Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Neurobiol Dis ; 115: 69-81, 2018 07.
Article in English | MEDLINE | ID: mdl-29627578

ABSTRACT

Adult neurogenesis is involved in certain hippocampus-dependent cognitive functions and is linked to psychiatric diseases including intellectual disabilities. The Coffin-Lowry syndrome (CLS) is a developmental disorder caused by mutations in the Rsk2 gene and characterized by intellectual disabilities associated with growth retardation. How RSK2-deficiency leads to cognitive dysfunctions in CLS is however poorly understood. Here, using Rsk2 Knock-Out mice, we characterized the impact of RSK2 deficiency on adult hippocampal neurogenesis in vivo. We report that the absence of RSK2 does not affect basal proliferation, differentiation and survival of dentate gyrus adult-born neurons but alters the maturation progression of young immature newborn neurons. Moreover, when RSK2-deficient mice were submitted to spatial learning, in contrast to wild-type mice, proliferation of adult generated neurons was decreased and no pro-survival effect of learning was observed. Thus, learning failed to recruit a selective population of young newborn neurons in association with deficient long-term memory recall. Given the proposed role of the dentate gyrus and of adult-generated newborn neurons in hippocampal-dependent pattern separation function, we explored this function in a delayed non-matching to place task and in an object-place pattern separation task and report severe deficits in spatial pattern separation in Rsk2-KO mice. Together, this study reveals a previously unknown role for RSK2 in the early stages of maturation and learning-dependent involvement of adult-born dentate gyrus neurons. These alterations associated with a deficit in the ability of RSK2-deficient mice to finely discriminate relatively similar spatial configurations, may contribute to cognitive dysfunction in CLS.


Subject(s)
Coffin-Lowry Syndrome/physiopathology , Disease Models, Animal , Hippocampus/physiopathology , Neurogenesis/physiology , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Spatial Behavior/physiology , Age Factors , Animals , Animals, Newborn , Coffin-Lowry Syndrome/genetics , Hippocampus/pathology , Male , Mice , Mice, Knockout , Ribosomal Protein S6 Kinases, 90-kDa/genetics
2.
Sci Rep ; 5: 14778, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26437780

ABSTRACT

The brain-derived neurotrophic factor BDNF plays a critical role in neuronal development and the induction of L-LTP at glutamatergic synapses in several brain regions. However, the cellular and molecular mechanisms underlying these BDNF effects have not been firmly established. Using in vitro cultures of cortical neurons from knockout mice for Pld1 and Rsk2, BDNF was observed to induce a rapid RSK2-dependent activation of PLD and to stimulate BDNF ERK1/2-CREB and mTor-S6K signalling pathways, but these effects were greatly reduced in Pld1(-/-) neurons. Furthermore, phospho-CREB did not accumulate in the nucleus, whereas overexpression of PLD1 amplified the BDNF-dependent nuclear recruitment of phospho-ERK1/2 and phospho-CREB. This BDNF retrograde signalling was prevented in cells silenced for the scaffolding protein PEA15, a protein which complexes with PLD1, ERK1/2, and RSK2 after BDNF treatment. Finally PLD1, ERK1/2, and RSK2 partially colocalized on endosomal structures, suggesting that these proteins are part of the molecular module responsible for BDNF signalling in cortical neurons.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Neurons/metabolism , Phospholipase D/genetics , Signal Transduction , Animals , Apoptosis Regulatory Proteins , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Endosomes/metabolism , Gene Expression Regulation , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/cytology , Neurons/drug effects , Phospholipase D/deficiency , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Primary Cell Culture , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
J Immunol ; 195(9): 4264-72, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26401001

