Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Commun Biol ; 4(1): 645, 2021 05 31.
Article in English | MEDLINE | ID: mdl-34059773

ABSTRACT

WDR62 is a scaffold protein involved in centriole duplication and spindle assembly during mitosis. Mutations in WDR62 can cause primary microcephaly and premature ovarian insufficiency. We have generated a genetrap mouse model deficient in WDR62 and characterised the developmental effects of WDR62 deficiency during meiosis in the testis. We have found that WDR62 deficiency leads to centriole underduplication in the spermatocytes due to reduced or delayed CEP63 accumulation in the pericentriolar matrix. This resulted in prolonged metaphase that led to apoptosis. Round spermatids that inherited a pair of centrioles progressed through spermiogenesis, however, manchette removal was delayed in WDR62 deficient spermatids due to delayed Katanin p80 accumulation in the manchette, thus producing misshapen spermatid heads with elongated manchettes. In mice, WDR62 deficiency resembles oligoasthenoteratospermia, a common form of subfertility in men that is characterised by low sperm counts, poor motility and abnormal morphology. Therefore, proper WDR62 function is necessary for timely spermatogenesis and spermiogenesis during male reproduction.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/genetics , Nerve Tissue Proteins/metabolism , Spermatogenesis/genetics , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Centrioles/metabolism , Cytoskeleton/metabolism , Female , Male , Meiosis , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Spermatids/metabolism , Spermatogenesis/physiology , Spermatozoa/metabolism , Testis/metabolism
2.
Hum Mol Genet ; 29(2): 248-263, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31816041

ABSTRACT

WDR62 mutations that result in protein loss, truncation or single amino-acid substitutions are causative for human microcephaly, indicating critical roles in cell expansion required for brain development. WDR62 missense mutations that retain protein expression represent partial loss-of-function mutants that may therefore provide specific insights into radial glial cell processes critical for brain growth. Here we utilized CRISPR/Cas9 approaches to generate three strains of WDR62 mutant mice; WDR62 V66M/V66M and WDR62R439H/R439H mice recapitulate conserved missense mutations found in humans with microcephaly, with the third strain being a null allele (WDR62stop/stop). Each of these mutations resulted in embryonic lethality to varying degrees and gross morphological defects consistent with ciliopathies (dwarfism, anophthalmia and microcephaly). We find that WDR62 mutant proteins (V66M and R439H) localize to the basal body but fail to recruit CPAP. As a consequence, we observe deficient recruitment of IFT88, a protein that is required for cilia formation. This underpins the maintenance of radial glia as WDR62 mutations caused premature differentiation of radial glia resulting in reduced generation of neurons and cortical thinning. These findings highlight the important role of the primary cilium in neocortical expansion and implicate ciliary dysfunction as underlying the pathology of MCPH2 patients.


Subject(s)
Cell Cycle Proteins/metabolism , Cilia/metabolism , Ciliopathies/genetics , Microcephaly/genetics , Microtubule-Associated Proteins/metabolism , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Anophthalmos/embryology , Anophthalmos/genetics , Anophthalmos/metabolism , Apoptosis/genetics , CRISPR-Cas Systems , Cell Cycle Proteins/genetics , Cells, Cultured , Cilia/genetics , Cilia/pathology , Ciliopathies/embryology , Ciliopathies/metabolism , Ciliopathies/pathology , Dwarfism/embryology , Dwarfism/genetics , Dwarfism/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcephaly/embryology , Microcephaly/metabolism , Microtubule-Associated Proteins/genetics , Mutation, Missense , Neocortex/embryology , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Neuroglia/cytology , Neuroglia/metabolism , Neurons/metabolism , Tumor Suppressor Proteins/genetics
3.
PLoS Biol ; 16(12): e2006613, 2018 12.
Article in English | MEDLINE | ID: mdl-30566428

