Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Dent Res ; 95(7): 752-60, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26927527

RESUMO

The ribosomal S6 kinase RSK2 is essential for osteoblast function, and inactivating mutations of RSK2 cause osteopenia in humans with Coffin-Lowry syndrome (CLS). Alveolar bone loss and premature tooth exfoliation are also consistently reported symptoms in CLS patients; however, the pathophysiologic mechanisms are unclear. Therefore, aiming to identify the functional relevance of Rsk2 for tooth development, we analyzed Rsk2-deficient mice. Here, we show that Rsk2 is a critical regulator of cementoblast function. Immunohistochemistry, histology, micro-computed tomography imaging, quantitative backscattered electron imaging, and in vitro assays revealed that Rsk2 is activated in cementoblasts and is necessary for proper acellular cementum formation. Cementum hypoplasia that is observed in Rsk2-deficient mice causes detachment and disorganization of the periodontal ligament and was associated with significant alveolar bone loss with age. Moreover, Rsk2-deficient mice display hypomineralization of cellular cementum with accumulation of nonmineralized cementoid. In agreement, treatment of the cementoblast cell line OCCM-30 with a Rsk inhibitor reduces formation of mineralization nodules and decreases the expression of cementum markers. Western blot analyses based on antibodies against Rsk1, Rsk2, and an activated form of the 2 kinases confirmed that Rsk2 is expressed and activated in differentiating OCCM-30 cells. To discriminate between periodontal bone loss and systemic bone loss, we additionally crossed Rsk2-deficient mice with transgenic mice overexpressing the osteoanabolic transcription factor Fra1. Fra1 overexpression clearly increases systemic bone volume in Rsk2-deficient mice but does not protect from alveolar bone loss. Our results indicate that cell autonomous cementum defects are causing early tooth loss in CLS patients. Moreover, we identify Rsk2 as a nonredundant regulator of cementum homeostasis, alveolar bone maintenance, and periodontal health, with all these features being independent of Rsk2 function in systemic bone formation.


Assuntos
Síndrome de Coffin-Lowry/genética , Cemento Dentário/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Animais , Western Blotting , Calcificação Fisiológica/fisiologia , Síndrome de Coffin-Lowry/enzimologia , Cemento Dentário/anatomia & histologia , Cemento Dentário/citologia , Cemento Dentário/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão por Filtração de Energia , Proteínas Quinases S6 Ribossômicas 90-kDa/deficiência , Microtomografia por Raio-X
2.
Dis Model Mech ; 8(11): 1389-400, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26398944

RESUMO

Plastic changes in synaptic properties are considered as fundamental for adaptive behaviors. Extracellular-signal-regulated kinase (ERK)-mediated signaling has been implicated in regulation of synaptic plasticity. Ribosomal S6 kinase 2 (RSK2) acts as a regulator and downstream effector of ERK. In the brain, RSK2 is predominantly expressed in regions required for learning and memory. Loss-of-function mutations in human RSK2 cause Coffin-Lowry syndrome, which is characterized by severe mental retardation and low IQ scores in affected males. Knockout of RSK2 in mice or the RSK ortholog in Drosophila results in a variety of learning and memory defects. However, overall brain structure in these animals is not affected, leaving open the question of the pathophysiological consequences. Using the fly neuromuscular system as a model for excitatory glutamatergic synapses, we show that removal of RSK function causes distinct defects in motoneurons and at the neuromuscular junction. Based on histochemical and electrophysiological analyses, we conclude that RSK is required for normal synaptic morphology and function. Furthermore, loss of RSK function interferes with ERK signaling at different levels. Elevated ERK activity was evident in the somata of motoneurons, whereas decreased ERK activity was observed in axons and the presynapse. In addition, we uncovered a novel function of RSK in anterograde axonal transport. Our results emphasize the importance of fine-tuning ERK activity in neuronal processes underlying higher brain functions. In this context, RSK acts as a modulator of ERK signaling.


