Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 264
Filter
1.
Clin Pharmacol Ther ; 95(4): 413-5, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24413269

ABSTRACT

Our understanding of cancer has grown considerably with recent advances in high-throughput genome and transcriptome sequencing, but techniques to comprehensively analyze protein activity are still in development. Methods to quantitatively measure the activation of signaling pathways within tumors at baseline and following therapeutic intervention will prove critical to the design of proper treatment regimens. Focusing on breast cancer, we present such a method to understand kinase signaling using multiplexed kinase inhibitor beads coupled with mass spectrometry (MIB/MS).


Subject(s)
Breast Neoplasms/pathology , Mass Spectrometry/methods , Protein Kinase Inhibitors/pharmacology , Adaptation, Physiological , Breast Neoplasms/therapy , Female , Gene Expression Profiling , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Signal Transduction/physiology , Transcriptome
2.
Oncogene ; 31(34): 3889-900, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22139075

ABSTRACT

Analysis of patient tumors suggests that multiple MAP3 kinases (MAP3Ks) are critical for growth and metastasis of cancer cells. MAP3Ks selectively control the activation of extracellular signal-regulated kinase 1/2 (ERK1/2), Jun N-terminal kinase (JNK), p38 and ERK5 in response to receptor tyrosine kinases and GTPases. We used MDA-MB-231 cells because of their ability to metastasize from the breast fat pad to distant lymph nodes for an orthotopic xenograft model to screen the function of seven MAP3Ks in controlling tumor growth and metastasis. Stable short hairpin RNA (shRNA) knockdown was used to inhibit the expression of each of the seven MAP3Ks, which were selected for their differential regulation of the MAPK network. The screen identified two MAP3Ks, MEKK2 and MLK3, whose shRNA knockdown caused significant inhibition of both tumor growth and metastasis. Neither MEKK2 nor MLK3 have been previously shown to regulate tumor growth and metastasis in vivo. These results demonstrated that MAP3Ks, which differentially activate JNK, p38 and ERK5, are necessary for xenograft tumor growth and metastasis of MDA-MB-231 tumors. The requirement for MAP3Ks signaling through multiple MAPK pathways explains why several members of the MAPK network are activated in cancer. MEKK2 was required for epidermal growth factor receptor and Her2/Neu activation of ERK5, with ERK5 being required for metastasis. Loss of MLK3 expression increased mitotic infidelity and apoptosis in vitro. Knockdown of MEKK2 and MLK3 resulted in increased apoptosis in orthotopic xenografts relative to control tumors in mice, inhibiting both tumor growth and metastasis; MEKK2 and MLK3 represent untargeted kinases in tumor biology for potential therapeutic development.


Subject(s)
Breast Neoplasms/genetics , Mammary Neoplasms, Experimental/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , RNA Interference , Animals , Apoptosis/genetics , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/genetics , HEK293 Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , MAP Kinase Kinase Kinase 2/genetics , MAP Kinase Kinase Kinase 2/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Metastasis , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Burden/genetics , Mitogen-Activated Protein Kinase Kinase Kinase 11
3.
Theriogenology ; 71(4): 707-15, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-18995894

ABSTRACT

The objective of this experiment was to evaluate corpus luteum blood flow (CLBF) as an early indicator of pregnancy status in bovine embryo recipients. Fifty crossbred beef cows were submitted to embryo transfer on Day 7 after estrus. On Days 7, 11, 13, 15, 17, 19, 21, 26, 33, and 40, a blood sample was taken, the CL examined using a color-flow Doppler ultrasound scanner, and video was recorded of each scanning session. Ultrasound data were grouped by the first day progesterone concentrations were <1 ng/mL (indicating early embryo loss, EEL) through Day 21 (EEL-17, n=3; EEL-19, n=9; EEL-21, n=3), absence of an embryo on Days 26, 33, or 40 (late embryo loss; LEL; n=12), or remained pregnant (P; n=23). The first decrease in CLBF of EEL-17, EEL-19, and EEL-21 cows compared to P cows occurred on Days 17, 19, and 21, respectively (P<0.05). There was no difference in CLBF between LEL and P cows on Days 17, 19, and 21. Six evaluators diagnosed pregnancy from randomized video clips on Days 17, 19, and 21. Evaluators made more (P<0.004) correct diagnoses on Day 19 than Day 17. Sensitivity (82.9+/-10.1%) was not affected by day. From Days 17 to 19, diagnostic specificity increased (P=0.046) from 43.2+/-3.0 to 54.3+/-3.0% but remained unchanged thereafter. Due to low specificity and sensitivity, evaluation of CLBF alone was insufficient for early pregnancy diagnosis.