ABSTRACT

Mutations of WD repeat domain 62 (WDR62) lead to autosomal recessive primary microcephaly (MCPH), and down-regulation of WDR62 expression causes the loss of neural progenitor cells (NPCs). However, how WDR62 is regulated and hence controls neurogenesis and brain size remains elusive. Here, we demonstrate that mitogen-activated protein kinase kinase kinase 3 (MEKK3) forms a complex with WDR62 to promote c-Jun N-terminal kinase (JNK) signaling synergistically in the control of neurogenesis. The deletion of Mekk3, Wdr62, or Jnk1 resulted in phenocopied defects, including premature NPC differentiation. We further showed that WDR62 protein is positively regulated by MEKK3 and JNK1 in the developing brain and that the defects of wdr62 deficiency can be rescued by the transgenic expression of JNK1. Meanwhile, WDR62 is also negatively regulated by T1053 phosphorylation, leading to the recruitment of F-box and WD repeat domain-containing protein 7 (FBW7) and proteasomal degradation. Our findings demonstrate that the coordinated reciprocal and bidirectional regulation among MEKK3, FBW7, WDR62, and JNK1, is required for fine-tuned JNK signaling for the control of balanced NPC self-renewal and differentiation during cortical development.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7/physiology , MAP Kinase Kinase Kinase 3/physiology , Microtubule-Associated Proteins/metabolism , Animals , Cell Differentiation , F-Box-WD Repeat-Containing Protein 7/genetics , Female , HEK293 Cells , Humans , MAP Kinase Kinase Kinase 3/genetics , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Mice, Transgenic , Microcephaly/genetics , Microcephaly/physiopathology , Mitogen-Activated Protein Kinase 8/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Phosphorylation , Protein Binding , Rats , Rats, Sprague-Dawley , Signal Transduction
4.
Cell Cycle ; 15(3): 413-24, 2016.
Article in English | MEDLINE | ID: mdl-26713495

ABSTRACT

Mitotic spindle organization is regulated by centrosomal kinases that potentiate recruitment of spindle-associated proteins required for normal mitotic progress including the microcephaly protein WD40-repeat protein 62 (WDR62). WDR62 functions underlie normal brain development as autosomal recessive mutations and wdr62 loss cause microcephaly. Here we investigate the signaling interactions between WDR62 and the mitotic kinase Aurora A (AURKA) that has been recently shown to cooperate to control brain size in mice. The spindle recruitment of WDR62 is closely correlated with increased levels of AURKA following mitotic entry. We showed that depletion of TPX2 attenuated WDR62 localization at spindle poles indicating that TPX2 co-activation of AURKA is required to recruit WDR62 to the spindle. We demonstrated that AURKA activity contributed to the mitotic phosphorylation of WDR62 residues Ser49 and Thr50 and phosphorylation of WDR62 N-terminal residues was required for spindle organization and metaphase chromosome alignment. Our analysis of several MCPH-associated WDR62 mutants (V65M, R438H and V1314RfsX18) that are mislocalized in mitosis revealed that their interactions and phosphorylation by AURKA was substantially reduced consistent with the notion that AURKA is a key determinant of WDR62 spindle recruitment. Thus, our study highlights the role of AURKA signaling in the spatiotemporal control of WDR62 at spindle poles where it maintains spindle organization.


Subject(s)
Aurora Kinase A/metabolism , Nerve Tissue Proteins/metabolism , Spindle Apparatus/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chromosome Segregation , HeLa Cells , Humans , Metaphase , Mice , Microscopy, Fluorescence , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction
5.
Eur J Pharmacol ; 758: 53-9, 2015 Jul 05.
Article in English | MEDLINE | ID: mdl-25820159

