Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(8): 102263, 2022 08.
Article in English | MEDLINE | ID: mdl-35843311

ABSTRACT

Mixed lineage kinase 3 (MLK3) is a serine/threonine mitogen-activated protein kinase kinase kinase that promotes the activation of multiple mitogen-activated protein kinase pathways and is required for invasion and proliferation of ovarian cancer cells. Inhibition of MLK activity causes G2/M arrest in HeLa cells; however, the regulation of MLK3 during ovarian cancer cell cycle progression is not known. Here, we found that MLK3 is phosphorylated in mitosis and that inhibition of cyclin-dependent kinase 1 (CDK1) prevented MLK3 phosphorylation. In addition, we observed that c-Jun N-terminal kinase, a downstream target of MLK3 and a direct target of MKK4 (SEK1), was activated in G2 phase when CDK2 activity is increased and then inactivated at the beginning of mitosis concurrent with the increase in CDK1 and MLK3 phosphorylation. Using in vitro kinase assays and phosphomutants, we determined that CDK1 phosphorylates MLK3 on Ser548 and decreases MLK3 activity during mitosis, whereas CDK2 phosphorylates MLK3 on Ser770 and increases MLK3 activity during G1/S and G2 phases. We also found that MLK3 inhibition causes a reduction in cell proliferation and a cell cycle arrest in ovarian cancer cells, suggesting that MLK3 is required for ovarian cancer cell cycle progression. Taken together, our results suggest that phosphorylation of MLK3 by CDK1 and CDK2 is important for the regulation of MLK3 and c-Jun N-terminal kinase activities during G1/S, G2, and M phases in ovarian cancer cell division.


Subject(s)
CDC2 Protein Kinase , Cyclin-Dependent Kinase 2 , Ovarian Neoplasms , CDC2 Protein Kinase/metabolism , Cell Division/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 2/metabolism , Female , G2 Phase Cell Cycle Checkpoints , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mitosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Phosphorylation , Mitogen-Activated Protein Kinase Kinase Kinase 11
2.
Mol Biol Cell ; 32(16): 1446-1458, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34106735

ABSTRACT

G protein beta-gamma (Gßγ) subunits anchor to the plasma membrane (PM) through the carboxy-terminal (CT) prenyl group in Gγ. This interaction is crucial for the PM localization and functioning of Gßγ, allowing GPCR-G protein signaling to proceed. The diverse Gγ family has 12 members, and we have recently shown that the signaling efficacies of major Gßγ effectors are Gγ-type dependent. This dependency is due to the distinct series of membrane-interacting abilities of Gγ. However, the molecular process allowing for Gßγ subunits to exhibit a discrete and diverse range of Gγ-type-dependent membrane affinities is unclear and cannot be explained using only the type of prenylation. The present work explores the unique designs of membrane-interacting CT residues in Gγ as a major source for this Gγ-type-dependent Gßγ signaling. Despite the type of prenylation, the results show signaling efficacy at the PM, and associated cell behaviors of Gßγ are governed by crucially located specific amino acids in the five to six residue preprenylation region of Gγ. The provided molecular picture of Gγ-membrane interactions may explain how cells gain Gγ-type-dependent G protein-GPCR signaling as well as how Gßγ elicits selective signaling at various subcellular compartments.


Subject(s)
Cell Membrane/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Protein Prenylation , Signal Transduction , GTP-Binding Protein beta Subunits/metabolism , HeLa Cells , Humans
3.
Mol Cell Biol ; 41(7): e0007821, 2021 06 23.
Article in English | MEDLINE | ID: mdl-33875576