ABSTRACT

Langerhans cells (LC), the dendritic cells of the epidermis, are distributed in a distinctive regularly spaced array. In the mouse, the LC array is established in the first few days of life from proliferating local precursors, but the regulating signaling pathways are not fully understood. We found that mice lacking the kinase phosphoinositide-dependent kinase 1 selectively lack LC. Deletion of the phosphoinositide-dependent kinase 1 target kinases, ribosomal S6 kinase 1 (Rsk1) and Rsk2, produced a striking perturbation in the LC network: LC density was reduced 2-fold, but LC size was increased by the same magnitude. Reduced LC numbers in Rsk1/2(-/-) mice was not due to accelerated emigration from the skin but rather to reduced proliferation at least in adults. Rsk1/2 were required for normal LC patterning in neonates, but not when LC were ablated in adults and replaced by bone marrow-derived cells. Increased LC size was an intrinsic response to reduced LC numbers, reversible on LC emigration, and could be observed in wild type epidermis where LC size also correlated inversely with LC density. Our results identify a key signaling pathway needed to establish a normal LC network and suggest that LC might maintain epidermal surveillance by increasing their "footprint" when their numbers are limited.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Cell Proliferation , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Animals , Animals, Newborn , Cell Count , Cell Movement , Cell Size , Cells, Cultured , Epidermal Cells , Epidermis/metabolism , Flow Cytometry , Langerhans Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics
4.
Mol Cell Biol ; 35(1): 132-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25332232

ABSTRACT

Rsk kinases play important roles in several cellular processes such as proliferation, metabolism, and migration. Until recently, Rsk activation was thought to be exclusively initiated by Erk1/2, but in dendritic cells (DC) Rsk is also activated by p38 mitogen-activated protein (MAP) kinase via its downstream substrates, MK2/3. How and why this noncanonical configuration of the MAP kinase pathway is adopted by these key immune cells are not known. We demonstrate that the Erk1/2-activated C-terminal kinase domain of Rsk is dispensable for p38-MK2/3 activation and show that compared with fibroblasts, a greater fraction of p38 and MK2/3 is located in the cytosol of DC prior to stimulation, suggesting a partial explanation for the operation of the noncanonical pathway of Rsk activation in these cells. p38/MK2/3-activated Rsk phosphorylated downstream targets and is physiologically important because in plasmacytoid DC (pDC) stimulated with Toll-like receptor 7 (TLR7) agonists, Erk1/2 activation is very weak relative to p38. As a result, Rsk activation is entirely p38 dependent. We show that this unusual configuration of MAP kinase signaling contributes substantially to production of type I interferons, a hallmark of pDC activation.


Subject(s)
Dendritic Cells/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Toll-Like Receptors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Proliferation , Cytokines/metabolism , Cytoplasm/metabolism , Cytosol/metabolism , Enzyme Activation , Gene Expression Regulation, Enzymologic , Hematopoietic Stem Cells/cytology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
5.
PLoS One ; 9(1): e84343, 2014.
Article in English | MEDLINE | ID: mdl-24416220

ABSTRACT

BACKGROUND: The RSK2 gene is responsible for Coffin-Lowry syndrome, an X-linked dominant genetic disorder causing mental retardation, skeletal growth delays, with craniofacial and digital abnormalities typically associated with this syndrome. Craniofacial and dental anomalies encountered in this rare disease have been poorly characterized. METHODOLOGY/PRINCIPAL FINDINGS: We examined, using X-Ray microtomographic analysis, the variable craniofacial dysmorphism and dental anomalies present in Rsk2 knockout mice, a model of Coffin-Lowry syndrome, as well as in triple Rsk1,2,3 knockout mutants. We report Rsk mutation produces surpernumerary teeth midline/mesial to the first molar. This highly penetrant phenotype recapitulates more ancestral tooth structures lost with evolution. Most likely this leads to a reduction of the maxillary diastema. Abnormalities of molar shape were generally restricted to the mesial part of both upper and lower first molars (M1). Expression analysis of the four Rsk genes (Rsk1, 2, 3 and 4) was performed at various stages of odontogenesis in wild-type (WT) mice. Rsk2 is expressed in the mesenchymal, neural crest-derived compartment, correlating with proliferative areas of the developing teeth. This is consistent with RSK2 functioning in cell cycle control and growth regulation, functions potentially responsible for severe dental phenotypes. To uncover molecular pathways involved in the etiology of these defects, we performed a comparative transcriptomic (DNA microarray) analysis of mandibular wild-type versus Rsk2-/Y molars. We further demonstrated a misregulation of several critical genes, using a Rsk2 shRNA knock-down strategy in molar tooth germs cultured in vitro. CONCLUSIONS: This study reveals RSK2 regulates craniofacial development including tooth development and patterning via novel transcriptional targets.