ABSTRACT

The novel pro-drug of 3'4'-dihydroxyflavonol, NP202, potently reduces myocardial infarct size resulting from ischemia-reperfusion (I/R) through mechanisms that remain to be fully defined. In this study, we investigated whether cardioprotection induced by NP202 depended on activation of the reperfusion injury survival kinase (RISK) pathways. We therefore examined the effects of PD98059 and LY294002, specific inhibitors of the MEK/ERK1/2 and PI3K/Akt pathways, respectively. In isolated cardiomyocytes, H2O2induced oxidative stress activated ERK1/2 and this was further enhanced by DiOHF, the active parent compound of NP202. Although oxidative stress did not stimulate Akt in cardiomyocytes, co-treatment with DiOHF substantially increased Akt phosphorylation. This suggests that DiOHF is a potent modulator of RISK pathways specifically in the context of stress stimulation. In anesthetised sheep, following 1h ischemia and 3h reperfusion, the contribution of the RISK pathways to NP202-mediated cardioprotection was determined by treating the animals with PD98059, LY294002 or vehicle prior to NP202 administration and reperfusion. Infarct size, as a percentage of the area-at-risk, was substantially reduced by NP202 (from 78±6 to 46±4%, P<0.05). Inhibition of MEK/ERK1/2 abolished the cardioprotective effects of NP202 (infarct size 81±4%), whereas inhibition of PI3K/Akt had no effect (infarct size 53±4%). Our combined cellular and animal studies indicate that NP202 potently protects against myocardial I/R injury through complex mechanisms that involved augmentation of MEK/ERK1/2 signaling, but not PI3K/Akt signaling.


Subject(s)
Flavonols/pharmacology , MAP Kinase Signaling System/drug effects , Myocardial Reperfusion Injury/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , Animals , Cardiotonic Agents/pharmacology , Chromones/therapeutic use , Drug Therapy, Combination , Female , Flavonoids/therapeutic use , Flavonols/therapeutic use , Morpholines/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Prodrugs/pharmacology , Prodrugs/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sheep
6.
J Cell Sci ; 128(3): 527-40, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25501809

ABSTRACT

WD40-repeat protein 62 (WDR62) is a spindle pole protein required for normal cell division and neuroprogenitor differentiation during brain development. Microcephaly-associated mutations in WDR62 lead to mitotic mislocalization, highlighting a crucial requirement for precise WDR62 spatiotemporal distribution, although the regulatory mechanisms are unknown. Here, we demonstrate that the WD40-repeat region of WDR62 is required for microtubule association, whereas the disordered C-terminal region regulates cell-cycle-dependent compartmentalization. In agreement with a functional requirement for the WDR62­JNK1 complex during neurogenesis, WDR62 specifically recruits JNK1 (also known as MAPK8), but not JNK2 (also known as MAPK9), to the spindle pole. However, JNK-mediated phosphorylation of WDR62 T1053 negatively regulated microtubule association, and loss of JNK signaling resulted in constitutive WDR62 localization to microtubules irrespective of cell cycle stage. In contrast, we identified that Aurora A kinase (AURKA) and WDR62 were in complex and that AURKA-mediated phosphorylation was required for the spindle localization of WDR62 during mitosis. Our studies highlight complex regulation of WDR62 localization, with opposing roles for JNK and AURKA in determining its spindle association.


Subject(s)
Aurora Kinase A/metabolism , Microtubules/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Nerve Tissue Proteins/metabolism , Spindle Apparatus/metabolism , Cell Cycle Proteins , Cell Line , HeLa Cells , Humans , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 8/genetics , Mitosis/genetics , Neurogenesis/genetics , Phosphorylation , Protein Structure, Tertiary
7.
Biochem Biophys Res Commun ; 453(3): 576-81, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25301550