Subject(s)
Cattle/physiology , Corpus Luteum/blood supply , Embryo Transfer/veterinary , Regional Blood Flow , Ultrasonography, Doppler, Color/veterinary , Animals , Female , Pregnancy
4.
Oncogene ; 27(32): 4434-45, 2008 Jul 24.
Article in English | MEDLINE | ID: mdl-18372913

ABSTRACT

Hyperactivation of ErbB signaling is implicated in metastatic breast cancer. However, the mechanisms that cause dysregulated ErbB signaling and promote breast carcinoma cell invasion remain poorly understood. One pathway leading to ErbB activation that remains unexplored in breast carcinoma cell invasion involves transactivation by G-protein-coupled receptors (GPCRs). Protease-activated receptor-1 (PAR1), a GPCR activated by extracellular proteases, is overexpressed in invasive breast cancer. PAR1 is also proposed to function in breast cancer invasion and metastasis, but how PAR1 contributes to these processes is not known. In this study, we report that proteolytic activation of PAR1 by thrombin induces persistent transactivation of EGFR and ErbB2/HER2 in invasive breast carcinoma, but not in normal mammary epithelial cells. PAR1-stimulated EGFR and ErbB2 transactivation leads to prolonged extracellular signal-regulated kinase-1 and -2 signaling and promotes breast carcinoma cell invasion. We also show that PAR1 signaling through Galpha(i/o) and metalloprotease activity is critical for ErbB transactivation and cellular invasion. Finally, we demonstrate that PAR1 expression in invasive breast carcinoma is essential for tumor growth in vivo assessed by mammary fat pad xenografts. These studies reveal a critical role for PAR1, a receptor activated by tumor-generated proteases, in hyperactivation of ErbB signaling that promotes breast carcinoma cell invasion.


Subject(s)
Breast Neoplasms/pathology , ErbB Receptors/genetics , Receptor, ErbB-2/genetics , Receptor, PAR-1/physiology , Transcriptional Activation , ADAM Proteins/physiology , ADAM17 Protein , Animals , Cell Line, Tumor , ErbB Receptors/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Fibroblasts/physiology , GTP-Binding Protein alpha Subunits/physiology , Humans , Mice , NIH 3T3 Cells , Neoplasm Invasiveness , Signal Transduction , Thrombin/pharmacology
5.
Technol Cancer Res Treat ; 6(6): 651-4, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17994796

ABSTRACT

Arsenic trioxide (ATO, Trisenox) is a potent anti-vascular agent and significantly enhances hyperthermia and radiation response. To understand the mechanism of the anti-tumor effect in vivo we imaged the binding of a fluorescently-labeled poly-caspase inhibitor (FLIVO) in real time before and 3 h or 24 h after injection of 8 mg/kg ATO. FSaII tumors were grown in dorsal skin-fold window chambers or on the rear limb and we observed substantial poly-caspase binding associated with vascular damage induced by ATO treatment at 3 and 24 h after ATO injection. Flow cytometric analysis of cells dissociated from the imaged tumor confirmed cellular uptake and binding of the FLIVO probe. Apoptosis appears to be a major mode of cell death induced by ATO in the tumor and the use of fluorescently tagged caspase inhibitors to assess cell death in live animals appears feasible to monitor and/or confirm anti-tumor effects of therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Arsenicals/pharmacology , Caspases/metabolism , Enzyme Inhibitors , Fluorescent Dyes , Oxides/pharmacology , Animals , Arsenic Trioxide , Female , Flow Cytometry , Mice , Mice, Nude
7.
Oncogene ; 26(22): 3159-71, 2007 May 14.
Article in English | MEDLINE | ID: mdl-17496913