Subject(s)
Craniofacial Abnormalities/enzymology , Head/growth & development , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/pathology , Abnormalities, Multiple/physiopathology , Animals , Craniofacial Abnormalities/pathology , Craniofacial Abnormalities/physiopathology , Enzyme Activation , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , MAP Kinase Signaling System , Male , Mice , Odontogenesis , Phenotype , RNA, Small Interfering/genetics , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Tooth/anatomy & histology , Tooth/growth & development
6.
J Neurosci ; 33(50): 19470-9, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24336713

ABSTRACT

More than 80 human X-linked genes have been associated with mental retardation and deficits in learning and memory. However, most of the identified mutations induce limited morphological alterations in brain organization and the molecular bases underlying neuronal clinical features remain elusive. We show here that neurons cultured from mice lacking ribosomal S6 kinase 2 (Rsk2), a model for the Coffin-Lowry syndrome (CLS), exhibit a significant delay in growth in a similar way to that shown by neurons cultured from phospholipase D1 (Pld1) knock-out mice. We found that gene silencing of Pld1 or Rsk2 as well as acute pharmacological inhibition of PLD1 or RSK2 in PC12 cells strongly impaired neuronal growth factor (NGF)-induced neurite outgrowth. Expression of a phosphomimetic PLD1 mutant rescued the inhibition of neurite outgrowth in PC12 cells silenced for RSK2, revealing that PLD1 is a major target for RSK2 in neurite formation. NGF-triggered RSK2-dependent phosphorylation of PLD1 led to its activation and the synthesis of phosphatidic acid at sites of neurite growth. Additionally, total internal reflection fluorescence microscopy experiments revealed that RSK2 and PLD1 positively control fusion of tetanus neurotoxin insensitive vesicle-associated membrane protein (TiVAMP)/VAMP-7 vesicles at sites of neurite outgrowth. We propose that the loss of function mutations in RSK2 that leads to CLS and neuronal deficits are related to defects in neuronal growth due to impaired RSK2-dependent PLD1 activity resulting in a reduced vesicle fusion rate and membrane supply.


Subject(s)
Neurites/metabolism , Phosphatidic Acids/biosynthesis , Phospholipase D/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Cells, Cultured , Coffin-Lowry Syndrome/genetics , Coffin-Lowry Syndrome/metabolism , Mice , Mice, Knockout , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Phosphorylation , Rats , Ribosomal Protein S6 Kinases, 90-kDa/genetics
7.
Neurobiol Dis ; 58: 156-68, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23742761

ABSTRACT

The Coffin-Lowry syndrome (CLS) is a syndromic form of intellectual disability caused by loss-of-function of the RSK2 serine/threonine kinase encoded by the rsk2 gene. Rsk2 knockout mice, a murine model of CLS, exhibit spatial learning and memory impairments, yet the underlying neural mechanisms are unknown. In the current study, we examined the performance of Rsk2 knockout mice in cued, trace and contextual fear memory paradigms and identified selective deficits in the consolidation and reconsolidation of hippocampal-dependent fear memories as task difficulty and hippocampal demand increase. Electrophysiological, biochemical and electron microscopy analyses were carried out in the dentate gyrus of the hippocampus to explore potential alterations in neuronal functions and structure. In vivo and in vitro electrophysiology revealed impaired synaptic transmission, decreased network excitability and reduced AMPA and NMDA conductance in Rsk2 knockout mice. In the absence of RSK2, standard measures of short-term and long-term potentiation (LTP) were normal, however LTP-induced CREB phosphorylation and expression of the transcription factors EGR1/ZIF268 were reduced and that of the scaffolding protein SHANK3 was blocked, indicating impaired activity-dependent gene regulation. At the structural level, the density of perforated and non-perforated synapses and of multiple spine boutons was not altered, however, a clear enlargement of spine neck width and post-synaptic densities indicates altered synapse ultrastructure. These findings show that RSK2 loss-of-function is associated in the dentate gyrus with multi-level alterations that encompass modifications of glutamate receptor channel properties, synaptic transmission, plasticity-associated gene expression and spine morphology, providing novel insights into the mechanisms contributing to cognitive impairments in CLS.