ABSTRACT

The c-Jun N-terminal Kinases (JNKs) play important roles in cell responses to stress or growth factor stimulation. The JNK1α1 isoform shares >90% identity with a predominantly neuronal JNK3α1 isoform, but JNK3α1 also includes a distinctive 38 amino acid N-terminal sequence. To address the outstanding question of the potential for these JNK isoforms to have different binding partners that mediate different biological actions, the work presented here refined the yeast two-hybrid approach to identify and categorize binding partners for JNK1α1 and JNK3α1. Specifically, site-directed mutagenesis of the JNK1α1 common docking (CD) domain that mediates typical JNK-binding domain (JBD)-dependent interactions, truncation of the distinctive JNK3 N-terminal domain (i.e. ΔN JNK3α1), and interaction evaluation in the yeast two-hybrid system defined the interacting partners as either JNK1-specific interactors (ATF7, FUS, KCNE4, PIAS1, SHANK1, TKT), typical JBD-dependent interactors shared by JNK1α1 and JNK3α1 (AKAP6, BMPR2, EEF1A1, GFAP, GRIP2, GTF2F1, HDAC2, MAP1B, MYO9B, PTPN2, RABGAP1, RUSC2, SUMO1, SYPL1, TOPBP1, ZNF668), or JNK3-specific partners (ATXN1, NNAT, PTGDS) dependent on interaction with the JNK3 N-terminal extension. The interacting partners ATF7, AKAP6, and ATXN1 were explored further as representatives of these different classes. Two potential JBDs were identified in ATF7 as important for its interaction with JNK1α1, but additionally an interaction between ATF7 and ΔN JNK3α1 was shown to be JBD-dependent, suggesting that the JNK3α1 N-terminus prevents interaction with some proteins. For the shared partner AKAP6, one of the multiple potential JBDs predicted by sequence analysis was important for the AKAP6-JNK interaction in the yeast screening system as well as in mammalian cells. Finally, the ATXN1-JNK3α1 interaction was dependent on the JNK3α1 N-terminus in a mammalian cell context. These studies therefore highlight a diversity of potential JNK-interacting partners with both JBD-dependent as well as JBD-independent modes of interaction.


Subject(s)
Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Binding Sites , Mitogen-Activated Protein Kinase 10/chemistry , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 8/chemistry , Mitogen-Activated Protein Kinase 8/genetics , Two-Hybrid System Techniques
8.
Biochem Biophys Res Commun ; 446(1): 248-54, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24589734

ABSTRACT

The stathmin (STMN) family of tubulin-binding phosphoproteins are critical regulators of interphase microtubule dynamics and organization in a broad range of cellular processes. c-Jun N-terminal kinase (JNK) signalling to STMN family proteins has been implicated specifically in neuronal maturation, degeneration and cell stress responses more broadly. Previously, we characterized mechanisms underlying JNK phosphorylation of STMN at proline-flanked serine residues (Ser25 and Ser38) that are conserved across STMN-like proteins. In this study, we demonstrated using in vitro kinase assays and alanine replacement of serine residues that JNK phosphorylated the STMN-like domain (SLD) of SCG10 on Ser73, consistent with our previous finding that STMN Ser38 was the primary JNK target site. In addition, we confirmed that a JNK binding motif ((41)KKKDLSL(47)) that facilitates JNK targeting of STMN is conserved in SCG10. In contrast, SCLIP was phosphorylated by JNK primarily on Ser60 which corresponds to Ser25 on STMN. Moreover, although the JNK-binding motif identified in STMN and SCG10 was not conserved in SCLIP, JNK phosphorylation of SCLIP was inhibited by a substrate competitive peptide (TI-JIP) highlighting kinase-substrate interaction as required for JNK targeting. Thus, STMN and SCG10 are similarly targeted by JNK but there are clear differences in JNK recognition and phosphorylation of the closely related family member, SCLIP.


Subject(s)
Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Stathmin/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Conserved Sequence , Humans , MAP Kinase Signaling System , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 8/genetics , Mutagenesis, Site-Directed , Phosphorylation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Stathmin/chemistry , Stathmin/genetics , Substrate Specificity
9.
J Biol Chem ; 289(4): 2157-69, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24302736