ABSTRACT

Mitogen-activated protein kinases (MAPKs) are members of a dynamic protein kinase network through which diverse stimuli regulate the spatio-temporal activities of complex biological systems. MAPKs regulate critical cellular functions required for homeostasis such as the expression of cytokines and proteases, cell cycle progression, cell adherence, motility and metabolism. MAPKs therefore influence cell proliferation, differentiation, survival, apoptosis and development. In vertebrates, five MAPK families are regulated by MAPK kinase kinase-MAPK kinase-MAPK (MKKK-MKK-MAPK) phosphorelay systems. There are at least 20 MKKKs that selectively phosphorylate and activate different combinations of the seven MKKs, resulting in a specific activation profile of members within the five MAPK families. MKKKs are differentially activated by upstream stimuli including cytokines, antigens, toxins and stress insults providing a mechanism to integrate the activation of different MAPKs with the cellular response to each stimulus. Thus, MKKKs can be considered as 'signaling hubs' that regulate the specificity of MAPK activation. In this review, we describe how the MKKK 'hub' function regulates the specificity of MAPK activation, highlighting MKKKs as targets for therapeutic intervention in cancer and other diseases.


Subject(s)
MAP Kinase Kinase Kinases/physiology , MAP Kinase Signaling System/physiology , Animals , Humans
8.
Oncogene ; 26(19): 2791-8, 2007 Apr 26.
Article in English | MEDLINE | ID: mdl-17057734

ABSTRACT

The controlled and specific re-activation of endogenous tumor suppressors in cancer cells represents an important therapeutic strategy to block tumor growth and subsequent progression. Other than ectopic delivery of tumor suppressor-encoded cDNA, there are no therapeutic tools able to specifically re-activate tumor suppressor genes that are silenced in tumor cells. Herein, we describe a novel approach to specifically regulate dormant tumor suppressors in aggressive cancer cells. We have targeted the Mammary Serine Protease Inhibitor (maspin) (SERPINB5) tumor suppressor, which is silenced by transcriptional and aberrant promoter methylation in aggressive epithelial tumors. Maspin is a multifaceted protein, regulating tumor cell homeostasis through inhibition of cell growth, motility and invasion. We have constructed artificial transcription factors (ATFs) made of six zinc-finger (ZF) domains targeted against 18-base pair (bp) unique sequences in the maspin promoter. The ZFs were linked to the activator domain VP64 and delivered in breast tumor cells. We found that the designed ATFs specifically interact with their cognate targets in vitro with high affinity and selectivity. One ATF was able to re-activate maspin in cell lines that comprise a maspin promoter silenced by epigenetic mechanisms. Consistently, we found that this ATF was a powerful inducer of apoptosis and was able to knock down tumor cell invasion in vitro. Moreover, this ATF was able to suppress MDA-MB-231 growth in a xenograft breast cancer model in nude mice. Our work suggests that ATFs could be used in cancer therapeutics as novel molecular switches to re-activate dormant tumor suppressors.