Subject(s)
Coffin-Lowry Syndrome/complications , Coffin-Lowry Syndrome/genetics , Dentate Gyrus/pathology , Fear , Memory Disorders/etiology , Mutation/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Synaptic Transmission/genetics , Animals , Conditioning, Psychological/physiology , Cues , Dentate Gyrus/ultrastructure , Disease Models, Animal , Electric Stimulation , Excitatory Postsynaptic Potentials/genetics , Freezing Reaction, Cataleptic/physiology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , N-Methylaspartate/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission/physiology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
8.
Int J Mol Sci ; 14(2): 3358-75, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23389038

ABSTRACT

The RSK2 protein is a member of the RSK serine-threonine protein kinase family and is encoded by the X-linked rps6ka3 gene in human. Highly heterogeneous loss-of-function mutations affecting this gene are responsible for a severe syndromic form of cognitive impairment, Coffin-Lowry syndrome. RSK2, which is highly conserved in mammals, acts at the distal end of the Ras-ERK signaling pathway and is activated in response to growth factors and neurotransmitters. RSK2 is highly expressed in the hippocampus, and Rsk2-KO mice display spatial learning and memory impairment. We recently showed that ERK1/2 activity is abnormally increased in the hippocampus of Rsk2-KO mice as well as the expression of the AMPA receptor subunit GluR2. The mechanism via which RSK2 deficiency affects the expression of GluR2 in neural cells was unknown. To address this issue we constitutively suppressed the expression of RSK2 in PC12 cells via vector-based shRNA in the present study. We show that Rsk2 silencing leads also to an elevation of ERK1/2 phosphorylation as well as of GluR2 expression and that the increased level of GluR2 expression results from the increased ERK1/2 activity on the transcription factor Sp1. Our results provide evidence that RSK2 modulates ERK1/2 activity on Sp1, which regulates GluR2 expression through transcriptional activation.

9.
Eur J Med Genet ; 56(3): 150-2, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23261961

ABSTRACT

Coffin-Lowry syndrome (CLS) is an X-linked disorder characterized by growth and psychomotor retardation, hypotonia and progressive skeletal changes. RPS6KA3 is the only gene known to be associated with CLS, and over 150 distinct inactivating mutations in this gene have so far been reported in CLS patients. However, no defect is found in about half of the CLS compatible patients by exon sequencing. We report here the first deep intronic mutation in RPS6KA3, which is associated with the retention of intronic sequences in the mRNAs. Indeed, this finding suggests that all the patients with a highly suggestive CLS clinical diagnosis, but in whom exon screening has failed to detect a mutation, should be reanalyzed at the RNA level.


Subject(s)
Coffin-Lowry Syndrome/diagnosis , Coffin-Lowry Syndrome/genetics , Mutation , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Alleles , Base Sequence , Exons , Humans , Introns , Male , Molecular Sequence Data , RNA, Messenger/genetics , Sequence Analysis, DNA , Young Adult
10.
Circ Res ; 112(1): 128-39, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-22997248