ABSTRACT

Dynamic microtubule changes after a cell stress challenge are required for cell survival and adaptation. Stathmin (STMN), a cytoplasmic microtubule-destabilizing phosphoprotein, regulates interphase microtubules during cell stress, but the signaling mechanisms involved are poorly defined. In this study ectopic expression of single alanine-substituted phospho-resistant mutants demonstrated that STMN Ser-38 and Ser-63 phosphorylation were specifically required to maintain interphase microtubules during hyperosmotic stress. STMN was phosphorylated on Ser-38 and Ser-63 in response to hyperosmolarity, heat shock, and arsenite treatment but rapidly dephosphorylated after oxidative stress treatment. Two-dimensional PAGE and Phos-tag gel analysis of stress-stimulated STMN phospho-isoforms revealed rapid STMN Ser-38 phosphorylation followed by subsequent Ser-25 and Ser-63 phosphorylation. Previously, we delineated stress-stimulated JNK targeting of STMN. Here, we identified cAMP-dependent protein kinase (PKA) signaling as responsible for stress-induced STMN Ser-63 phosphorylation. Increased cAMP levels induced by cholera toxin triggered potent STMN Ser-63 phosphorylation. Osmotic stress stimulated an increase in PKA activity and elevated STMN Ser-63 and CREB (cAMP-response element-binding protein) Ser-133 phosphorylation that was substantially attenuated by pretreatment with H-89, a PKA inhibitor. Interestingly, PKA activity and subsequent phosphorylation of STMN were augmented in the absence of JNK activation, indicating JNK and PKA pathway cross-talk during stress regulation of STMN. Taken together our study indicates that JNK- and PKA-mediated STMN Ser-38 and Ser-63 phosphorylation are required to preserve interphase microtubules in response to hyperosmotic stress.


Subject(s)
Interphase/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Microtubules/metabolism , Osmotic Pressure/physiology , Signal Transduction/physiology , Stathmin/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases , JNK Mitogen-Activated Protein Kinases/genetics , Microtubules/genetics , PC12 Cells , Phosphorylation/physiology , Rats , Stathmin/genetics
10.
Biochim Biophys Acta ; 1843(2): 253-64, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24184208

ABSTRACT

The c-Jun N-terminal kinases (JNKs) are a group of stress-activated protein kinases that regulate gene expression changes through specific phosphorylation of nuclear transcription factor substrates. To address the mechanisms underlying JNK nuclear entry, we employed a semi-intact cell system to demonstrate for the first time that JNK1 nuclear entry is dependent on the importin α2/ß1 heterodimer and independent of importins α3, α4, ß2, ß3, 7 and 13. However, quantitative image analysis of JNK1 localization following exposure of cells to either arsenite or hyperosmotic stress did not indicate its nuclear accumulation. Extending our analyses to define the dynamics of nuclear trafficking of JNK1, we combined live cell imaging analyses with fluorescence recovery after photobleaching (FRAP) protocols. Subnuclear and subcytoplasmic bleaching protocols revealed the slowed movement of JNK1 in both regions in response to hyperosmotic stress. Strikingly, while movement into the nucleus of green fluorescent protein (GFP) or transport of a GFP-T-antigen fusion protein as estimated by initial rates and time to reach half-maximal recovery (t1/2) measures remained unaltered, hyperosmotic stress slowed the nuclear entry of GFP-JNK1. In contrast, arsenite exposure which did not alter the initial rates of nuclear accumulation of GFP, GFP-T-antigen or GFP-JNK1, decreased the t1/2 for nuclear accumulation of both GFP and GFP-JNK1. Thus, our results challenge the paradigm of increased nuclear localization of JNK broadly in response to all forms of stress-activation and are consistent with enhanced interactions of stress-activated JNK1 with scaffold and substrate proteins throughout the nucleus and the cytosol under conditions of hyperosmotic stress.


Subject(s)
Cell Nucleus/metabolism , Intracellular Space/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Osmotic Pressure , Sorbitol/pharmacology , Stress, Physiological , Animals , Antigens, Polyomavirus Transforming/metabolism , Arsenites/pharmacology , Cell Nucleus/drug effects , Enzyme Activation/drug effects , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Intracellular Space/drug effects , Karyopherins/metabolism , Kinetics , Mice , Osmotic Pressure/drug effects , Phosphorylation/drug effects , Protein Transport/drug effects , Rats , Stress, Physiological/drug effects , Subcellular Fractions/enzymology
11.
Biochim Biophys Acta ; 1843(3): 483-94, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24286865