ABSTRACT

Mixed-lineage kinase 3 (MLK3) activates mammalian mitogen-activated protein kinase (MAPK) signaling pathways in response to cytokines and stress stimuli. MLK3 is important for proliferation, migration, and invasion of different types of human tumor cells. We observed that endogenous MLK3 was localized to both the cytoplasm and the nucleus in immortalized ovarian epithelial (T80) and ovarian cancer cells, and mutation of arginines 474 and 475 within a putative MLK3 nuclear localization sequence (NLS) resulted in exclusion of MLK3 from the nucleus. The large tumor suppressor (LATS) Ser/Thr kinase regulates cell proliferation, morphology, apoptosis, and mitotic exit in response to cell-cell contact. RNA interference (RNAi)-mediated knockdown of LATS1 increased nuclear, endogenous MLK3 in T80 cells. LATS1 phosphorylated MLK3 on Thr477, which is within the putative NLS, and LATS1 expression enhanced the association between MLK3 and the adapter protein 14-3-3ζ. Thr477 is essential for MLK3-14-3-3ζ association and MLK3 retention in the cytoplasm, and a T477A MLK3 mutant had predominantly nuclear localization and significantly increased invasiveness of SKOV3 ovarian cancer cells. This study identified a novel link between the MAPK and Hippo/LATS1 signaling pathways. Our results reveal LATS1 as a novel regulator of MLK3 that controls MLK3 nuclear/cytoplasmic localization and MLK3-dependent ovarian cancer cell invasion.


Subject(s)
Epithelial Cells/metabolism , MAP Kinase Kinase Kinases/metabolism , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Cell Movement/genetics , Female , Humans , MAP Kinase Signaling System/physiology , Ovarian Neoplasms/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , Mitogen-Activated Protein Kinase Kinase Kinase 11
4.
Exp Cell Res ; 394(1): 112167, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32649943

ABSTRACT

Nicotine is the major addictive component of cigarette smoke and although it is not considered carcinogenic, it can enhance or inhibit cancer cell proliferation depending on the type of cancer. Nicotine mediates its effects through nicotinic acetylcholine receptors (nAChRs), which are expressed in many different neuronal and non-neuronal cell types. We observed that the nAChR α4, α5, α7 subunits were expressed in ovarian cancer (OC) cells. Nicotine inhibited the proliferation of SKOV3 and TOV112D OC cells, which have TP53 mutation and wild-type KRAS, but did not inhibit the proliferation of TOV21G or HEY OC cells, which have KRAS mutation and wild-type TP53. Exposure to nicotine for 96 h led to a significant reduction in the amounts of activated extracellular signal-regulated kinase (ERK) and activated p38 mitogen-activated protein kinases (MAPKs) in SKOV3 cells, and in activated ERK in TOV112D cells. In addition, SKOV3 and TOV112D invasion and spheroid formation were substantially inhibited by siRNA knockdown of mixed lineage kinase 3 (MLK3), or MEK inhibition. Nicotine treatment reduced SKOV3 and TOV112D spheroid invasion and compaction but did not significantly affect spheroid formation. Furthermore, SKOV3 spheroid invasion was blocked by p38 inhibition with SB202190, but not by MEK inhibition with U0126; whereas TOV112D spheroid invasion was reduced by MEK inhibition, but not by p38 inhibition. These results indicate that nicotine can suppress spheroid invasion and compaction as well as proliferation in SKOV3 and TOV112D OC cells; and p38 and ERK MAPK signaling pathways are important mediators of these responses.


Subject(s)
Nicotine/pharmacology , Ovarian Neoplasms/drug therapy , Receptors, Nicotinic/drug effects , Signal Transduction/drug effects , Carcinoma, Ovarian Epithelial/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Imidazoles/pharmacology , Ovarian Neoplasms/metabolism , Phosphorylation/drug effects , Pyridines/pharmacology , Receptors, Nicotinic/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Cell Signal ; 58: 34-43, 2019 06.
Article in English | MEDLINE | ID: mdl-30849518

