dominant negative with respect to PDE3B activation. Thymidine incorporation was greater in F/B* cells than in F/V cells and was inhibited to a greater extent by PDE3 inhibitors than by rolipram, a PDE4 inhibitor. In F/B cells, IGF-1-induced phosphorylation of the apoptotic protein BAD was inhibited by the PDE3 inhibitor cilostamide. Activated PKB phosphorylated and activated rPDE3B in vitro. These results suggest that PDE3B, not PDE4, is a target of PKB and that activated PDE3B may regulate cAMP pools that modulate effects of PKB on thymidine incorporation and BAD phosphorylation in FDCP2 cells.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Hematopoietic Stem Cells/enzymology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Thymidine/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/physiology , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cell Line , Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3 , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Insulin-Like Growth Factor I/physiology , Mice , Molecular Sequence Data , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection , bcl-Associated Death Protein
3.
Oncogene ; 18(47): 6564-72, 1999 Nov 11.
Article in English | MEDLINE | ID: mdl-10597260

ABSTRACT

To investigate the role of protein kinase C (PKC) in apoptotic signaling induced by cytokine withdrawal, we expressed PKC-alpha, -delta and -epsilon individually in the 32D myeloid progenitor cells. The parental and PKC-delta- and PKC-epsilon-transfected 32D cells underwent apoptosis within 24 h in the absence of interleukin 3. In contrast, expression of PKC-alpha inhibited the onset of apoptosis as determined by genomic DNA fragmentation and flow cytometric analysis. Correlating with the inhibition of apoptosis, PKC-alpha transfectants exhibited increased activity of the endogenous Akt serine/threonine kinase. Furthermore, PKC-alpha, but not PKC-delta or -epsilon, specifically activated overexpressed Akt. PKC-alpha-induced Akt activity was partially dependent on phosphoinositol 3' kinase (PI 3'K) since a PI 3'K inhibitor was able to suppress PKC-alpha-induced Akt activation. Both basal and interleukin 3-stimulated phosphorylation of Akt on serine 473 was enhanced in the PKC-alpha and Akt contransfectants. Coexpression of wild type Akt and PKC-alpha resulted in greater suppression of apoptosis than PKC-alpha expression alone. Together, our results demonstrate that suppression of apoptosis by PKC-alpha correlates with its ability of activating endogenous Akt. Furthermore, activation of overexpressed Akt by PKC-alpha is consistent with their synergistic effect on suppressing apoptosis, providing the strong evidence of cross talk between Akt and PKC-alpha.


Subject(s)
Apoptosis , Interleukin-3/pharmacology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Androstadienes/pharmacology , Apoptosis/drug effects , Cell Line , DNA Replication , Enzyme Inhibitors/pharmacology , Interleukin-3/administration & dosage , Isoenzymes/genetics , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C-alpha , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Serine/metabolism , Transfection , Wortmannin
4.
Oncogene ; 18(17): 2681-9, 1999 Apr 29.
Article in English | MEDLINE | ID: mdl-10348342

ABSTRACT

The ErbB/HER family of receptor tyrosine kinases consists of four receptors that bind a large number of growth factor ligands sharing an epidermal growth factor- (EGF)-like motif. Whereas ErbB-1 binds seven different ligands whose prototype is EGF, the three families of neuregulins (NRGs) activate ErbB-3 and/or ErbB-4. Here we characterize a fourth neuregulin, NRG-4, that acts through ErbB-4. The predicted pro-NRG-4 is a transmembrane protein carrying a unique EGF-like motif and a short cytoplasmic domain. A synthetic peptide encompassing the full-length EGF-like domain can induce growth of interleukin-dependent cells ectopically expressing ErbB-4, but not cells expressing the other three ErbB proteins or their combinations. Consistent with specificity to ErbB-4, NRG-4 can displace an ErbB-4-bound NRG-1 and can activate signaling downstream of this receptor. Expression of NRG-4 mRNA was detected in the adult pancreas and weakly in muscle; other tissues displayed no detectable NRG-4 mRNA. The primary structure and the pattern of expression of NRG-4, together with the strict specificity of this growth factor to ErbB-4, suggest a physiological role distinct from that of the known ErbB ligands.