ABSTRACT

RATIONALE: Cardiac myocyte hypertrophy is the main compensatory response to chronic stress on the heart. p90 ribosomal S6 kinase (RSK) family members are effectors for extracellular signal-regulated kinases that induce myocyte growth. Although increased RSK activity has been observed in stressed myocytes, the functions of individual RSK family members have remained poorly defined, despite being potential therapeutic targets for cardiac disease. OBJECTIVE: To demonstrate that type 3 RSK (RSK3) is required for cardiac myocyte hypertrophy. METHODS AND RESULTS: RSK3 contains a unique N-terminal domain that is not conserved in other RSK family members. We show that this domain mediates the regulated binding of RSK3 to the muscle A-kinase anchoring protein scaffold, defining a novel kinase anchoring event. Disruption of both RSK3 expression using RNA interference and RSK3 anchoring using a competing muscle A-kinase anchoring protein peptide inhibited the hypertrophy of cultured myocytes. In vivo, RSK3 gene deletion in the mouse attenuated the concentric myocyte hypertrophy induced by pressure overload and catecholamine infusion. CONCLUSIONS: Taken together, these data demonstrate that anchored RSK3 transduces signals that modulate pathologic myocyte growth. Targeting of signaling complexes that contain select kinase isoforms should provide an approach for the specific inhibition of cardiac myocyte hypertrophy and for the development of novel strategies for the prevention and treatment of heart failure.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cardiomegaly/enzymology , Myocytes, Cardiac/enzymology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , A Kinase Anchor Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Newborn , Binding Sites , COS Cells , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Chlorocebus aethiops , Disease Models, Animal , Female , Genotype , HEK293 Cells , Humans , Immunoprecipitation , Isoproterenol , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Phenotype , Protein Interaction Domains and Motifs , Protein Interaction Mapping , RNA Interference , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction , Transduction, Genetic , Transfection
11.
Eur J Med Genet ; 55(5): 335-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22490425

ABSTRACT

The Coffin-Lowry syndrome (CLS) is a rare but well-defined X-linked semidominant syndrome characterized by psychomotor and growth retardation, and progressive skeletal changes. CLS is caused by loss of function mutations in the Rps6ka3 gene encoding the ribosomal S6 kinase 2 (RSK2) protein. A distinctive paroxysmal disorder has been described in some CLS patients, characterized by episodes of sudden falling, without apparent alteration of consciousness, usually induced by unexpected tactile or auditory stimuli. Duration of episodes is very short, usually lasting a few seconds. The appellation "Stimulus-induced drop episodes" (SIDEs) was proposed for these non-epileptic events in CLS patients. SIDEs are clinically heterogeneous; with some patients exhibiting cataplexy-like events characterized by sudden hypotonia and collapse, and others hyperekplexia-like episodes with a startle response. The pathophysiology of SIDEs is not well understood.


Subject(s)
Coffin-Lowry Syndrome/physiopathology , Animals , Cataplexy/epidemiology , Cataplexy/genetics , Cataplexy/physiopathology , Coffin-Lowry Syndrome/epidemiology , Coffin-Lowry Syndrome/genetics , Epilepsy/physiopathology , Humans , Mutation , Prevalence , Reflex, Startle , Ribosomal Protein S6 Kinases, 90-kDa/genetics
12.
Neuropsychopharmacology ; 37(5): 1288-96, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22218090

ABSTRACT

It has been established that mu opioid receptors activate the ERK1/2 signaling cascade both in vitro and in vivo. The Ser/Thr kinase RSK2 is a direct downstream effector of ERK1/2 and has a role in cellular signaling, cell survival growth, and differentiation; however, its role in biological processes in vivo is less well known. Here we determined whether RSK2 contributes to mu-mediated signaling in vivo. Knockout mice for the rsk2 gene were tested for main morphine effects, including analgesia, tolerance to analgesia, locomotor activation, and sensitization to this effect, as well as morphine withdrawal. The deletion of RSK2 reduced acute morphine analgesia in the tail immersion test, indicating a role for this kinase in mu receptor-mediated nociceptive processing. All other morphine effects and adaptations to chronic morphine were unchanged. Because the mu opioid receptor and RSK2 both show high density in the habenula, we specifically downregulated RSK2 in this brain metastructure using an adeno-associated-virally mediated shRNA approach. Remarkably, morphine analgesia was significantly reduced, as observed in the total knockout animals. Together, these data indicate that RSK2 has a role in nociception, and strongly suggest that a mu opioid receptor-RSK2 signaling mechanism contributes to morphine analgesia at the level of habenula. This study opens novel perspectives for both our understanding of opioid analgesia, and the identification of signaling pathways operating in the habenular complex.