ABSTRACT

G protein αq-coupled receptors (Gq-GPCRs) primarily signal through GαqGTP mediated phospholipase Cß (PLCß) stimulation and the subsequent hydrolysis of phosphatidylinositol 4, 5 bisphosphate (PIP2). Though Gq-heterotrimer activation results in both GαqGTP and Gßγ, unlike Gi/o-receptors, it is unclear if Gq-coupled receptors employ Gßγ as a major signal transducer. Compared to Gi/o- and Gs-coupled receptors, we observed that most cell types exhibit a limited free Gßγ generation upon Gq-pathway and Gαq/11 heterotrimer activation. We show that cells transfected with Gαq or endogenously expressing more than average-levels of Gαq/11 compared to Gαs and Gαi exhibit a distinct signaling regime primarily characterized by recovery-resistant PIP2 hydrolysis. Interestingly, the elevated Gq-expression is also associated with enhanced free Gßγ generation and signaling. Furthermore, the gene GNAQ, which encodes for Gαq, has recently been identified as a cancer driver gene. We also show that GNAQ is overexpressed in tumor samples of patients with Kidney Chromophobe (KICH) and Kidney renal papillary (KIRP) cell carcinomas in a matched tumor-normal sample analysis, which demonstrates the clinical significance of Gαq expression. Overall, our data indicates that cells usually express low Gαq levels, likely safeguarding cells from excessive calcium as wells as from Gßγ signaling.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Signal Transduction , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Expression , HeLa Cells , Humans , Hydrolysis , Phospholipase C beta/metabolism , Transfection
6.
Cell Signal ; 39: 66-73, 2017 11.
Article in English | MEDLINE | ID: mdl-28757353

ABSTRACT

Mixed Lineage Kinase 3 (MLK3), a member of the MLK subfamily of protein kinases, is a mitogen-activated protein (MAP) kinase kinase kinase (MAP3K) that activates MAPK signalling pathways and regulates cellular responses such as proliferation, invasion and apoptosis. MLK4ß, another member of the MLK subfamily, is less extensively studied, and the regulation of MLK4ß by stress stimuli is not known. In this study, the regulation of MLK4ß and MLK3 by osmotic stress, thermostress and heat shock protein 90 (Hsp90) inhibition was investigated in ovarian cancer cells. MLK3 and MLK4ß protein levels declined under conditions of prolonged osmotic stress, heat stress or exposure to the Hsp90 inhibitor geldanamycin (GA); and MLK3 protein declined faster than MLK4ß. Similar to MLK3, the reduction in MLK4ß protein in cells exposed to heat or osmotic stresses occurred via a mechanism that involves the E3 ligase, carboxy-terminus of Hsc70-interacting protein (CHIP). Both heat shock protein 70 (Hsp70) and CHIP overexpression led to polyubiquitination and a decrease in endogenous MLK4ß protein, and MLK4ß was ubiquitinated by CHIP in vitro. In untreated cells and cells exposed to osmotic and heat stresses for short time periods, small interfering RNA (siRNA) knockdown of MLK4ß elevated the levels of activated MLK3, c-Jun N-terminal kinase (JNK) and p38 MAPKs. Furthermore, MLK3 binds to MLK4ß, and this association is regulated by osmotic stress. These results suggest that in the early response to stressful stimuli, MLK4ß-MLK3 binding is important for regulating MLK3 activity and MAPK signalling, and after prolonged periods of stress exposure, MLK4ß and MLK3 proteins decline via CHIP-dependent degradation. These findings provide insight into how heat and osmotic stresses regulate MLK4ß and MLK3, and reveal an important function for MLK4ß in modulating MLK3 activity in stress responses.


Subject(s)
Heat-Shock Response , MAP Kinase Kinase Kinases/metabolism , Osmotic Pressure , Ovarian Neoplasms/enzymology , Ubiquitin-Protein Ligases/metabolism , Benzoquinones/pharmacology , Cell Line, Tumor , Female , HEK293 Cells , HSC70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/pharmacology , MAP Kinase Kinase Kinases/genetics , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Mitogen-Activated Protein Kinase Kinase Kinase 11
7.
Exp Cell Res ; 331(2): 292-308, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25281303