ABSTRACT

Although cytokine-driven STAT3 phosphorylation and activation are often transient, persistent activation of STAT3 is a hallmark of a range of pathologies and underpins altered transcriptional responses. As triggers in disease frequently include combined increases in inflammatory cytokine and reactive oxygen species levels, we report here how oxidative stress impacts on cytokine-driven STAT3 signal transduction events. In the model system of murine embryonic fibroblasts (MEFs), combined treatment with the interleukin-6 family cytokine Leukemia Inhibitory Factor (LIF) and hydrogen peroxide (H2O2) drove persistent STAT3 phosphorylation whereas STAT3 phosphorylation increased only transiently in response to LIF alone and was not increased by H2O2 alone. Surprisingly, increases in transcript levels of the direct STAT3 gene target SOCS3 were delayed during the combined LIF + H2O2 treatment, leading us to probe the impact of oxidative stress on STAT3 regulatory events. Indeed, LIF + H2O2 prolonged JAK activation, delayed STAT3 nuclear localisation, and caused relocalisation of nuclear STAT3 phosphatase TC-PTP (TC45) to the cytoplasm. In exploring the nuclear import/ export pathways, we observed disruption of nuclear/cytoplasmic distributions of Ran and importin-alpha3 in cells exposed to H2O2 and the resultant reduced nuclear trafficking of Classical importin-alpha/3-dependent protein cargoes. CRM1-mediated nuclear export persisted despite the oxidative stress insult, with sustained STAT3 Y705 phosphorylation enhancing STAT3 nuclear residency. Our studies thus reveal for the first time the striking impact of oxidative stress to sustain STAT3 phosphorylation and nuclear retention following disruption of multiple regulatory events, with significant implications for STAT3 function.


Subject(s)
Hydrogen Peroxide/pharmacology , Interleukin-6/metabolism , Leukemia Inhibitory Factor/pharmacology , Oxidative Stress/physiology , STAT3 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , HeLa Cells , Humans , Janus Kinases/metabolism , Leukemia Inhibitory Factor/metabolism , Mice , Oxidative Stress/drug effects , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , src-Family Kinases/metabolism
12.
Biochem J ; 456(2): 149-61, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24032640

ABSTRACT

DiOHF (3',4'-dihydroxyflavonol) is cardioprotective against I/R (ischaemia/reperfusion) injury. The biological activities of flavonols are associated with kinase modulation to alter cell signalling. We thus investigated the effects of DiOHF on the activation of MAPKs (mitogen-activated protein kinases) that regulate the cardiac stress response. In an ovine model of I/R, JNK (c-Jun N-terminal kinase), p38(MAPK), ERK (extracellular-signal-regulated kinase) and Akt were activated, and NP202, a pro-drug of DiOHF, reduced infarct size and inhibited JNK and p38(MAPK) activation, whereas ERK and Akt phosphorylation were unaltered. Similarly, in cultured myoblasts, DiOHF pre-treatment preserved viability and inhibited activation of JNK and p38(MAPK), but not ERK in response to acute oxidative and chemotoxic stress. Furthermore, DiOHF prevented stress-activation of the direct upstream regulators MKK4/7 (MAPK kinase 4/7) and MKK3/6 respectively. We utilized small-molecule affinity purification and identified CaMKII (Ca(2+)/calmodulin-dependent protein kinase II) as a kinase targeted by DiOHF and demonstrated potent CaMKII inhibition by DiOHF in vitro. Moreover, the specific inhibition of CaMKII with KN-93, but not KN-92, prevented oxidative stress-induced activation of JNK and p38(MAPK). The present study indicates DiOHF inhibition of CaMKII and attenuation of MKK3/6→p38(MAPK) and MKK4/7→JNK signalling as a requirement for the protective effects of DiOHF against stress stimuli and myocardial I/R injury.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Flavonols/pharmacology , MAP Kinase Signaling System , Myocardial Reperfusion Injury/drug therapy , Animals , Arsenites/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Hydrogen Peroxide/pharmacology , MAP Kinase Kinase 4/metabolism , Mice , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Oxidants/pharmacology , Oxidative Stress/drug effects , Phosphorylation , Protein Processing, Post-Translational , Rats , Sheep, Domestic , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Cell Sci ; 125(Pt 21): 5096-109, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22899712