Subject(s)
Breast Neoplasms/genetics , Genes, Tumor Suppressor , Response Elements/physiology , Serpins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Humans , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Invasiveness/pathology , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Serpins/metabolism , Tumor Cells, Cultured , Zinc Fingers
9.
Oncogene ; 25(45): 6092-100, 2006 Oct 05.
Article in English | MEDLINE | ID: mdl-16652142

ABSTRACT

Mer (MerTK) is a receptor tyrosine kinase important in platelet aggregation, as well as macrophage cytokine secretion and clearance of apoptotic cells. Mer is not normally expressed in thymocytes or lymphocytes; however, ectopic Mer RNA transcript and protein expression is found in a subset of acute lymphoblastic leukemia cell lines and patient samples, suggesting a role in leukemogenesis. To investigate the oncogenic potential of Mer in vivo, we created a transgenic mouse line (Mer(Tg)) that expresses Mer in the hematopoietic lineage under control of the Vav promoter. Ectopic expression and activation of the transgenic Mer protein was demonstrated in lymphocytes and thymocytes of the Mer(Tg) mice. At 12-24 months of age, greater than 55% of the Mer(Tg) mice, compared to 12% of the wild type, developed adenopathy, hepatosplenomegaly, and circulating lymphoblasts. Histopathological analysis and flow cytometry were consistent with T-cell lymphoblastic leukemia/lymphoma. Mer may contribute to leukemogenesis by activation of Akt and ERK1/2 anti-apoptotic signals, which were upregulated in Mer(Tg) mice. Additionally, a significant survival advantage was noted in Mer(Tg) lymphocytes compared to wild-type lymphocytes after dexamethasone treatment. These data suggest that Mer plays a cooperative role in leukemogenesis and may be an effective target for biologically based leukemia/lymphoma therapy.


Subject(s)
Leukemia, T-Cell/genetics , Lymphoma, T-Cell/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Apoptosis , Base Sequence , DNA Primers , Flow Cytometry , Humans , Intercellular Signaling Peptides and Proteins/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , c-Mer Tyrosine Kinase
10.
Oncogene ; 25(36): 4998-5010, 2006 Aug 17.
Article in English | MEDLINE | ID: mdl-16568086

ABSTRACT

Mammary tumor cells are required to degrade the surrounding matrix and disseminate in order to metastasize, and both of these processes are controlled by a tumor cell-signaling network that remains poorly defined. MEKK1 is a MAPKKK that regulates both the extracellular signal regulated kinase (ERK1/2) and the c-Jun amino terminal kinase (JNK) signaling pathways. MEKK1 signaling regulates migration through control of cell adhesion and is required for inducible expression of urokinase-type plasminogen activator (uPA). MEKK1-deficient mice with mammary gland-targeted expression of the polyoma middle T antigen (PyMT) transgene develop primary mammary tumors at a rate and frequency similar to wild-type littermates, indicating that MEKK1 deficiency does not affect PyMT-mediated transformation. However, MEKK1-/- mice display significantly delayed tumor cell dissemination and lung metastasis. Delayed MEKK1-dependent tumor dissemination is associated with markedly reduced tumor uPA expression, gelatinase activity, and prolonged tumor basement membrane integrity. siRNA-mediated MEKK1 knockdown inhibits uPA activity, cell migration and invasion in MDA-MB-231 human breast cancer cells. Thus MEKK1 controls tumor progression by regulating both the migration and proteolysis aspects of tumor cell invasiveness. To our knowledge, this is the first example of a MAPKKK that regulates metastasis through control of tumor invasiveness.


Subject(s)
Antigens, Polyomavirus Transforming/physiology , MAP Kinase Kinase Kinase 1/physiology , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis , Animals , Base Sequence , DNA Primers , Disease Progression , Lung Neoplasms/secondary , MAP Kinase Kinase Kinase 1/genetics , Mice , Mice, Knockout , RNA, Small Interfering
11.
Curr Genomics ; 6: 501-513, 2005.
Article in English | MEDLINE | ID: mdl-23335855