Subject(s)
Analgesics, Opioid/pharmacology , Habenula/drug effects , Habenula/metabolism , Morphine/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Drug Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine Dependence/drug therapy , Morphine Dependence/etiology , Morphine Dependence/genetics , Motor Activity/drug effects , Motor Activity/genetics , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nociception/drug effects , Nociception/physiology , Pain Measurement/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Signal Transduction/drug effects , Transduction, Genetic
13.
J Neurochem ; 119(3): 447-59, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21838783

ABSTRACT

Coffin-Lowry syndrome is a syndromic form of mental retardation caused by mutations of the Rps6ka3 gene encoding ribosomal s6 kinase (RSK)2. RSK2 belongs to a family containing four members in mammals: RSK1-4. RSKs are serine/threonine kinases and cytosolic substrates of extracellular signal-regulated kinase (ERK) in the Ras/MAPK signaling pathway. RSK2 is highly expressed in the hippocampus, and mrsk2_KO mice display spatial learning and memory impairment. In the present study, we provide evidence of abnormally increased phosphorylation of ERK1/2 in the hippocampus of mrsk2_KO mice. Further studies based on cultured hippocampal neurons revealed that glutamate activates ERK1/2 and RSKs, and confirmed a stronger activation of ERK1/2 in mrsk2_KO neurons than in WT cells. We, thus, provide further evidence that RSK2 exerts a feedback inhibitory effect on the ERK1/2 pathway. We also observed a transient sequestration of P-ERK1/2 in the cytoplasm upon glutamate stimulation. In addition, the transcription factors cAMP response element binding and Ets LiKe gene1 show over-activation in RSK2-deficient neurons. Finally, c-Fos, Zif268 and Arc were significantly over-expressed in mrsk2_KO neurons upon glutamate stimulation. Importantly, the increased phosphorylation of other RSK family members observed in mutant neurons was unable to compensate for RSK2 deficiency. This aberrant ERK1/2 signaling can influence various neuronal functions, and thus play a significant role in cognitive dysfunction in mrsk2_KO mice and in the Coffin-Lowry syndrome.


Subject(s)
Coffin-Lowry Syndrome/genetics , Disease Models, Animal , Hippocampus/enzymology , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Animals , Cells, Cultured , Coffin-Lowry Syndrome/enzymology , Cognition Disorders/enzymology , Cognition Disorders/genetics , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/physiology , Mutation , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/physiology
14.
Learn Mem ; 18(9): 574-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21852432

ABSTRACT

RSK2 is a Ser/Thr kinase acting in the Ras/MAPK pathway. Rsk2 gene deficiency leads to the Coffin-Lowry Syndrome, notably characterized by cognitive deficits. We found that mrsk2 knockout mice are unable to associate an aversive stimulus with context in a lithium-induced conditioned place aversion task requiring both high-order cognition and emotional processing. Virally mediated shRNA-RSK2 knockdown in the habenula, whose involvement in cognition is receiving increasing attention, also ablated contextual conditioning. RSK2 signaling in the habenula, therefore, is essential for this task. Our study reveals a novel role for RSK2 in cognitive processes and uncovers the critical implication of an intriguing brain structure in place aversion learning.


Subject(s)
Avoidance Learning/physiology , Habenula/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Antimanic Agents/pharmacology , Avoidance Learning/radiation effects , COS Cells , Chlorocebus aethiops , Conditioning, Operant/drug effects , Habenula/drug effects , Lithium Chloride/pharmacology , Luminescent Proteins/genetics , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection/methods
15.
Hum Genet ; 129(3): 255-69, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21116650

ABSTRACT

Coffin-Lowry syndrome (CLS) is a syndromic form of mental retardation caused by loss of function mutations in the X-linked RPS6KA3 gene, which encodes RSK2, a serine/threonine kinase acting in the MAPK/ERK pathway. The mouse invalidated for the Rps6ka3 (Rsk2-KO) gene displays learning and long-term spatial memory deficits. In the current study, we compared hippocampal gene expression profiles from Rsk2-KO and normal littermate mice to identify changes in molecular pathways. Differential expression was observed for 100 genes encoding proteins acting in various biological pathways, including cell growth and proliferation, cell death and higher brain function. The twofold up-regulated gene (Gria2) was of particular interest because it encodes the subunit GLUR2 of the AMPA glutamate receptor. AMPA receptors mediate most fast excitatory synaptic transmission in the central nervous system. We provide evidence that in the hippocampus of Rsk2-KO mice, expression of GLUR2 at the mRNA and at the protein levels is significantly increased, whereas basal AMPA receptor-mediated transmission in the hippocampus of Rsk2-KO mice is significantly decreased. This is the first time that such deregulations have been demonstrated in the mouse model of the Coffin-Lowry syndrome. Our findings suggest that a defect in AMPA neurotransmission and plasticity contribute to mental retardation in CLS patients.