ABSTRACT

The impact of aberrant centrosomes and/or spindles on asymmetric cell division in embryonic development indicates the tight regulation of bipolar spindle formation and positioning that is required for mitotic progression and cell fate determination. WD40-repeat protein 62 (WDR62) was recently identified as a spindle pole protein linked to the neurodevelopmental defect of microcephaly but its roles in mitosis have not been defined. We report here that the in utero electroporation of neuroprogenitor cells with WDR62 siRNAs induced their cell cycle exit and reduced their proliferative capacity. In cultured cells, we demonstrated cell-cycle-dependent accumulation of WDR62 at the spindle pole during mitotic entry that persisted until metaphase-anaphase transition. Utilizing siRNA depletion, we revealed WDR62 function in stabilizing the mitotic spindle specifically during metaphase. WDR62 loss resulted in spindle orientation defects, decreased the integrity of centrosomes displaced from the spindle pole and delayed mitotic progression. Additionally, we revealed JNK phosphorylation of WDR62 is required for maintaining metaphase spindle organization during mitosis. Our study provides the first functional characterization of WDR62 and has revealed requirements for JNK/WDR62 signaling in mitotic spindle regulation that may be involved in coordinating neurogenesis.


Subject(s)
Cell Cycle Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Metaphase , Microtubule-Associated Proteins/metabolism , Protein Processing, Post-Translational , Spindle Apparatus/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Proliferation , Centrosome/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Female , Gene Knockdown Techniques , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Microcephaly , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Nerve Tissue Proteins , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Phosphorylation , Prophase , Protein Transport , RNA, Small Interfering/genetics
14.
J Biol Chem ; 286(2): 1576-87, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21056972

ABSTRACT

Excessive proliferation and stabilization of the microtubule (MT) array in cardiac myocytes can accompany pathological cardiac hypertrophy, but the molecular control of these changes remains poorly characterized. In this study, we examined MT stabilization in two independent murine models of heart failure and revealed increases in the levels of post-translationally modified stable MTs, which were closely associated with STAT3 activation. To explore the molecular signaling events contributing to control of the cardiac MT network, we stimulated cardiac myocytes with an α-adrenergic agonist phenylephrine (PE), and observed increased tubulin content without changes in detyrosinated (glu-tubulin) stable MTs. In contrast, the hypertrophic interleukin-6 (IL6) family cytokines increased both the glu-tubulin content and glu-MT density. When we examined a role for ERK in regulating cardiac MTs, we showed that the MEK/ERK-inhibitor U0126 increased glu-MT density in either control cardiac myocytes or following exposure to hypertrophic agents. Conversely, expression of an activated MEK1 mutant reduced glu-tubulin levels. Thus, ERK signaling antagonizes stabilization of the cardiac MT array. In contrast, inhibiting either JAK2 with AG490, or STAT3 signaling with Stattic or siRNA knockdown, blocked cytokine-stimulated increases in glu-MT density. Furthermore, the expression of a constitutively active STAT3 mutant triggered increased glu-MT density in the absence of hypertrophic stimulation. Thus, STAT3 activation contributes substantially to cytokine-stimulated glu-MT changes. Taken together, our results highlight the opposing actions of STAT3 and ERK pathways in the regulation of MT changes associated with cardiac myocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Microtubules/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cardiomegaly/pathology , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Cells, Cultured , Disease Models, Animal , Humans , Interleukin-6/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Processing, Post-Translational/physiology , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Tubulin/genetics , Tubulin/metabolism
15.
J Biol Chem ; 285(37): 29001-13, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20630875

ABSTRACT

The cell stress response encompasses the range of intracellular events required for adaptation to stimuli detrimental to cell survival. Although the c-Jun N-terminal kinase (JNK) is a stress-activated kinase that can promote either cell survival or death in response to detrimental stimuli, the JNK-regulated mechanisms involved in survival are not fully characterized. Here we show that in response to hyperosmotic stress, JNK phosphorylates a key cytoplasmic microtubule regulatory protein, stathmin (STMN), on conserved Ser-25 and Ser-38 residues. In in vitro biochemical studies, we identified STMN Ser-38 as the critical residue required for efficient phosphorylation by JNK and identified a novel kinase interaction domain in STMN required for recognition by JNK. We revealed that JNK was required for microtubule stabilization in response to hyperosmotic stress. Importantly, we also demonstrated a novel cytoprotective function for STMN, as the knockdown of STMN levels by siRNA was sufficient to augment viability in response to hyperosmotic stress. Our findings show that JNK targeting of STMN represents a novel stress-activated cytoprotective mechanism involving microtubule network changes.