ABSTRACT

We previously demonstrated that the expression of intercellular adhesion molecule-1 (ICAM-1) by skeletal muscle cells after muscle overload contributes to ensuing regenerative and hypertrophic processes in skeletal muscle. The objective of the present study is to reveal mechanisms through which skeletal muscle cell expression of ICAM-1 augments regenerative and hypertrophic processes of myogenesis. This was accomplished by genetically engineering C2C12 myoblasts to stably express ICAM-1, and by inhibiting the adhesive and signaling functions of ICAM-1 through the use of a neutralizing antibody or cell penetrating peptide, respectively. Expression of ICAM-1 by cultured skeletal muscle cells augmented myoblast-myoblast adhesion, myotube formation, myonuclear number, myotube alignment, myotube-myotube fusion, and myotube size without influencing the ability of myoblasts to proliferate or differentiate. ICAM-1 augmented myotube formation, myonuclear accretion, and myotube alignment through a mechanism involving adhesion-induced activation of ICAM-1 signaling, as these dependent measures were reduced via antibody and peptide inhibition of ICAM-1. The adhesive and signaling functions of ICAM-1 also facilitated myotube hypertrophy through a mechanism involving myotube-myotube fusion, protein synthesis, and Akt/p70s6k signaling. Our findings demonstrate that ICAM-1 expression by skeletal muscle cells augments myogenesis, and establish a novel mechanism through which the inflammatory response facilitates growth processes in skeletal muscle.


Subject(s)
Intercellular Adhesion Molecule-1/biosynthesis , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Animals , CD11a Antigen/biosynthesis , CD11b Antigen/biosynthesis , Cell Adhesion/physiology , Cell Differentiation , Cell Fusion , Cell Line , Cell Proliferation , Intercellular Adhesion Molecule-1/genetics , Mice , Muscle Fibers, Skeletal/metabolism , Myoblasts/physiology , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Mol Cell Biol ; 34(16): 3132-43, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24912674

ABSTRACT

Mixed-lineage kinase 3 (MLK3) activates mitogen-activated protein kinase (MAPK) signaling pathways and has important functions in migration, invasion, proliferation, tumorigenesis, and apoptosis. We investigated the role of the E3 ligase carboxyl terminus of Hsc70-interacting protein (CHIP) in the regulation of MLK3 protein levels. We show that CHIP interacts with MLK3 and, together with the E2 ubiquitin-conjugating enzyme UbcH5 (UbcH5a, -b, -c, or -d), ubiquitinates MLK3 in vitro. CHIP or Hsp70 overexpression promoted endogenous MLK3 ubiquitination and induced a decline in MLK3 protein levels in cells with Hsp90 inhibition. Furthermore, CHIP overexpression caused a proteasome-dependent reduction in exogenous MLK3 protein. Geldanamycin (GA), heat shock, and osmotic shock treatments also reduced the level of MLK3 protein via a CHIP-dependent mechanism. In addition, CHIP depletion in ovarian cancer SKOV3 cells increased cell invasion, and the enhancement of invasiveness was abrogated by small interfering RNA (siRNA)-mediated knockdown of MLK3. Thus, CHIP modulates MLK3 protein levels in response to GA and stress stimuli, and CHIP-dependent regulation of MLK3 is required for suppression of SKOV3 ovarian cancer cell invasion.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Ubiquitin-Protein Ligases/genetics , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Movement/genetics , Enzyme Inhibitors/pharmacology , HEK293 Cells , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/biosynthesis , HSP90 Heat-Shock Proteins/biosynthesis , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Response , Humans , Lactams, Macrocyclic/pharmacology , MAP Kinase Kinase Kinases/biosynthesis , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System , Neoplasm Invasiveness , Osmotic Pressure , Protein Binding , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Sorbitol/pharmacology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitination , Mitogen-Activated Protein Kinase Kinase Kinase 11
9.
Can J Physiol Pharmacol ; 91(4): 268-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23627838

ABSTRACT

Mitogen-activated protein kinase (MAPK) signaling pathways are composed of a phosphorelay signaling module where an activated MAP kinase kinase kinase (MAP3K) phosphorylates and activates a MAPK kinase (MAP2K) that in turn phosphorylates and activates a MAPK. The biological outcome of MAPK signaling is the regulation of cellular responses such as proliferation, differentiation, migration, and apoptosis. The MAP3K mixed lineage kinase 3 (MLK3) phosphorylates MAP2Ks to activate multiple MAPK signaling pathways, and MLK3 also has functions in cell signaling that are independent of its kinase activity. The recent elucidation of essential functions for MLK3 in tumour cell proliferation, migration, and invasion has drawn attention to the MLKs as potential therapeutic targets for cancer treatments. The mounting evidence that suggests a role for MLK3 in tumourigenesis and establishment of the malignant phenotype is the focus of this review.


