Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 21(21)2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33114754

ABSTRACT

Extracellular signal-regulated kinase 3 (ERK3), known also as mitogen-activated protein kinase 6 (MAPK6), is an atypical member of MAPK kinase family, which has been poorly studied. Little is known regarding its function in biological processes, yet this atypical kinase has been suggested to play important roles in the migration and invasiveness of certain cancers. The lack of tools, such as a selective inhibitor, hampers the study of ERK3 biology. Here, we report the crystal structure of the kinase domain of this atypical MAPK kinase, providing molecular insights into its distinct ATP binding pocket compared to the classical MAPK ERK2, explaining differences in their inhibitor binding properties. Medium-scale small molecule screening identified a number of inhibitors, several of which unexpectedly exhibited remarkably high inhibitory potencies. The crystal structure of CLK1 in complex with CAF052, one of the most potent inhibitors identified for ERK3, revealed typical type-I binding mode of the inhibitor, which by structural comparison could likely be maintained in ERK3. Together with the presented structural insights, these diverse chemical scaffolds displaying both reversible and irreversible modes of action, will serve as a starting point for the development of selective inhibitors for ERK3, which will be beneficial for elucidating the important functions of this understudied kinase.


Subject(s)
Adenosine Triphosphate/metabolism , Mitogen-Activated Protein Kinase 6/chemistry , Mitogen-Activated Protein Kinase 6/metabolism , Small Molecule Libraries/pharmacology , Binding Sites , Crystallography, X-Ray , Humans , Mitogen-Activated Protein Kinase 6/antagonists & inhibitors , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Small Molecule Libraries/chemistry
2.
Int J Mol Sci ; 21(11)2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32516969

ABSTRACT

Extracellular signal-regulated kinase 3 (ERK3) is an atypical member of the mitogen-activated protein kinase (MAPK) family. It harbors a kinase domain in the N-terminus and a long C-terminus extension. The C-terminus extension comprises a conserved in ERK3 and ERK4 (C34) region and a unique C-terminus tail, which was shown to be required for the interaction of ERK3 with the cytoskeletal protein septin 7. Recent studies have elucidated the role of ERK3 signaling in promoting the motility and invasiveness of cancer cells. However, little is known about the intramolecular regulation of the enzymatic activity and cellular functions of ERK3. In this study, we investigated the role of the elongated C-terminus extension in regulating ERK3 kinase activity and its ability to promote cancer cell migration and invasion. Our study revealed that the deletion of the C-terminus tail greatly diminishes the ability of ERK3 to promote the migration and invasion of lung cancer cells. We identified two molecular mechanisms underlying this effect. Firstly, the deletion of the C-terminus tail decreases the kinase activity of ERK3 towards substrates, including the oncogenic protein steroid receptor co-activator 3 (SRC-3), an important downstream target for ERK3 signaling in cancer. Secondly, in line with the previous finding that the C-terminus tail mediates the interaction of ERK3 with septin 7, we found that the depletion of septin 7 abolished the ability of ERK3 to promote migration, indicating that septin 7 acts as a downstream effector for ERK3-induced cancer cell migration. Taken together, the findings of this study advance our understanding of the molecular regulation of ERK3 signaling by unraveling the role of the C-terminus tail in regulating ERK3 kinase activity and functions in cancer cells. These findings provide useful insights for the development of therapeutic agents targeting ERK3 signaling in cancer.


Subject(s)
Mitogen-Activated Protein Kinase 6/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Protein Interaction Domains and Motifs , Cell Movement/genetics , Enzyme Activation , Humans , Mitogen-Activated Protein Kinase 6/chemistry , Mitogen-Activated Protein Kinase 6/genetics , Neoplasms/pathology , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs/genetics , Signal Transduction
3.
Mol Cell Biol ; 32(13): 2467-78, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22508986

ABSTRACT

Mitogen-activated protein kinase-activated protein (MAPKAP) kinase 5 (MK5) deficiency is associated with reduced extracellular signal-regulated kinase 3 (ERK3) (mitogen-activated protein kinase 6) levels, hence we utilized the MK5 knockout mouse model to analyze the physiological functions of the ERK3/MK5 signaling module. MK5-deficient mice displayed impaired dendritic spine formation in mouse hippocampal neurons in vivo. We performed large-scale interaction screens to understand the neuronal functions of the ERK3/MK5 pathway and identified septin7 (Sept7) as a novel interacting partner of ERK3. ERK3/MK5/Sept7 form a ternary complex, which can phosphorylate the Sept7 regulators Binders of Rho GTPases (Borgs). In addition, the brain-specific nucleotide exchange factor kalirin-7 (Kal7) was identified as an MK5 interaction partner and substrate protein. In transfected primary neurons, Sept7-dependent dendrite development and spine formation are stimulated by the ERK3/MK5 module. Thus, the regulation of neuronal morphogenesis is proposed as the first physiological function of the ERK3/MK5 signaling module.