Subject(s)
MAP Kinase Kinase 4/metabolism , Microtubules/metabolism , Stathmin/metabolism , Animals , Cell Survival/physiology , MAP Kinase Kinase 4/genetics , Mice , Microtubules/genetics , Osmotic Pressure/physiology , Phosphorylation , Protein Structure, Tertiary , Stathmin/genetics
16.
Biochem J ; 430(2): 345-54, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20594188

ABSTRACT

The JNKs (c-Jun N-terminal kinases) are stress-activated serine/threonine kinases that can regulate both cell death and cell proliferation. We have developed a cell system to control JNK re-expression at physiological levels in JNK1/2-null MEFs (murine embryonic fibroblasts). JNK re-expression restored basal and stress-activated phosphorylation of the c-Jun transcription factor and attenuated cellular proliferation with increased cells in G1/S-phase of the cell cycle. To explore JNK actions to regulate cell proliferation, we evaluated a role for the cytosolic protein, STMN (stathmin)/Op18 (oncoprotein 18). STMN, up-regulated in a range of cancer types, plays a crucial role in the control of cell division through its regulation of microtubule dynamics of the mitotic spindle. In JNK1/2-null or c-Jun-null MEFs or cells treated with c-Jun siRNA (small interfering RNA), STMN levels were significantly increased. Furthermore, a requirement for JNK/cJun signalling was demonstrated by expression of wild-type c-Jun, but not a phosphorylation-defective c-Jun mutant, being sufficient to down-regulate STMN. Critically, shRNA (small hairpin RNA)-directed STMN down-regulation in JNK1/2-null MEFs attenuated proliferation. Thus JNK/c-Jun regulation of STMN levels provides a novel pathway in regulation of cell proliferation with important implications for understanding the actions of JNK as a physiological regulator of the cell cycle and tumour suppressor protein.


Subject(s)
Cell Proliferation , Down-Regulation , Fibroblasts/cytology , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Stathmin/metabolism , Animals , Cells, Cultured , Fibroblasts/enzymology , Fibroblasts/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Phosphorylation , Proto-Oncogene Proteins c-jun/genetics , Stathmin/genetics
17.
Biochim Biophys Acta ; 1804(3): 463-75, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19900593

ABSTRACT

c-Jun N-terminal kinases (JNKs), first characterized as stress-activated members of the mitogen-activated protein kinase (MAPK) family, have become a focus of inhibitor screening strategies following studies that have shown their critical roles in the development of a number of diseases, such as diabetes, neurodegeneration and liver disease. We discuss recent advances in the discovery and development of ATP-competitive and ATP-noncompetitive JNK inhibitors. Because understanding the modes of actions of these inhibitors and improving their properties will rely on a better understanding of JNK structure, JNK catalytic mechanisms and substrates, recent advances in these areas of JNK biochemistry are also considered. In addition, the use of JNK gene knockout animals is continuing to reveal in vivo functions for these kinases, with tissue-specific roles now being dissected with tissue-specific knockouts. These latest advances highlight the many challenges now faced, particularly in the directed targeting of the JNK isoforms in specific tissues.


Subject(s)
JNK Mitogen-Activated Protein Kinases/chemistry , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Protein Kinase Inhibitors/chemistry , Animals , Catalysis , Diabetes Mellitus/drug therapy , Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Gene Knockdown Techniques , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , Liver Diseases/drug therapy , Liver Diseases/enzymology , Liver Diseases/genetics , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/genetics , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary/genetics
SELECTION OF CITATIONS
SEARCH DETAIL