Subject(s)
Cell Transformation, Neoplastic/genetics , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Neoplasms/pathology , Signal Transduction , Mitogen-Activated Protein Kinase Kinase Kinase 11
10.
Exp Cell Res ; 318(14): 1641-8, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22652451

ABSTRACT

Mixed lineage kinase 3 (MLK3) is a mitogen-activated protein kinase kinase kinase (MAP3K) that activates MAPK signaling pathways and regulates cellular responses such as proliferation, migration and apoptosis. Here we report high levels of total and phospho-MLK3 in ovarian cancer cell lines in comparison to immortalized nontumorigenic ovarian epithelial cell lines. Using small interfering RNA (siRNA)-mediated gene silencing, we determined that MLK3 is required for the invasion of SKOV3 and HEY1B ovarian cancer cells. Furthermore, mlk3 silencing substantially reduced matrix metalloproteinase (MMP)-1, -2, -9 and -12 gene expression and MMP-2 and -9 activities in SKOV3 and HEY1B ovarian cancer cells. MMP-1, -2, -9 and-12 expression, and MLK3-induced activation of MMP-2 and MMP-9 requires both extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) activities. In addition, inhibition of activator protein-1 (AP-1) reduced MMP-1, MMP-9 and MMP-12 gene expression. Collectively, these findings establish MLK3 as an important regulator of MMP expression and invasion in ovarian cancer cells.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Matrix Metalloproteinases/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Female , Gene Expression Profiling , Humans , MAP Kinase Kinase Kinases/genetics , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Ovarian Neoplasms/metabolism , Phosphorylation , Real-Time Polymerase Chain Reaction , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , Mitogen-Activated Protein Kinase Kinase Kinase 11
11.
Methods Mol Biol ; 661: 59-73, 2010.
Article in English | MEDLINE | ID: mdl-20811976

ABSTRACT

The stress-activated protein kinase/c-jun N-terminal kinases (SAPK/JNKs) are mitogen-activated protein kinases (MAPKs) that are activated by stressful and inflammatory stimuli and regulate cellular responses such as proliferation, differentiation, and apoptosis. The SAPK/JNKs are phosphorylated and activated by the MAP kinase kinases (MAP2Ks), SEK1/MKK4 and MKK7. These MAP2Ks are phosphorylated and activated by upstream stress-activated MAPK kinase kinases (MAP3Ks). Upon activation, SAPK/JNKs translocate to the nucleus and phosphorylate transcription factors, ultimately resulting in the modulation of gene expression. We have analyzed the activation of SAPK/JNK and stress-activated MAP3Ks using in vitro kinase assays. In addition, we have studied the role of different MAP3Ks in SAPK/JNK signaling by silencing specific MAP3K expression with RNAi and then analyzing the effect on activation of SAPK/JNKs and other MAPKs.


Subject(s)
Enzyme Assays/methods , JNK Mitogen-Activated Protein Kinases/metabolism , Animals , Antibodies/immunology , Antibody Specificity , Enzyme Activation , Gene Knockdown Techniques , Humans , JNK Mitogen-Activated Protein Kinases/immunology , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Phosphoproteins/immunology , Phosphorylation , RNA, Small Interfering/genetics
12.
Small GTPases ; 1(3): 183-186, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21686274

ABSTRACT

Mammalian mitogen-activated protein kinase (MAPK) signaling pathways respond to diverse extracellular signals and coordinate a range of cellular responses. Mixed lineage kinase 3 (MLK3) is a member of the mixed lineage kinase family of MAPK kinase kinases (MAP3Ks) that functions to regulate multiple MAPK signaling pathways. Activated forms of the Rho GTP ases, Rac and Cdc42, interact with MLK3 through the Cdc42/Rac-interactive binding (CRIB) motif and promote MLK3 catalytic activity. Our recent findings demonstrate that merlin, the product of the neurofibromatosis type 2 (NF2) tumor suppressor gene, is a physiological inhibitor of MLK3. Our results suggest that merlin inhibits MLK3 activity by blocking the Cdc42-MLK3 interaction. In this commentary, the effect of merlin on Cdc42-mediated activation of MLK3 and MAPK signaling will be discussed.