Subject(s)
Coffin-Lowry Syndrome/genetics , Hippocampus/enzymology , Receptors, AMPA/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Animals , Coffin-Lowry Syndrome/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, AMPA/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Synaptic Transmission/genetics , Up-Regulation
16.
Eur J Hum Genet ; 18(6): 627-33, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19888300

ABSTRACT

Coffin-Lowry syndrome (CLS) is a syndromic form of X-linked mental retardation, which is characterized in male patients by psychomotor and growth retardation and various skeletal anomalies. Typical facial changes and specific clinical and radiological signs in the hand are useful aids in the diagnosis. CLS is caused by mutations in the RPS6KA3 gene located at Xp22.2, which encodes RSK2, a growth-factor-regulated protein kinase. RPS6KA3 mutations are extremely heterogeneous and lead to loss of phosphotransferase activity in the RSK2 kinase, most often because of premature termination of translation.


Subject(s)
Coffin-Lowry Syndrome/diagnosis , Coffin-Lowry Syndrome/genetics , Abnormalities, Multiple/genetics , Bone Diseases, Developmental/diagnosis , Bone Diseases, Developmental/epidemiology , Bone Diseases, Developmental/genetics , Coffin-Lowry Syndrome/epidemiology , Humans , Male , Models, Biological , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/physiology
17.
Mol Cell Neurosci ; 42(2): 134-41, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19555761

ABSTRACT

Mutations in Ribosomal s6 kinase 2 (Rsk2) are associated with severe neuronal dysfunction in Coffin-Lowry syndrome (CLS) patients, flies and mice. So far, the mechanisms of how Rsk2 regulates development, maintenance and activity of neurons are not understood. We have investigated the consequences of Rsk2 deficiency in mouse spinal motoneurons. Survival of isolated Rsk2 deficient motoneurons is not reduced, but these cells grow significantly longer neurites. Conversely, overexpression of a constitutively active form of Rsk2 leads to reduced axon growth. Increased axon growth in Rsk2 deficient neurons was accompanied by higher Erk 1/2 phosphorylation, and the knockout phenotype could be rescued by pharmacological inhibition of MAPK/Erk kinase (Mek). These data indicate that Rsk2 negatively regulates axon elongation via the MAPK pathway. Thus, the functional defects observed in the nervous system of CLS patients and animal models with Rsk2 deficiency might be caused by dysregulated neurite growth rather than primary neurodegeneration.


Subject(s)
Axons/physiology , Motor Neurons , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Cell Survival , Female , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/cytology , Motor Neurons/physiology , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Spinal Cord/cytology
18.
Mol Cell Biol ; 29(8): 2105-17, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19223461

ABSTRACT

Dysregulation of the receptor tyrosine kinase fibroblast growth factor receptor 3 (FGFR3) plays a pathogenic role in a number of human hematopoietic malignancies and solid tumors. These include t(4;14) multiple myeloma associated with ectopic expression of FGFR3 and t(4;12)(p16;p13) acute myeloid leukemia associated with expression of a constitutively activated fusion tyrosine kinase, TEL-FGFR3. We recently reported that FGFR3 directly tyrosine phosphorylates RSK2 at Y529, which consequently regulates RSK2 activation. Here we identified Y707 as an additional tyrosine in RSK2 that is phosphorylated by FGFR3. Phosphorylation at Y707 contributes to RSK2 activation, through a putative disruption of the autoinhibitory alphaL-helix on the C terminus of RSK2, unlike Y529 phosphorylation, which facilitates ERK binding. Moreover, we found that FGFR3 interacts with RSK2 through residue W332 in the linker region of RSK2 and that this association is required for FGFR3-dependent phosphorylation of RSK2 at Y529 and Y707, as well as the subsequent RSK2 activation. Furthermore, in a murine bone marrow transplant assay, genetic deficiency in RSK2 resulted in a significantly delayed and attenuated myeloproliferative syndrome induced by TEL-FGFR3 as compared with wild-type cells, suggesting a critical role of RSK2 in FGFR3-induced hematopoietic transformation. Our current and previous findings represent a paradigm for tyrosine phosphorylation-dependent regulation of serine-threonine kinases.