Subject(s)
ErbB Receptors/physiology , Glycoproteins/physiology , Nerve Growth Factors/physiology , Amino Acid Sequence , Base Sequence , Cell Division/physiology , Humans , Ligands , Molecular Sequence Data , Neuregulins , Organ Specificity , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , Receptor, ErbB-4
5.
Endocrinology ; 140(5): 1972-83, 1999 May.
Article in English | MEDLINE | ID: mdl-10218944

ABSTRACT

GH exerts a variety of metabolic and growth-promoting effects. GH induces activation of the GH receptor (GHR)-associated cytoplasmic tyrosine kinase, JAK2, resulting in tyrosine phosphorylation of the GHR and activation of STAT (signal transducer and activator of transcription), Ras-mitogen-activated protein kinase, and phosphoinositol 3-kinase signaling pathways, among others. GH-stimulated tyrosine phosphorylation of insulin receptor substrate (IRS) proteins has been demonstrated in vitro and in vivo. IRS-1 is a multiply phosphorylated cytoplasmic docking protein involved in metabolic and proliferative signaling by insulin, IL-4, and other cytokines, but the physiological role of IRS-1 in GH signaling is unknown. In this study, as noted by others, we detected in murine 3T3-F442A pre-adipocytes GH-dependent tyrosine phosphorylation of IRS-1 and specific GH-induced coimmunoprecipitation with JAK2 of a tyrosine phosphoprotein consistent with IRS-1. We further examined this interaction by in vitro affinity precipitation experiments with glutathione-S-transferase fusion proteins incorporating regions of rat IRS-1 and, as a source of JAK2, extracts of 3T3-F442A cells. Fusion proteins containing amino-terminal regions of IRS-1 that include the pleckstrin homology, phosphotyrosine-binding, and Shc and IRS-1 NPXY-binding domains, but not those containing other IRS-1 regions or glutathione-S-transferase alone, bound JAK2 from cell extracts. Tyrosine-phosphorylated JAK2 resulting from GH stimulation was included in the amino-terminal IRS-1 fusion precipitates; however, neither tyrosine phosphorylation of JAK2 nor treatment of cells with GH before extraction was necessary for the specific JAK2-IRS-1 interaction to be detected. In contrast, in this assay, specific insulin receptor association with the IRS-1 phosphotyrosine-binding, and Shc and IRS-1 NPXY-binding domains was insulin and phosphotyrosine dependent, as previously shown. To test for significance of IRS-1 with regard to GH signaling, IRS- and GHR-deficient 32D cells were stably reconstituted with the rabbit (r) GHR, either alone (32D-rGHR) or with IRS-1 (32D-rGHR-IRS-1). As assayed by three independent methods, GH induced proliferation in 32D-rGHR cells, even in the absence of transfected IRS-1. Notably, however, GH-induced proliferation was markedly enhanced in cells expressing IRS-1. Similarly, GH-induced mitogen-activated protein kinase activation was significantly augmented in IRS-1-expressing cells relative to that in cells harboring no IRS-1. These results indicate that IRS-1 enhances GH-induced proliferative signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Cell Division/drug effects , Human Growth Hormone/pharmacology , Phosphoproteins/pharmacology , Proto-Oncogene Proteins , 3T3 Cells , Animals , Binding Sites , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Glutathione Transferase/genetics , Humans , Immunosorbent Techniques , Insulin Receptor Substrate Proteins , Janus Kinase 2 , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Rabbits , Rats , Receptor, Insulin/metabolism , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Recombinant Fusion Proteins/metabolism , Shc Signaling Adaptor Proteins , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1
6.
J Biol Chem ; 274(17): 12075-80, 1999 Apr 23.
Article in English | MEDLINE | ID: mdl-10207032

ABSTRACT

CSF-1 is equipotent to insulin in its ability to stimulate 2-[3H]deoxyglucose uptake in 3T3-L1 adipocytes expressing the colony stimulating factor-1 receptor/insulin receptor chimera (CSF1R/IR). However, CSF-1-stimulated glucose uptake and glycogen synthesis is reduced by 50% in comparison to insulin in 3T3-L1 cells expressing a CSF1R/IR mutated at Tyr960 (CSF1R/IRA960). CSF-1-treated adipocytes expressing the CSF1R/IRA960 were impaired in their ability to phosphorylate insulin receptor substrate 1 (IRS-1) but not in their ability to phosphorylate IRS-2. Immunoprecipitation of IRS proteins followed by Western blotting revealed that the intact CSF1R/IR co-precipitates with IRS-2 from CSF-1-treated cells. In contrast, the CSF1R/IRA960 co-precipitates poorly with IRS-2. These observations suggest that Tyr960 is important for interaction of the insulin receptor cytoplasmic domain with IRS-2, but it is not essential to the ability of the insulin receptor tyrosine kinase to use IRS-2 as a substrate. These observations also suggest that in 3T3-L1 adipocytes, tyrosine phosphorylation of IRS-2 by the insulin receptor tyrosine kinase is not sufficient for maximal stimulation of receptor-regulated glucose transport or glycogen synthesis.