13.
Biochim Biophys Acta ; 1793(12): 1811-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19782705

ABSTRACT

Mixed lineage kinase 3 (MLK3) is a mitogen activated protein kinase kinase kinase (MAP3K) that activates multiple MAPK signaling pathways. Nuclear factor kappa B (NF-kappaB) is a transcription factor that has important functions in inflammation, immunity and cell survival. We found that silencing mlk3 expression with RNA interference (RNAi) in SKOV3 human ovarian cancer epithelial cells and NIH-3T3 murine fibroblasts led to a reduction in the level of the inhibitor of kappa B alpha (IkappaBalpha) protein. In addition, we observed enhanced basal IkappaB kinase (IKK) activity in HEK293 cells transiently transfected with MLK3 siRNA and in NIH3T3 cells stably expressing MLK3 shRNA (shMLK3). Furthermore, the basal level of NF-kappaB-dependent gene transcription was elevated in shMLK3 cells. Silencing mlk3 expression conferred resistance of cells to etoposide-induced apoptotic cell death and overexpression of wild type MLK3 (MLK3-WT) or kinase-dead MLK3 (MLK3-KD) promoted apoptotic cell death and cleavage of poly (ADP-ribose) polymerase (PARP). Overexpression of MLK3-WT or MLK3-KD enhanced etoposide-induced apoptotic cell death and cleavage of PARP. These data suggest that MLK3 functions to limit IKK activity, and depleting MLK3 helps protect cells from etoposide-induced cell death through activation of IKK-dependent signaling.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Etoposide/pharmacology , I-kappa B Kinase/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Animals , Apoptosis/genetics , Cell Line, Tumor , Humans , I-kappa B Kinase/genetics , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System/genetics , Mice , NIH 3T3 Cells , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Mitogen-Activated Protein Kinase Kinase Kinase 11
14.
Cell Signal ; 21(11): 1620-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19586614

ABSTRACT

Mixed lineage kinase 3 (MLK3) is a mitogen-activated protein kinase kinase kinase (MAP3K) that activates multiple mitogen-activated protein kinase (MAPK) pathways in response to growth factors, stresses and the pro-inflammatory cytokine, tumor necrosis factor (TNF). MLK3 is required for optimal activation of stress activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) signaling by TNF, however, the mechanism by which MLK3 is recruited and activated by the TNF receptor remains poorly understood. Here we report that both TNF and interleukin-1 beta (IL-1 beta) stimulation rapidly activate MLK3 kinase activity. We observed that TNF stimulates an interaction between MLK3 and TNF receptor associated factor (TRAF) 2 and IL-1 beta stimulates an interaction between MLK3 and TRAF6. RNA interference (RNAi) of traf2 or traf6 dramatically impairs MLK3 activation by TNF indicating that TRAF2 and TRAF6 are critically required for MLK3 activation. We show that TNF also stimulates ubiquitination of MLK3 and MLK3 can be conjugated with lysine 48 (K48)- and lysine 63 (K63)-linked polyubiquitin chains. Our results suggest that K48-linked ubiquitination directs MLK3 for proteosomal degradation while K63-linked ubiquitination is important for MLK3 kinase activity. These results reveal a novel mechanism for MLK3 activation by the pro-inflammatory cytokines TNF and IL-1 beta.