Subject(s)
Dendrites/metabolism , Dendrites/ultrastructure , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Protein Serine-Threonine Kinases/metabolism , Septins/metabolism , Animals , Base Sequence , DNA Primers/genetics , GTP-Binding Protein Regulators/metabolism , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HeLa Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 6/chemistry , Mitogen-Activated Protein Kinase 6/genetics , Models, Neurological , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Neurogenesis/physiology , Neurons/metabolism , Neurons/ultrastructure , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Septins/chemistry , Septins/genetics , Transfection
4.
J Biol Chem ; 286(15): 13603-11, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21317288

ABSTRACT

The class I p21-activated kinases (Pak1-3) regulate many essential biological processes, including cytoskeletal rearrangement, cell cycle progression, apoptosis, and cellular transformation. Although many Pak substrates, including elements of MAPK signaling cascades, have been identified, it is likely that additional substrates remain to be discovered. Identification of such substrates, and determination of the consequences of their phosphorylation, is essential for a better understanding of class I Pak activity. To identify novel class I Pak substrates, we used recombinant Pak2 to screen high density protein microarrays. This approach identified the atypical MAPK Erk3 as a potential Pak2 substrate. Solution-based in vitro kinase assays using recombinant Erk3 confirmed the protein microarray results, and phospho-specific antisera identified serine 189, within the Erk3 activation loop, as a site directly phosphorylated by Pak2 in vitro. Erk3 protein is known to shuttle between the cytoplasm and the nucleus, and we showed that selective inhibition of class I Pak kinase activity in cells promoted increased nuclear accumulation of Erk3. Pak inhibition in cells additionally reduced the extent of Ser(189) phosphorylation and inhibited the formation of Erk3-Prak complexes. Collectively, our results identify the Erk3 protein as a novel class I Pak substrate and further suggest a role for Pak kinase activity in atypical MAPK signaling.


Subject(s)
Mitogen-Activated Protein Kinase 6/chemistry , p21-Activated Kinases/chemistry , Active Transport, Cell Nucleus/physiology , Animals , Cell Line , Cell Nucleus/enzymology , Cell Nucleus/genetics , Cytoplasm/enzymology , Cytoplasm/genetics , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 6/genetics , Mitogen-Activated Protein Kinase 6/metabolism , Phosphorylation/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera , Substrate Specificity/physiology , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
5.
J Biol Chem ; 284(29): 19392-401, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19473979

ABSTRACT

ERK3 and ERK4 are atypical MAPKs in which the canonical TXY motif within the activation loop of the classical MAPKs is replaced by SEG. Both ERK3 and ERK4 bind, translocate, and activate the MAPK-activated protein kinase (MK) 5. The classical MAPKs ERK1/2 and p38 interact with downstream MKs (RSK1-3 and MK2-3, respectively) through conserved clusters of acidic amino acids, which constitute the common docking (CD) domain. In contrast to the classical MAPKs, the interaction between ERK3/4 and MK5 is strictly dependent on phosphorylation of the SEG motif of these kinases. Here we report that the conserved CD domain is dispensable for the interaction of ERK3 and ERK4 with MK5. Using peptide overlay assays, we have defined a novel MK5 interaction motif (FRIEDE) within both ERK4 and ERK3 that is essential for binding to the C-terminal region of MK5. This motif is located within the L16 extension lying C-terminal to the CD domain in ERK3 and ERK4 and a single isoleucine to lysine substitution in FRIEDE totally abrogates binding, activation, and translocation of MK5 by both ERK3 and ERK4. These findings are the first to demonstrate binding of a physiological substrate via this region of the L16 loop in a MAPK. Furthermore, the link between activation loop phosphorylation and accessibility of the FRIEDE interaction motif suggests a switch mechanism for these atypical MAPKs in which the phosphorylation status of the activation loop regulates the ability of both ERK3 and ERK4 to bind to a downstream effector.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Binding Sites/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoblotting , Intracellular Signaling Peptides and Proteins/genetics , Microscopy, Confocal , Mitogen-Activated Protein Kinase 6/chemistry , Mitogen-Activated Protein Kinase 6/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
6.
Mol Cell Biol ; 23(13): 4542-58, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12808096

ABSTRACT

Mitogen-activated protein (MAP) kinases are stable enzymes that are mainly regulated by phosphorylation and subcellular targeting. Here we report that extracellular signal-regulated kinase 3 (ERK3), unlike other MAP kinases, is an unstable protein that is constitutively degraded in proliferating cells with a half-life of 30 min. The proteolysis of ERK3 is executed by the proteasome and requires ubiquitination of the protein. Contrary to other protein kinases, the catalytic activity of ERK3 is not responsible for its short half-life. Instead, analysis of ERK1/ERK3 chimeras revealed the presence of two destabilization regions (NDR1 and -2) in the N-terminal lobe of the ERK3 kinase domain that are both necessary and sufficient to target ERK3 and heterologous proteins for proteasomal degradation. To assess the physiological relevance of the rapid turnover of ERK3, we monitored the expression of the kinase in different cellular models of differentiation. We observed that ERK3 markedly accumulates during differentiation of PC12 and C2C12 cells into the neuronal and muscle lineage, respectively. The accumulation of ERK3 during myogenic differentiation is associated with the time-dependent stabilization of the protein. Terminal skeletal muscle differentiation is accompanied by cell cycle withdrawal. Interestingly, we found that expression of stabilized forms of ERK3 causes G(1) arrest in NIH 3T3 cells. We propose that ERK3 biological activity is regulated by its cellular abundance through the control of protein stability.


Subject(s)
CDC2-CDC28 Kinases , Cell Differentiation , Cysteine Endopeptidases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 6/metabolism , Ubiquitin/metabolism , 3T3 Cells , Animals , Cyclin-Dependent Kinases/metabolism , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 6/chemistry , Rats , Recombinant Fusion Proteins/metabolism , S Phase , Time Factors , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...