Subject(s)
Hematopoiesis , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Binding Sites , Bone Marrow Transplantation , Mice , Myeloproliferative Disorders/etiology , Phosphorylation , Protein Binding/physiology , Tyrosine/metabolism
19.
J Neurochem ; 107(5): 1325-34, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18823370

ABSTRACT

The Coffin-Lowry syndrome, a rare syndromic form of X-linked mental retardation, is caused by loss-of-function mutations in the hRSK2 (RPS6KA3) gene. To further investigate RSK2 (90-kDa ribosomal S6 kinase) implication in cognitive processes, a mrsk2_KO mouse has previously been generated as an animal model of Coffin-Lowry syndrome. The aim of the present study was to identify possible neurochemical dysregulation associated with the behavioral and morphological abnormalities exhibited by mrsk2_KO mice. A cortical dopamine level increase was found in mrsk2_KO mice that was accompanied by an over-expression of dopamine receptor of type 2 and the dopamine transporter. We also detected an increase of total and phosphorylated extracellular regulated kinase that may be responsible for the increased level of tyrosine hydroxylase phosphorylation also observed. By taking into consideration previously reported data, our results strongly suggest that the dopaminergic dysregulation in mrsk2_KO mice may be caused, at least in part, by tyrosine hydroxylase hyperactivity. This cortical hyperdopaminergia may explain some non-cognitive but also cognitive alterations exhibited by mrsk2_KO mice.


Subject(s)
Coffin-Lowry Syndrome/metabolism , Disease Models, Animal , Dopamine/metabolism , Small-Conductance Calcium-Activated Potassium Channels/deficiency , Animals , Brain/metabolism , Brain/pathology , Chromatography, High Pressure Liquid/methods , Coffin-Lowry Syndrome/pathology , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Dopamine beta-Hydroxylase/genetics , Dopamine beta-Hydroxylase/metabolism , Eukaryotic Initiation Factor-2/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Tyrosine 3-Monooxygenase/metabolism
20.
Proc Natl Acad Sci U S A ; 105(24): 8434-9, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18550821

ABSTRACT

Exocytosis of neurotransmitters and hormones occurs through the fusion of secretory vesicles with the plasma membrane. This highly regulated process involves key proteins, such as SNAREs, and specific lipids at the site of membrane fusion. Phospholipase D (PLD) has recently emerged as a promoter of membrane fusion in various exocytotic events potentially by providing fusogenic cone-shaped phosphatidic acid. We show here that PLD1 is regulated by ribosomal S6 kinase 2 (RSK2)-dependent phosphorylation. RSK2 is activated by a high K(+)-induced rise in cytosolic calcium. Expression of inactive RSK2 mutants or selective knockdown of endogenous RSK2 dramatically affects the different kinetic components of the exocytotic response in chromaffin cells. RSK2 physically interacts with and stimulates PLD activity through the phosphorylation of Thr-147 in the PLD1 amino-terminal phox homology domain. Expression of PLD1 phosphomimetic mutants fully restores secretion in cells depleted of RSK2, suggesting that RSK2 is a critical upstream signaling element in the activation of PLD1 to produce the lipids required for exocytosis. We propose that PLD-related defects in neuronal and endocrine activities could contribute to the effect observed after the loss-of-function mutations in Rsk2 that lead to Coffin-Lowry syndrome, an X-linked form of growth and mental retardation.


Subject(s)
Calcium/metabolism , Chromaffin Cells/metabolism , Coffin-Lowry Syndrome/enzymology , Exocytosis , Phospholipase D/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Chromaffin Cells/enzymology , Coffin-Lowry Syndrome/genetics , Exocytosis/genetics , PC12 Cells , Phosphatidic Acids/metabolism , Phosphorylation , Rats , Ribosomal Protein S6 Kinases, 90-kDa/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...