Subject(s)
Interleukin-1beta/pharmacology , MAP Kinase Kinase Kinases/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Line , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 6/genetics , Ubiquitination , Mitogen-Activated Protein Kinase Kinase Kinase 11
15.
J Cell Biochem ; 106(1): 33-41, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19009561

ABSTRACT

Activating ras mutations are frequently found in malignant tumors of the pancreas, colon, lung and other tissues. RAS activates a number of downstream pathways that ultimately cause cellular transformation. Several recent studies suggested that one of those pathways involves Aurora kinases. Overexpression of Aurora-B kinase can augment transformation by oncogenic RAS, however the mechanism was not determined. The cooperative effect of high levels of Aurora kinase is important since this kinase is frequently overexpressed in human tumors. We have used two Aurora kinase inhibitors to test their effect on RAS signaling. We find that these inhibitors have no effect on the phosphorylation of MEK1/2 or MAPK in response to RAS. Furthermore, inhibiting Aurora kinases in human cancer cells with or without activated RAS did not change the length of the cell cycle nor induce apoptosis suggesting that these kinases do not play a direct role in these key cellular responses to activated RAS. Overexpression of Aurora B can cause cells to become polyploid. Also, inducing polyploidy with cytochalasin D was reported to induce neoplastic transformation, suggesting that Aurora overexpression may cooperate with RAS indirectly by inducing polyploidy. We find that inducing polyploidy with cytochalasin D or blebbistatin does not enhance transformation by oncogenic RAS. Our observations argue against a direct role for Aurora kinases in the RAS-MAPK pathway, and suggest that the polyploid state does not enhance transformation by RAS.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Aurora Kinase B , Aurora Kinases , Benzamides/pharmacology , Cell Cycle , Cell Line, Tumor , Fibroblasts/metabolism , Histones/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , NIH 3T3 Cells , Phosphorylation , Polyploidy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Rats , raf Kinases/metabolism
16.
Proc Natl Acad Sci U S A ; 103(12): 4463-8, 2006 Mar 21.
Article in English | MEDLINE | ID: mdl-16537381

ABSTRACT

The Ras --> Raf --> MEK1/2 --> extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) pathway couples mitogenic signals to cell proliferation. B-Raf and Raf-1 function within an oligomer wherein they are regulated in part by mutual transactivation. The MAPK kinase kinase (MAP3K) mixed-lineage kinase 3 (MLK3) is required for mitogen activation of B-Raf and cell proliferation. Here we show that the kinase activity of MLK3 is not required for support of B-Raf activation. Instead, MLK3 is a component of the B-Raf/Raf-1 complex and is required for maintenance of the integrity of this complex. We show that the activation of ERK and the proliferation of human schwannoma cells bearing a loss-of-function mutation in the neurofibromatosis 2 (NF2) gene require MLK3. We find that merlin, the product of NF2, blunts the activation of both ERK and c-Jun N-terminal kinase (JNK). Finally, we demonstrate that merlin and MLK3 can interact in situ and that merlin can disrupt the interactions between B-Raf and Raf-1 or those between MLK3 and either B-Raf or Raf-1. Thus, MLK3 is part of a multiprotein complex and is required for ERK activation. The levels of this complex may be negatively regulated by merlin.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Neurofibromin 2/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Multienzyme Complexes/metabolism , Mutation , Neurofibromin 2/genetics , Rats , Tumor Suppressor Proteins/genetics , Mitogen-Activated Protein Kinase Kinase Kinase 11
17.
Cell Cycle ; 3(10): 1227-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15467451

ABSTRACT

The extracellular signal-regulated kinase (ERK) group of MAPKs is essential for cell proliferation, including that stimulated by mitogens, oncogenic ras and raf. The Raf kinases (especially B-Raf) are ERK-specific, mitogen-activated MAP3Ks. Mixed lineage kinase-3 (MLK3) is a MAP3K previously thought to be a selective regulator of the JNK group of MAPKs. Surprisingly, we found that silencing of mlk3 by RNAi suppresses mitogen and cytokine activation not only of JNK but of ERK and p38 as well. Silencing mlk3 also blocks mitogen-stimulated phosphorylation of B-Raf at Thr598 and Ser601-a step required for B-Raf activation. Finally, silencing mlk3 prevents serum-stimulated cell proliferation and the proliferation of tumor cells bearing either oncogenic Ki-Ras or loss of function neurofibromatosis-1 (NF1) or NF2 mutations. The proliferation of tumor cells with activating mutations in B-raf or raf-1 are unaffected by silencing mlk3. These results define a new role for MLK3 in B-Raf activation, ERK signaling and cell proliferation. Accordingly, targeting MLK3 could be beneficial to the treatment of tumors with activated receptor tyrosine kinase or ras mutations, and to the treatment of NF1 or NF2 tumors.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Animals , Cell Proliferation , Drosophila , Humans , Signal Transduction , Mitogen-Activated Protein Kinase Kinase Kinase 11
18.
Nat Cell Biol ; 6(8): 770-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258589