Assuntos
Transporte Axonal , Axônios/enzimologia , Síndrome de Coffin-Lowry/enzimologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neurônios Motores/enzimologia , Junção Neuromuscular/enzimologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transmissão Sináptica , Animais , Axônios/patologia , Síndrome de Coffin-Lowry/genética , Síndrome de Coffin-Lowry/patologia , Modelos Animais de Doenças , Regulação para Baixo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Potenciais Pós-Sinápticos Excitadores , Predisposição Genética para Doença , Potenciais Pós-Sinápticos em Miniatura , Mitocôndrias/enzimologia , Neurônios Motores/patologia , Junção Neuromuscular/patologia , Plasticidade Neuronal , Fenótipo , Terminações Pré-Sinápticas/enzimologia , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Fatores de Tempo
3.
J Neurochem ; 119(3): 447-59, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21838783

RESUMO

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.


Assuntos
Síndrome de Coffin-Lowry/genética , Modelos Animais de Doenças , Hipocampo/enzimologia , Sistema de Sinalização das MAP Quinases/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/deficiência , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Animais , Células Cultivadas , Síndrome de Coffin-Lowry/enzimologia , Transtornos Cognitivos/enzimologia , Transtornos Cognitivos/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Mutação , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia
4.
Cell Mol Neurobiol ; 30(8): 1401-6, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21061166

RESUMO

Coffin-Lowry syndrome (CLS) is a syndromic form of X-linked mental retardation, characterized in male patients by psychomotor and growth retardation and various skeletal anomalies. CLS is caused by mutations in the RPS6KA3 gene, which encodes RSK2, a growth factor-regulated protein kinase. Cognitive deficiencies in CLS patients are prominent, but markedly variable in severity, even between siblings. However, the vast majority of patients are severely affected, with mental retardation ranging from moderate to profound. We used a RSK2-KO mouse model that shows no obvious brain abnormalities at the anatomical and histological levels to study the function of RSK2 in neurosecretion. Behavioral studies revealed normal motor coordination, but a profound retardation in spatial learning and a deficit in long-term spatial memory, providing evidence that RSK2 plays similar roles in mental functioning both in mice and human. We found that associative LTP at cortical inputs to the lateral amygdala was blocked in Rsk2 KO mice. Using an RNA interference rescue strategy in PC12 cells, we were able to demonstrate that RSK2 regulates catecholamine release through the phosphorylation of PLD. These results provide the first molecular evidence that RSK2 could regulate neurotransmitter release by activating PLD production of lipids required for exocytosis.


Assuntos
Síndrome de Coffin-Lowry/enzimologia , Neurossecreção , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Exocitose , Humanos , Modelos Biológicos , Terminações Pré-Sinápticas/enzimologia
5.
Dev Biol ; 347(2): 348-59, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20832397

RESUMO

Coffin-Lowry Syndrome (CLS) is an X-linked genetic disorder associated with cognitive and behavioural impairments. CLS patients present with loss-of-function mutations in the RPS6KA3 gene encoding the mitogen-activated protein kinase (MAPK)-activated kinase p90 ribosomal S6 kinase 2 (Rsk2). Although Rsk2 is expressed in the embryonic brain, its function remains largely uncharacterized. To this end, we isolated murine cortical precursors at embryonic day 12 (E12), a timepoint when neuronal differentiation is initiated, and knocked-down Rsk2 expression levels using shRNA. We performed similar experiments in vivo using in utero electroporations to express shRNA against Rsk2. Rsk2 knockdown resulted in a significant decrease in neurogenesis and an increase in the proportion of proliferating Pax6-positive radial precursor cells, indicating that Rsk2 is essential for cortical radial precursors to differentiate into neurons. In contrast, reducing Rsk2 levels in vitro or in vivo had no effect on the generation of astrocytes. Thus, Rsk2 loss-of-function, as seen in CLS, perturbs the differentiation of neural precursors into neurons, and maintains them instead as proliferating radial precursor cells, a defect that may underlie the cognitive dysfunction seen in CLS.