ABSTRACT

The availability of complete genomes and global gene expression profiling has greatly facilitated analysis of complex genetic regulatory systems. We describe the use of a bioinformatics strategy for analyzing the cis-regulatory design of genes diferentially regulated during viral infection of a target cell. The large-scale transcriptional activity of human embryonic kidney (HEK293) cells to reovirus (serotype 3 Abney) infection was measured using the Affymetrix HU-95Av2 gene array. Comparing the 2000 base pairs of 5' upstream sequence for the most differentially expressed genes revealed highly preserved sequence regions, which we call "modules". Higher-order patterns of modules, called "super-modules", were significantly over-represented in the 5' upstream regions of transcriptionally responsive genes. These supermodules contain binding sites for multiple transcription factors and tend to define the role of genes in processes associated with reovirus infection. The supermodular design encodes a cis-regulatory logic for transducing upstream signaling for the control of expression of genes involved in similar biological processes. In the case of reovirus infection, these processes recapitulate the integrated response of cells including signal transduction, transcriptional regulation, cell cycle control, and apoptosis. The computational strategies described for analyzing gene expression data to discover cis-regulatory features and associating them with pathological processes represents a novel approach to studying the interaction of a pathogen with its target cells.

14.
J Virol ; 75(23): 11275-83, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11689607

ABSTRACT

Viral infection often perturbs host cell signaling pathways including those involving mitogen-activated protein kinases (MAPKs). We now show that reovirus infection results in the selective activation of c-Jun N-terminal kinase (JNK). Reovirus-induced JNK activation is associated with an increase in the phosphorylation of the JNK-dependent transcription factor c-Jun. Reovirus serotype 3 prototype strains Abney (T3A) and Dearing (T3D) induce significantly more JNK activation and c-Jun phosphorylation than does the serotype 1 prototypic strain Lang (T1L). T3D and T3A also induce more apoptosis in infected cells than T1L, and there was a significant correlation between the ability of these viruses to phosphorylate c-Jun and induce apoptosis. However, reovirus-induced apoptosis, but not reovirus-induced c-Jun phosphorylation, is inhibited by blocking TRAIL/receptor binding, suggesting that apoptosis and c-Jun phosphorylation involve parallel rather than identical pathways. Strain-specific differences in JNK activation are determined by the reovirus S1 and M2 gene segments, which encode viral outer capsid proteins (sigma1 and mu1c) involved in receptor binding and host cell membrane penetration. These same gene segments also determine differences in the capacity of reovirus strains to induce apoptosis, and again a significant correlation between the capacity of T1L x T3D reassortant reoviruses to both activate JNK and phosphorylate c-Jun and to induce apoptosis was shown. The extracellular signal-related kinase (ERK) is also activated in a strain-specific manner following reovirus infection. Unlike JNK activation, ERK activation could not be mapped to specific reovirus gene segments, suggesting that ERK activation and JNK activation are triggered by different events during virus-host cell interaction.


Subject(s)
Capsid Proteins , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Reoviridae/physiology , Animals , Apoptosis/physiology , Capsid/genetics , Cell Line , Enzyme Activation , Hemagglutinins , JNK Mitogen-Activated Protein Kinases , Mice , Receptors, Tumor Necrosis Factor/physiology , Reoviridae/genetics , Signal Transduction , Viral Proteins/genetics
15.
Oncogene ; 20(47): 6910-9, 2001 Oct 18.
Article in English | MEDLINE | ID: mdl-11687970