Subject(s)
Adipocytes/metabolism , Glucose/metabolism , Phosphoproteins/metabolism , Receptor, Insulin/metabolism , Tyrosine/metabolism , 3T3 Cells , Animals , Biological Transport , Cell Membrane/metabolism , Cytoplasm/metabolism , Enzyme Activation , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Ligands , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Receptor, Insulin/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Recombinant Fusion Proteins/metabolism , Tyrosine/genetics
7.
J Immunol ; 162(8): 4864-75, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10202031

ABSTRACT

In FDCP2 myeloid cells, IL-4 activated cyclic nucleotide phosphodiesterases PDE3 and PDE4, whereas IL-3, granulocyte-macrophage CSF (GM-CSF), and phorbol ester (PMA) selectively activated PDE4. IL-4 (not IL-3 or GM-CSF) induced tyrosine phosphorylation of insulin-receptor substrate-2 (IRS-2) and its association with phosphatidylinositol 3-kinase (PI3-K). TNF-alpha, AG-490 (Janus kinase inhibitor), and wortmannin (PI3-K inhibitor) inhibited activation of PDE3 and PDE4 by IL-4. TNF-alpha also blocked IL-4-induced tyrosine phosphorylation of IRS-2, but not of STAT6. AG-490 and wortmannin, not TNF-alpha, inhibited activation of PDE4 by IL-3. These results suggested that IL-4-induced activation of PDE3 and PDE4 was downstream of IRS-2/PI3-K, not STAT6, and that inhibition of tyrosine phosphorylation of IRS molecules might be one mechnism whereby TNF-alpha could selectively regulate activities of cytokines that utilized IRS proteins as signal transducers. RO31-7549 (protein kinase C (PKC) inhibitor) inhibited activation of PDE4 by PMA. IL-4, IL-3, and GM-CSF activated mitogen-activated protein (MAP) kinase and protein kinase B via PI3-K signals; PMA activated only MAP kinase via PKC signals. The MAP kinase kinase (MEK-1) inhibitor PD98059 inhibited IL-4-, IL-3-, and PMA-induced activation of MAP kinase and PDE4, but not IL-4-induced activation of PDE3. In FDCP2 cells transfected with constitutively activated MEK, MAP kinase and PDE4, not PDE3, were activated. Thus, in FDCP2 cells, PDE4 can be activated by overlapping MAP kinase-dependent pathways involving PI3-K (IL-4, IL-3, GM-CSF) or PKC (PMA), but selective activation of PDE3 by IL-4 is MAP kinase independent (but perhaps IRS-2/PI3-K dependent).


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Hematopoietic Stem Cells/enzymology , Interleukin-3/pharmacology , Interleukin-4/pharmacology , Protein Serine-Threonine Kinases , Androstadienes/pharmacology , Animals , Biological Transport , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Activation/drug effects , Enzyme Activation/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Insulin/metabolism , STAT6 Transcription Factor , Signal Transduction/immunology , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Trans-Activators/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/metabolism , Tyrphostins/pharmacology , Wortmannin
8.
Mol Cell Biol ; 19(5): 3816-28, 1999 May.
Article in English | MEDLINE | ID: mdl-10207105

ABSTRACT

In the present study, we investigated the potential role of insulin-like growth factor I (IGF-I) receptor (IGF-IR) in cell proliferation by overexpressing it in 32D myeloid progenitor cells. The overexpression of IGF-IR caused the transfectants to proliferate in response to IGF-I in the absence of insulin receptor substrate (IRS) expression. The activation of overexpressed wild-type IGF-IR, but not that of an ATP-binding mutant of IGF-IR, resulted in the increased tyrosine phosphorylation of several intracellular proteins, including SHC, Src homology 2-containing inositol-5-phosphatase, protein kinase C-delta, and Erk2. Grb2 association with SHC and mitogen-activated protein kinase (MAPK) activity was also enhanced in response to IGF-I stimulation. Interestingly, the stimulation of the IGF-IR transfectants with interleukin 4 (IL-4) also resulted in strong mitogenesis independent of IRS expression. Moreover, IGF-I and/or IL-4 induced long-term cell growth of the IGF-IR transfectants. IL-4 was able to synergize with IGF-I for DNA synthesis, even in the parental 32D cells and a pro-B-cell line, Baf3, indicating the physiological importance of the two growth factors in hematopoietic cell proliferation. IL-4 stimulation of the IGF-IR transfectants resulted in enhanced tyrosine phosphorylation of SHC, Erk2, and signal transducer and activator of transcription 6 (STAT6) proteins. Both IL-4 and IGF-I were able to induce c-myc early response gene expression, and this expression was maximal in the presence of both factors. Finally, we demonstrated that a MAPK kinase inhibitor was able to suppress mitogenesis of the IGF-IR transfectants in response to IGF-I and/or IL-4. Together, our results suggest that IL-4 synergizes with IGF-I for hematopoietic cell proliferation, likely through cross talk between SHC/Grb2/MAPK and STAT6 pathways and through c-myc gene up-regulation.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Cell Division , Insulin-Like Growth Factor I/pharmacology , Interleukin-4/metabolism , Phosphoproteins/genetics , Receptor, IGF Type 1/genetics , Stem Cells/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Enzyme Activation , GRB2 Adaptor Protein , Gene Expression Regulation/genetics , Genes, myc/genetics , Hematopoiesis , Insulin Receptor Substrate Proteins , Mitogens/metabolism , Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, IGF Type 1/metabolism , STAT6 Transcription Factor , Shc Signaling Adaptor Proteins , Trans-Activators/metabolism , Transfection
9.
Oncogene ; 18(10): 1855-66, 1999 Mar 11.
Article in English | MEDLINE | ID: mdl-10086340