ABSTRACT

The ERK group of mitogen-activated protein kinases (MAPKs) is essential for cell proliferation stimulated by mitogens, oncogenic ras and raf (ref. 1). All MAPKs are activated by MAP3K/MEK/MAPK core pathways and the Raf proto-oncoproteins, especially B-Raf, are ERK-specific MAP3Ks (refs 1-3). Mixed lineage kinase-3 (MLK3) is a MAP3K that was thought to be a cytokine-activated, and comparatively selective, regulator of the JNK group of MAPKs (refs 1, 4-6). Here we report that silencing of mlk3 by RNAi suppressed mitogen and cytokine activation not only of JNK but of ERK and p38 as well. Silencing mlk3 also blocked mitogen-stimulated phosphorylation of B-Raf at Thr 598 and Ser 601, a step required for B-Raf activation. Furthermore, silencing mlk3 prevented serum-stimulated cell proliferation and the proliferation of tumour cells bearing either oncogenic Ki-Ras or loss-of-function neurofibromatosis-1 (NF1) or NF2 mutations. The proliferation of tumour cells containing activating B-raf or raf-1 mutations was unaffected by silencing mlk3. Our results define an unexpected role for MLK3 in mitogen regulation of B-Raf, ERK and cell proliferation.


Subject(s)
Cell Division , MAP Kinase Kinase Kinases/physiology , Mitogen-Activated Protein Kinases/physiology , Mitogens/pharmacology , Proto-Oncogene Proteins c-raf/physiology , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme Activation , Gene Silencing , HT29 Cells , Humans , Jurkat Cells , MAP Kinase Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/drug effects , Proto-Oncogene Proteins c-raf/metabolism , RNA Interference , Serine/chemistry , Threonine/chemistry , Umbilical Veins/cytology , Mitogen-Activated Protein Kinase Kinase Kinase 11
20.
Am J Physiol Cell Physiol ; 285(4): C823-30, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12801888

ABSTRACT

Smad4, the common Smad, is central for transforming growth factor (TGF)-beta superfamily ligand signaling. Smad4 has been shown to be constitutively phosphorylated (Nakao A, Imamura T, Souchelnytskyi S, Kawabata M, Ishisaki A, Oeda E, Tamaki K, Hanai J, Heldin C-H, Miyazono K, and ten Dijke P. EMBO J 16: 5353-5362, 1997), but the site(s) of phosphorylation, the kinase(s) that performs this phosphorylation, and the significance of the phosphorylation of Smad4 are currently unknown. This report describes the identification of a consensus ERK phosphorylation site in the linker region of Smad4 at Thr276. Our data show that ERK can phosphorylate Smad4 in vitro but not Smad4 with mutated Thr276. Flag-tagged Smad4-T276A mutant protein accumulates less efficiently in the nucleus after stimulation by TGF-beta and is less efficient in generating a transcriptional response than Smad4 wild-type protein. Tryptic phosphopeptide mapping identified a phosphopeptide in Smad4 wild-type protein that was absent in phosphorylated Smad4-T276A mutant protein. Our results suggest that MAP kinase can phosphorylate Thr276 of Smad4 and that phosphorylation can lead to enhanced TGF-beta-induced nuclear accumulation and, as a consequence, enhanced transcriptional activity of Smad4.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transforming Growth Factor beta/pharmacology , Amino Acid Sequence/genetics , Animals , Cell Line , Consensus Sequence , Genetic Linkage , LLC-PK1 Cells , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Mutation , Phosphoproteins/metabolism , Phosphorylation , Protein Structure, Tertiary/genetics , Swine , Threonine/metabolism , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...