ABSTRACT

TRAIL (TNF-related apoptosis-inducing ligand) induces apoptosis in susceptible cells by binding to death receptors 4 (DR4) and 5 (DR5). TRAIL preferentially induces apoptosis in transformed cells and the identification of mechanisms by which TRAIL-induced apoptosis can be enhanced may lead to novel cancer chemotherapeutic strategies. Here we show that reovirus infection induces apoptosis in cancer cell lines derived from human breast, lung and cervical cancers. Reovirus-induced apoptosis is mediated by TRAIL and is associated with the release of TRAIL from infected cells. Reovirus infection synergistically and specifically sensitizes cancer cell lines to killing by exogenous TRAIL. This sensitization both enhances the susceptibility of previously resistant cell lines to TRAIL-induced apoptosis and reduces the amount of TRAIL needed to kill already sensitive lines. Sensitization is not associated with a detectable change in the expression of TRAIL receptors in reovirus-infected cells. Sensitization is associated with an increase in the activity of the death receptor-associated initiator caspase, caspase 8, and is inhibited by the peptide IETD-fmk, suggesting that reovirus sensitizes cancer cells to TRAIL-induced apoptosis in a caspase 8-dependent manner. Reovirus-induced sensitization of cells to TRAIL is also associated with increased cleavage of PARP, a substrate of the effector caspases 3 and 7.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Caspases/physiology , Membrane Glycoproteins/pharmacology , Neoplasms/pathology , Neoplasms/virology , Orthoreovirus, Mammalian/physiology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Caspase 8 , Caspase 9 , Doxorubicin/pharmacology , HeLa Cells , Humans , Membrane Glycoproteins/physiology , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerases/metabolism , RNA, Neoplasm/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/physiology
16.
Biotechniques ; 31(3): 608-10, 612, 614, passim, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11570504

ABSTRACT

Apoptosis is dependent on the activation of a group of proteolytic enzymes called caspases. Caspase activation can be detected by immunoblotting using caspase-specific antibodies or by caspase activity measurement employing pro-fluorescent substrates that become fluorescent upon cleavage by the caspase. Most of these methods require the preparation of cell extracts and, therefore, are not suitable for the detection of active caspases within the living cell. Using FAM-VAD-FMK, we have developed a simple and sensitive assay for the detection of caspase activity in living cells. FAM-VAD-FMK is a carboxyfluorescein (FAM) derivative of benzyloxycarbonyl-valine-alanine-aspartic acid-fluoromethyl ketone (zVAD-FMK), which is a potent broad-spectrum inhibitor of caspases. FAM-VAD-FMK enters the cell and irreversibly binds to activated caspases. Cells containing bound FAM-VAD-FMK can be analyzed by flow cytometry, fluorescence microscopy, or a fluorescence plate reader. Using FAM-VAD-FMK, we have measured caspase activation in live non-adherent and adherent cells. We show that FAM-VAD-FMK labeled Jurkat and HeLa cells that had undergone apoptosis following treatment with camptothecin or staurosporine. Non-stimulated negative control cells were not stained. Pretreatment with the general caspase inhibitor zVAD-FMK blocked caspase-specific staining in induced Jurkat and HeLa cells. Pretreatment of staurosporine-induced Jurkat cells with FAM-VAD-FMK inhibited affinity labeling of caspase-3, -6, and -7, blocked caspase-specific cell staining, and led to the inhibition of apoptosis. In contrast, the fluorescent control inhibitor FAM-FA-FMK had no effect. Measurement of caspase activation in 96-well plates showed a 3- to 5-fold increase in FAM-fluorescence in staurosporine-treated cells compared to control cells. In summary, we show that FAM-VAD-FMK is a versatile and specific tool for detecting activated caspases in living cells.


Subject(s)
Caspase Inhibitors , Caspases/metabolism , Enzyme Inhibitors , Fluorescent Dyes , Affinity Labels , Amino Acid Chloromethyl Ketones , Apoptosis/drug effects , Camptothecin/pharmacology , Enzyme Activation , Flow Cytometry , Fluoresceins , HeLa Cells , Humans , Jurkat Cells , Microscopy, Fluorescence , Staurosporine/pharmacology
17.
Biochim Biophys Acta ; 1547(1): 167-73, 2001 May 05.
Article in English | MEDLINE | ID: mdl-11343802