ABSTRACT

The role of cbl-b in signaling by the epidermal growth factor receptor (EGFR) was studied and compared with c-cbl. We demonstrate in vivo, that cbl-b, like c-cbl, is phosphorylated and recruited to the EGFR upon EGF stimulation and both cbl proteins can bind to the Grb2 adaptor protein. To investigate the functional role of cbl proteins in EGFR signaling, we transfected cbl-b or c-cbl into 32D cells overexpressing the EGFR (32D/EGFR). This cell line is absolutely dependent on exogenous IL-3 or EGF for sustained growth. 32D/EGFR cells overexpressing cbl-b showed markedly inhibited growth in EGF compared to c-cbl transfectants and vector controls. This growth inhibition by cbl-b was the result of a dramatic increase in the number of cells undergoing apoptosis. Consistent with this finding, cbl-b overexpression markedly decreased the amplitude and duration of AKT activation upon EGF stimulation compared to either vector controls or c-cbl overexpressing cells. In addition, the duration of EGF mediated MAP kinase and Jun kinase activation in cells overexpressing cbl-b is shortened. These data demonstrate that cbl-b inhibits EGF-induced cell growth and that cbl-b and c-cbl have distinct roles in EGF mediated signaling.


Subject(s)
ErbB Receptors/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligases , Animals , Cell Line, Transformed , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-cbl , Rabbits
10.
Int J Oncol ; 13(5): 1061-7, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9772300

ABSTRACT

Ligands of the EGF/Heregulin family control the growth of epithelial cells by binding to receptors of the erbB family. By searching a large database of cDNA sequences at Human Genome Sciences Inc. we have identified a new encoded protein sequence containing all the conserved elements of the EGF/Heregulin family. The same sequence has recently been independently identified as NRG-3. The EGF-like domain of NRG-3 was generated as a recombinant protein in E. coli and used to test the specificity of receptor binding. In human breast cancer cells and in 32D cells transfected by erbB family members, NRG-3 activated multiple erbB family members. These include EGF receptor (erbB1) and erbB4 when expressed individually and erbB2 and erbB3 when expressed together. Recombinant NRG-3 altered the growth of human breast cancer cells growing in vitro. NRG-3 was expressed in cell lines derived from breast cancer. These results indicate that NRG-3 is a potential regulator of normal and malignant breast epithelial cells in vivo.


Subject(s)
Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Oncogene Proteins v-erbB/metabolism , Amino Acid Sequence , Carrier Proteins/chemistry , Cell Division , Cell Line , Databases, Factual , Epidermal Growth Factor/chemistry , Epithelial Cells/metabolism , Humans , Molecular Sequence Data , Neuregulins , Oncogene Proteins v-erbB/genetics , Protein Conformation , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tumor Cells, Cultured
11.
EMBO J ; 17(20): 5948-63, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9774339

ABSTRACT

Virulence of poxviruses, the causative agents of smallpox, depends on virus-encoded growth factors related to the mammalian epidermal growth factor (EGF). Here we report that the growth factors of Shope fibroma virus, Myxoma virus and vaccinia virus (SFGF, MGF and VGF) display unique patterns of specificity to ErbB receptor tyrosine kinases; whereas SFGF is a broad-specificity ligand, VGF binds primarily to ErbB-1 homodimers, and the exclusive receptor for MGF is a heterodimer comprised of ErbB-2 and ErbB-3. In spite of 10- to 1000-fold lower binding affinity to their respective receptors, the viral ligands are mitogenically equivalent or even more potent than their mammalian counterparts. This remarkable enhancement of cell growth is due to attenuation of receptor degradation and ubiquitination, which leads to sustained signal transduction. Our results imply that signal potentiation and precise targeting to specific receptor combinations contribute to cell transformation at sites of poxvirus infection, and they underscore the importance of the often ignored low-affinity ligand-receptor interactions.