ABSTRACT

MEKK2 and MEKK3 are mitogen-activated protein kinase kinase kinases (MAP3 kinases) of 70 and 71 kDa respectively that are markedly homologous (94%) in their kinase domains. Both MEKK2 and MEKK3 are able to activate the Jun kinase pathway in vivo. However, following routine immunoprecipitation in Triton X-100, MEKK2 but not MEKK3 is able to effectively phosphorylate both SEK-1 and MEK-1 and to undergo autophosphorylation. Unexpectedly, both MEKK2 and MEKK3 are functional in an in vitro kinase assay when cells are solubilized with the closely related detergent, NP-40. Given the high homology between these kinases, we set out to relate this differential sensitivity to Triton X-100 to differences in primary structure. A set of chimeric molecules were generated and the loss of activity in Triton X-100 mapped to kinase domain II/III and specifically to serine 390 of MEKK3 and valine 384 of MEKK2, residues immediately N-terminal to the active site lysine. Mutation of serine 390 of MEKK3 to a valine (as is found in MEKK2) conferred catalytic activity to MEKK3 in Triton X-100 whereas the reciprocal alteration of valine 384 of MEKK2 to a serine conferred lack of activity in Triton X-100 to MEKK2. Search of the protein database identified only three kinases, MEKK3, Pbs2p and Dd-PKI, with a serine or threonine at this site. The presence of a serine or threonine adjacent to the active site lysine in protein kinases is rare and, in MEKK3, results in detergent instability.


Subject(s)
Detergents , MAP Kinase Kinase Kinases/chemistry , Amino Acid Sequence , Binding Sites , Catalysis , Cell Line , Enzyme Stability , Humans , Lysine/chemistry , MAP Kinase Kinase Kinase 2 , MAP Kinase Kinase Kinase 3 , MAP Kinase Kinase Kinases/biosynthesis , MAP Kinase Kinase Kinases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Octoxynol , Polyethylene Glycols , Serine/chemistry , Transfection , Valine/chemistry
18.
Proc Natl Acad Sci U S A ; 98(8): 4599-604, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11274363

ABSTRACT

Cross-linking of the high-affinity IgE receptor (FcepsilonRI) on mast cells with IgE and multivalent antigen triggers mitogen-activated protein (MAP) kinase activation and cytokine gene expression. We report here that MAP kinase kinase 4 (MKK4) gene disruption does not affect either MAP kinase activation or cytokine gene expression in response to cross-linking of FcepsilonRI in embryonic stem cell-derived mast cells. MKK7 is activated in response to cross-linking of FcepsilonRI, and this activation is inhibited by MAP/ERK kinase (MEK) kinase 2 (MEKK2) gene disruption. In addition, expression of kinase-inactive MKK7 in the murine mast cell line MC/9 inhibits c-Jun NH(2)-terminal kinase (JNK) activation in response to cross-linking of FcepsilonRI, whereas expression of kinase-inactive MKK4 does not affect JNK activation by this stimulus. However, FcepsilonRI-induced activation of the tumor necrosis factor-alpha (TNF-alpha) gene promoter is not affected by expression of kinase-inactive MKK7. We describe an alternative pathway by which MEKK2 activates MEK5 and big MAP kinase1/extracellular signal-regulated kinase 5 in addition to MKK7 and JNK, and interruption of this pathway inhibits TNF-alpha promoter activation. These findings suggest that JNK activation by antigen cross-linking is dependent on the MEKK2-MKK7 pathway, and cytokine production in mast cells is regulated in part by the signaling complex MEKK2-MEK5-ERK5.


Subject(s)
Gene Expression Regulation/physiology , MAP Kinase Kinase Kinases/physiology , Mast Cells/enzymology , Mitogen-Activated Protein Kinase Kinases/physiology , Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/genetics , Cell Line , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 5 , MAP Kinase Kinase 7 , MAP Kinase Kinase Kinase 2 , Promoter Regions, Genetic
19.
J Biol Chem ; 276(3): 2189-99, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11053415