Subject(s)
Poxviridae/pathogenicity , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Dimerization , ErbB Receptors/metabolism , ErbB Receptors/physiology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/virology , Humans , Mice , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/physiology , Receptor, ErbB-3 , Receptor, ErbB-4
12.
J Biol Chem ; 273(41): 26908-14, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9756938

ABSTRACT

Activation of tyrosine kinases by numerous growth factor and cytokine receptors leads to tyrosine phosphorylation of the insulin receptor substrate (IRS)-proteins. Tyrosine-phosphorylated motifs on the IRS proteins bind to the SH2 domains in proteins that mediate downstream signals, including phosphatidylinositol 3'-kinase, GRB-2, and SHP-2. We investigated the function of the two SHP-2 binding COOH-terminal tyrosines of IRS-1 by replacing them with phenylalanine (IRS-1(FCT)). IRS-1(FCT) failed to bind SHP-2 or mediate its tyrosine phosphorylation during insulin stimulation. Although several reports suggest a critical role for SHP-2 in insulin stimulated mitogen-activated protein kinase activation and cell proliferation, IRS-1(FCT) mediated these effects normally in 32D cells. Indeed, IRS-1(FCT) exhibited increased tyrosine phosphorylation, phosphatidylinositol 3'-kinase binding and activation of protein synthesis in response to insulin. These results suggest that SHP-2 attentuates the phosphorylation and downstream signal transmission of IRS-1 and that the interaction of IRS-1 and SHP-2 is an important regulatory event which attenuates insulin metabolic responses.


Subject(s)
Insulin/metabolism , Phosphoproteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Tyrosine/metabolism , Animals , CHO Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cricetinae , Enzyme Activation , Humans , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/chemistry , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Rats
13.
Mol Cell Biol ; 18(10): 5888-98, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9742106

ABSTRACT

To investigate the potential role of protein kinase C-delta (PKC-delta) in insulin-like growth factor I receptor (IGF-IR)-mediated cell transformation, an oncogenic gag-IGF-IR beta-fusion receptor lacking the entire extracellular domain, which was designated NM1, and a full-length IGF-IR were coexpressed with either wild-type PKC-delta (PKC-deltaWT) or an ATP-binding mutant of PKC-delta (PKC-deltaK376R) in NIH 3T3 fibroblasts. While overexpression of PKC-deltaWT did not affect NM1- and IGF-IR-induced focus and colony formation of NIH 3T3 cells, expression of PKC-deltaK376R severely impaired these events. In contrast, NM1-mediated cell growth in monolayer was not affected by coexpressing PKC-deltaK376R. PKC-deltaWT and PKC-deltaK376R were constitutively phosphorylated on a tyrosine residue(s) in the NM1- and IGF-IR-expressing cells and were associated with them in an IGF-I-independent manner. Activated IGF-IR was able to phosphorylate purified PKC-delta in vitro and stimulated its kinase activity. Furthermore, the level of endogenous PKC-delta protein was up-regulated through transcriptional activation in response to long-term IGF-IR activation. Taken together, our results demonstrate that PKC-delta plays an important role in IGF-IR-mediated cell transformation, probably via association of the receptor with PKC-delta and its activation through protein up-regulation and tyrosine phosphorylation. Competition with endogenous PKC-delta for NM1 and IGF-IR association by PKC-deltaK376R is probably an important mechanism underlying the PKC-deltaK376R-mediated inhibition of cell transformation by NM1 and IGF-IR.


Subject(s)
Cell Transformation, Neoplastic , Gene Products, gag/genetics , Isoenzymes/metabolism , Protein Kinase C/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , 3T3 Cells , Animals , Cell Division , Enzyme Activation , Gene Expression , Isoenzymes/genetics , Mice , Mutation , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C-delta , Protein-Tyrosine Kinases/metabolism , Receptor, IGF Type 1/genetics , Tyrosine/metabolism
14.
Mol Cell Biol ; 18(10): 6090-101, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9742126

ABSTRACT

The recently isolated second family of neuregulins, NRG2, shares its primary receptors, ErbB-3 and ErbB-4, and induction of mammary cell differentiation with NRG1 isoforms, suggesting functional redundancy of the two growth factor families. To address this possibility, we analyzed receptor specificity of NRGs by using an engineered cellular system. The activity of isoform-specific but partly overlapping patterns of specificities that collectively activate all eight ligand-stimulatable ErbB dimers was revealed. Specifically, NRG2-alpha [corrected], like NRG1-beta [corrected], emerges as a narrow-specificity ligand, whereas NRG2-beta [corrected] is a pan-ErbB ligand that binds with different affinities to all receptor combinations, including those containing ErbB-1, but excluding homodimers of ErbB-2. The latter protein, however, displayed cooperativity with the direct NRG receptors. Apparently, signaling by all NRGs is funneled through the mitogen-activated protein kinase (MAPK). However, the duration and potency of MAPK activation depend on the identity of the stimulatory ligand-receptor ternary complex. We conclude that the NRG-ErbB network represents a complex and nonredundant machinery developed for fine-tuning of signal transduction.