ABSTRACT

The intensity and duration of an inflammatory response depends on the balance of factors that favor perpetuation versus resolution. At sites of inflammation, neutrophils adherent to other cells or matrix components are exposed to tumor necrosis factor-alpha (TNFalpha). Although TNFalpha has been implicated in induction of pro-inflammatory responses, it may also inhibit the intensity of neutrophilic inflammation by promoting apoptosis. Since TNFalpha is not only an important activator of the stress-induced pathways leading to p38 MAPk and c-Jun N-terminal kinase (JNK) but also a potent effector of apoptosis, we investigated the effects of TNFalpha on the JNK pathway in adherent human neutrophils and the potential involvement of this pathway in neutrophil apoptosis. Stimulation with TNFalpha was found to result in beta2 integrin-mediated activation of the cytoplasmic tyrosine kinases Pyk2 and Syk, and activation of a three-part MAPk module composed of MEKK1, MKK7, and/or MKK4 and JNK1. JNK activation was attenuated by blocking antibodies to beta2 integrins, the tyrosine kinase inhibitors, genistein, and tyrphostin A9, a Pyk2-specific inhibitor, and piceatannol, a Syk-specific inhibitor. Exposure of adherent neutrophils to TNFalpha led to the rapid onset of apoptosis that was demonstrated by augmented annexin V binding and caspase-3 cleavage. TNFalpha-induced increases in annexin V binding to neutrophils were attenuated by blocking antibodies to beta2 integrins, and the caspase-3 cleavage was attenuated by tyrphostin A9. Hence, exposure of adherent neutrophils to TNFalpha leads to utilization of the JNK-signaling pathways that may contribute to diverse functional responses including induction of apoptosis and subsequent resolution of the inflammatory response.


Subject(s)
CD18 Antigens/physiology , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/enzymology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Cell Adhesion , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Neutrophils/cytology
20.
J Biol Chem ; 276(7): 5093-100, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11073940

ABSTRACT

MEKK2 and MEKK3 are two closely related mitogen-activated protein kinase (MAPK) kinase kinases. The kinase domains of MEKK2 and MEKK3 are nearly identical, although their N-terminal regulatory domains are significantly divergent. By yeast two-hybrid library screening, we have identified MEK5, the MAPK kinase in the big mitogen-activated protein kinase 1 (BMK1)/ERK5 pathway, as a binding partner for MEKK2. MEKK2 expression stimulates BMK1/ERK5 activity, the downstream substrate for MEK5. Compared with MEKK3, MEKK2 activated BMK1/ERK5 to a greater extent, which might correlate with a higher affinity MEKK2-MEK5 interaction. A dominant negative form of MEK5 blocked the activation of BMK1/ERK5 by MEKK2, whereas activation of c-Jun N-terminal kinase (JNK) was unaffected, showing that MEK5 is a specific downstream effector of MEKK2 in the BMK1/ERK5 pathway. Activation of BMK1/ERK5 by epidermal growth factor and H2O2 in Cos7 and HEK293 cells was completely blocked by a kinase-inactive MEKK3 (MEKK3kin(-)), whereas MEKK2kin(-) had no effect. However, in D10 T cells, expression of MEKK2kin(-) but not MEKK3kin(-) inhibited BMK1/ERK5 activity. Two-hybrid screening also identified Lck-associated adapter/Rlk- and Itk-binding protein (Lad/RIBP), a T cell adapter protein, as a binding partner for MEKK2. MEKK2 and Lad/RIBP colocalize at the T cell contact site with antigen-loaded presenting cells, demonstrating cotranslocation of MEKK2 and Lad/RIBP during T cell activation. MEKK3 neither binds Lad/RIBP nor is recruited to the T cell contact with antigen presenting cell. MEKK2 and MEKK3 are differentially associated with signaling from specific upstream receptor systems, whereas both activate the MEK5-BMK1/ERK5 pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , MAP Kinase Kinase Kinases/physiology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , COS Cells , Cell Line , Epidermal Growth Factor/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Lymphocyte Activation , MAP Kinase Kinase 5 , MAP Kinase Kinase Kinase 2 , MAP Kinase Kinase Kinase 3 , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 7 , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , Oxidants/pharmacology , T-Lymphocytes/drug effects , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...