Subject(s)
ErbB Receptors/metabolism , Glycoproteins/metabolism , Nerve Growth Factors/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Enzyme Activation , ErbB Receptors/biosynthesis , Glycoproteins/pharmacology , Isomerism , Ligands , Nerve Growth Factors/pharmacology , Neuregulins , Phosphorylation , Proto-Oncogene Proteins/biosynthesis , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-3 , Receptor, ErbB-4
15.
Cancer Res ; 58(15): 3415-22, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9699674

ABSTRACT

The epidermal growth factor-like receptor tyrosine kinase (ErbB) family is frequently overexpressed in a variety of human carcinomas, including breast cancer. To assist in characterizing the role of ErbB-4 in breast cancer, we generated three specific hammerhead ribozymes targeted to the ErbB-4 mRNA. These ribozymes, Rz6, Rz21, and Rz29, efficiently catalyzed the specific cleavage of ErbB-4 message in a cell-free system. We demonstrated that the neuregulin-induced mitogenic effect was abolished in ribozyme Rz29- and Rz6-transfected 32D/ErbB-4 cells. Inhibition of mitogenesis was characterized by ribozyme-mediated down-regulation of ErbB-4 expression. In addition, we provide the first evidence that different threshold levels of ErbB-4 expression and activation correlate with different responses to neuregulin stimulation. High levels of ErbB-4 expression, phosphorylation, and homodimerization are necessary for neuregulin-stimulated, interleukin 3-independent cell proliferation in the 32D/E4 cells. In the case of Rz29-transfected 32D/E4 cells, low levels of ErbB-4 expression allowed neuregulin-induced phosphorylation but were insufficient to couple the activated receptor to cellular signaling. Furthermore, expression of the functional ErbB-4 ribozyme in T47D human breast carcinoma cells led to a down-regulation of endogenous ErbB-4 expression and a reduction of anchorage-independent colony formation. These studies support the use of ErbB-4 ribozymes to define the role of ErbB-4 receptors in human cancers.


Subject(s)
ErbB Receptors/physiology , Glycoproteins/pharmacology , RNA, Catalytic/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Division/physiology , Cell-Free System , Cells, Cultured , DNA/biosynthesis , Down-Regulation , ErbB Receptors/biosynthesis , ErbB Receptors/metabolism , Hematopoietic System/cytology , Hematopoietic System/enzymology , Humans , Interleukin-3/pharmacology , Mice , Neuregulins , Phosphorylation , RNA, Catalytic/pharmacology , RNA, Messenger/metabolism , Receptor, ErbB-4 , Stimulation, Chemical , Substrate Specificity
16.
Proc Natl Acad Sci U S A ; 95(12): 6809-14, 1998 Jun 09.
Article in English | MEDLINE | ID: mdl-9618494

ABSTRACT

Interleukin 3-dependent murine 32D cells do not detectably express members of the ErbB receptor family and do not proliferate in response to known ligands for these receptors. 32D transfectants were generated expressing human ErbB4 alone (32D.E4) or with ErbB2 (32D.E2/E4). Epidermal growth factor (EGF), neuregulin 1-beta (NRG1-beta), betacellulin (BTC), transforming growth factor-alpha (TGF-alpha), heparin binding-EGF (HB-EGF), and amphiregulin were analyzed for their ability to mediate mitogenesis in these transfectants. 32D.E4 responded mitogenically to NRG1-beta and BTC. Surprisingly, EGF also induced significant DNA synthesis and TGF-alpha was negligibly mitogenic on 32D.E4 cells, whereas HB-EGF and amphiregulin were inactive. Although coexpression of ErbB2 with ErbB4 in 32D.E2/E4 cells did not significantly alter DNA synthesis in response to NRG1-beta or BTC, it greatly enhanced mitogenesis elicited by EGF and TGF-alpha and unmasked the ability of HB-EGF to induce proliferation. EGF-related ligands that exhibited potent mitogenic activity on 32D.E2/E4 cells at low concentrations induced adherence, morphological alterations, and up-regulation of the Mac-1 integrin and FcgammaRII/III at higher concentrations. While 125I-EGF could be specifically crosslinked to both 32D.E4 and 32D.E2/E4 cells, its crosslinking capacity was greatly enhanced in the cotransfected cells. The ability of the various ligands to mediate proliferation and/or adhesion in the two transfectants correlated with their capacity to induce substrate tyrosine phosphorylation and to initiate and sustain activation of mitogen-activated protein kinase. We conclude that the ability of ErbB4 to mediate signal transduction through EGF-like ligands is broader than previously assumed and can be profoundly altered by the concomitant expression of ErbB2.


Subject(s)
ErbB Receptors/genetics , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins , Receptor, ErbB-2/genetics , Signal Transduction/genetics , Amphiregulin , Animals , Antineoplastic Agents/pharmacology , Betacellulin , Cell Line , EGF Family of Proteins , Epidermal Growth Factor/pharmacology , Gene Expression Regulation/drug effects , Glycoproteins/pharmacology , Growth Substances/pharmacology , Heparin-binding EGF-like Growth Factor , Humans , Ligands , Mice , Neuregulins , Receptor, ErbB-4 , Signal Transduction/drug effects , Transfection , Transforming Growth Factor alpha/pharmacology
17.
J Biol Chem ; 273(17): 10496-505, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9553109

ABSTRACT

The ErbB signaling network consists of four transmembrane receptor tyrosine kinases and more than a dozen ligands sharing an epidermal growth factor (EGF) motif. The multiplicity of ErbB-specific ligands is incompletely understood in terms of signal specificity because all ErbB molecules signal through partially overlapping pathways. Here we addressed the action of epiregulin, a recently isolated ligand of ErbB-1. By employing a set of factor-dependent cell lines engineered to express individual ErbBs or their combinations, we found that epiregulin is the broadest specificity EGF-like ligand so far characterized: not only does it stimulate homodimers of both ErbB-1 and ErbB-4, it also activates all possible heterodimeric ErbB complexes. Consistent with its relaxed selectivity, epiregulin binds the various receptor combinations with an affinity that is approximately 100-fold lower than the affinity of ligands with more stringent selectivity, including EGF. Nevertheless, epiregulin's action upon most receptor combinations transmits a more potent mitogenic signal than does EGF. This remarkable discrepancy between binding affinity and bioactivity is permitted by a mechanism that prevents receptor down-regulation, and results in a weak, but prolonged, state of receptor activation.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Animals , CHO Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation , Cell Line , Cricetinae , Dimerization , Down-Regulation , Enzyme Activation , Epiregulin , Ligands , Phosphorylation , Signal Transduction , Tyrosine/metabolism
18.
Clin Cancer Res ; 4(4): 993-1004, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9563895

ABSTRACT

Growth factor receptors provide unique opportunities for development of targeted anticancer therapy. Members of the type I receptor tyrosine kinase family, including epidermal growth factor (EGF) receptor (EGFR) and ErbB-2/neu, are often overexpressed in various human cancer cells, including breast. Recently, it has been shown that both ErbB-3 and ErbB-4 are receptors for heregulin (HRG)/Neu differentiation factor. Eight chimeric toxins composed of the extracellular and EGF-like domains of four different HRG isoforms and truncated Pseudomonas exotoxin (PE38KDEL) were constructed. The fusion proteins exhibited activity similar to the native HRG in inducing ErbB receptors phosphorylation. The EGF-like domain of HRG13 and HRGbeta2 fused to PE38KDEL showed the highest cytotoxic activity, with a IC50 of < or = 0.001 ng/ml. The alpha isoforms that were fused to PE38KDEL were 100-fold less active than the beta isoforms. The HRG-Pseudomonas exotoxin (PE) toxins show extremely high activity against cells expressing ErbB-4 receptor, alone or together with other members of the ErbB receptor family. Cells that do not express ErbB-4 but express ErbB-3 receptor, together with the ErbB-2 or EGFR, exhibited moderate sensitivity to HRG-PE toxins. HRG-PE toxins have little or no activity against cells expressing EGFR, ErbB-2, or ErbB-3 alone. More than an 80% tumor regression was achieved by intratumor injection of 1 microg of fusion proteins per day for 5 days. Continuous i.p. administration of EGF-like domain of HRGbeta1-PE38KDEL for 7 days via a miniosmotic pump at a dose of 40 microg/kg/day inhibited the growth of ErbB-4 receptor positive but not ErbB-4 receptor negative cell lines in athymic nude mice. We conclude that there is therapeutic potential of HRG-PE toxins in the therapy of cancers overexpressing the ErbB-4 or ErbB-2 plus ErbB-3 receptors.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/drug effects , Exotoxins/pharmacology , Genes, erbB/drug effects , Glycoproteins/pharmacology , Proto-Oncogene Proteins/drug effects , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Colony-Forming Units Assay , Exotoxins/pharmacokinetics , Exotoxins/therapeutic use , Female , Glycoproteins/pharmacokinetics , Glycoproteins/therapeutic use , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/pharmacology , Immunotoxins/therapeutic use , Mice , Mice, Nude , Phosphorylation , Receptor, ErbB-3 , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Stomach Neoplasms/drug therapy , Tumor Cells, Cultured/drug effects , Tyrosine/metabolism
19.
Proc Natl Acad Sci U S A ; 95(1): 172-7, 1998 Jan 06.
Article in English | MEDLINE | ID: mdl-9419348

ABSTRACT

Interleukin (IL)-4-mediated nuclear signaling by Stat6 has been implicated in lymphoid cell proliferation and the transcriptional activation of genes encoding major histocompatability complex (MHC) class II molecules and Fc receptors. To investigate IL-4-mediated transcriptional events, we cloned two naturally occurring human Stat6 isoforms, Stat6b and Stat6c, that encoded an NH2-terminal truncation or an SH2 domain deletion, respectively. Stat6 variant mRNAs were differentially expressed in many human tissues. To elucidate the biologic role of each isoform, we examined the consequences of overexpression in IL-4-responsive FDC-P2 cells. Stat6 and Stat6b (to a lesser extent) enhanced DNA synthesis, up-regulated endogenous MHC class II and Fcgamma receptors, and became tyrosine phosphorylated in response to IL-4 stimulation. In contrast, Stat6c, which lacks functionally critical SH2 domain residues, unexpectedly inhibited IL-4-mediated mitogenesis and cell surface antigen expression and was not tyrosine phosphorylated. Although Stat6c only modestly diminished endogenous Stat6 tyrosine phosphorylation, it abolished endogenous Stat6 FcgammaRI and Iepsilon DNA binding activity and FcgammaRI-luciferase reporter transcriptional activation. Our results indicate that the molecular mechanism of inhibition by Stat6c was due to suppression of endogenous Stat6 dimer formation. Thus, Stat6b and Stat6c are naturally occurring attenuated and dominant negative Stat6 variants, respectively, that affect IL-4-mediated biologic responses through differential transcriptional regulation.


Subject(s)
Interleukin-4/physiology , Signal Transduction , Trans-Activators/antagonists & inhibitors , Amino Acid Sequence , Cell Line , DNA/metabolism , DNA, Complementary/isolation & purification , Dimerization , Humans , Molecular Sequence Data , Phosphorylation , RNA, Messenger/metabolism , STAT6 Transcription Factor , Spectrometry, Fluorescence , Thymidine/metabolism , Trans-Activators/genetics , Transcriptional Activation , Tyrosine/metabolism
20.
EMBO J ; 16(18): 5608-17, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9312020

ABSTRACT

Interleukin-3 (IL-3)-dependent murine 32D cells do not detectably express epidermal growth factor receptors (EGFRs) and do not proliferate in response to EGF, heregulin (HRG) or other known EGF-like ligands. Here, we report that EGF specifically binds to and can be crosslinked to 32D transfectants co-expressing ErbB2 and ErbB3 (32D.E2/E3), but not to transfectants expressing either ErbB2 or ErbB3 individually. [125I]EGF-crosslinked species detected in 32D. E2/E3 cells were displaced by HRG and betacellulin (BTC) but not by other EGF-like ligands that were analyzed. EGF, BTC and HRG also induced receptor tyrosine phosphorylation, activation of downstream signaling molecules and proliferation of 32D.E2/E3 cells. 32D transfectants were also generated which expressed an ErbB3-EGFR chimera alone (32D.E3-E1) or in combination with ErbB2 (32D. E2/E3-E1). While HRG stimulation of 32D.E3-E1 cells resulted in DNA synthesis and receptor phosphorylation, EGF and BTC were inactive. However, EGF and BTC were as effective as HRG in mediating signaling when ErbB2 was co-expressed with the chimera in the 32D.E2/E3-E1 transfectant. These results provide evidence that ErbB2/ErbB3 binding sites for EGF and BTC are formed by a previously undescribed mechanism that requires co-expression of two distinct receptors. Additional data utilizing MDA MB134 human breast carcinoma cells, which naturally express ErbB2 and ErbB3 in the absence of EGFRs, supported the results obtained employing 32D cells and suggest that EGF and BTC may contribute to the progression of carcinomas that co-express ErbB2 and ErbB3.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/physiology , Growth Substances/pharmacology , Intercellular Signaling Peptides and Proteins , Proto-Oncogene Proteins/physiology , Receptor, ErbB-2/physiology , Signal Transduction/drug effects , Animals , Betacellulin , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Enzyme Activation , Epidermal Growth Factor/metabolism , ErbB Receptors/biosynthesis , ErbB Receptors/drug effects , Humans , Kinetics , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Polymerase Chain Reaction , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/drug effects , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/drug effects , Receptor, ErbB-3 , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/physiology , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...