Assuntos
Síndrome de Coffin-Lowry/etiologia , Neurogênese/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Animais , Sequência de Bases , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/enzimologia , Síndrome de Coffin-Lowry/embriologia , Síndrome de Coffin-Lowry/enzimologia , Síndrome de Coffin-Lowry/genética , Modelos Animais de Doenças , Células-Tronco Embrionárias , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Neurogênese/genética , Gravidez , RNA Interferente Pequeno/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 90-kDa/genética
6.
Ann N Y Acad Sci ; 1152: 201-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19161391

RESUMO

Together with the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins, fusogenic cone-shaped lipids, such as phosphatidic acid (PA), have been recently shown to be important actors in membrane fusion during exocytosis. Phospholipase D (PLD) appears to be the main provider of PA at the exocytotic site in neuroendocrine cells. We show here that ribosomal S6 kinase 2 (RSK2) stimulates PLD activity through the phosphorylation of Thr147 in the PLD1 amino-terminal Phox-homology domain. In PC12 cells, depletion of RSK2 dramatically prevents PA synthesis at exocytotic sites and inhibits hormone release. 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.


Assuntos
Síndrome de Coffin-Lowry/enzimologia , Exocitose , Células Neuroendócrinas/enzimologia , Células Neuroendócrinas/metabolismo , Fosfolipase D/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Exocitose/efeitos dos fármacos , Células PC12 , Ácidos Fosfatídicos/biossíntese , Fosfolipase D/genética , Inibidores de Proteínas Quinases/farmacologia , Ratos , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 90-kDa/genética
7.
Proc Natl Acad Sci U S A ; 105(24): 8434-9, 2008 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-18550821

RESUMO

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.


Assuntos
Cálcio/metabolismo , Células Cromafins/metabolismo , Síndrome de Coffin-Lowry/enzimologia , Exocitose , Fosfolipase D/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Células Cromafins/enzimologia , Síndrome de Coffin-Lowry/genética , Exocitose/genética , Células PC12 , Ácidos Fosfatídicos/metabolismo , Fosforilação , Ratos , Proteínas Quinases S6 Ribossômicas 90-kDa/genética
8.
Hum Genet ; 122(5): 541-3, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17717706

RESUMO

Heterogeneous mutations in the X-linked gene RPS6KA3, encoding the protein kinase RSK2, are responsible for Coffin-Lowry Syndrome. Here we have further studied a male patient with a highly suggestive clinical diagnosis of CLS but in whom no mutation was found by exon sequencing. Western blot analysis revealed a protein much larger than the normal expected size. Sequencing of the RSK2 cDNA, showed the presence of an in-frame tandem duplication of exons 17-20. The mutated RSK2 protein was found to be inactive in an in-vitro kinase assay. This event, which was the result of a homologous unequal recombination between Alu sequences, is the first reported large duplication of the RPS6KA3 gene. Our finding provides further evidence that immunoblot analysis, or a molecular assay capable to detect large genomic mutational events, is essential for patients with a highly suggestive CLS clinical diagnosis but remaining without mutation after exon sequencing.


Assuntos
Síndrome de Coffin-Lowry/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , Síndrome de Coffin-Lowry/enzimologia , DNA/genética , Éxons , Duplicação Gênica , Humanos , Masculino , RNA Mensageiro/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/química , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Sequências de Repetição em Tandem
9.
PLoS One ; 2(1): e151, 2007 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-17225856

RESUMO

The transcriptional activity of the integrated HIV provirus is dependent on the chromatin organization of the viral promoter and the transactivator Tat. Tat recruits the cellular pTEFb complex and interacts with several chromatin-modifying enzymes, including the histone acetyltransferases p300 and PCAF. Here, we examined the interaction of Tat with activation-dependent histone kinases, including the p90 ribosomal S6 kinase 2 (RSK2). Dominant-negative RSK2 and treatment with a small-molecule inhibitor of RSK2 kinase activity inhibited the transcriptional activity of Tat, indicating that RSK2 is important for Tat function. Reconstitution of RSK2 in cells from subjects with a genetic defect in RSK2 expression (Coffin-Lowry syndrome) enhanced Tat transactivation. Tat interacted with RSK2 and activated RSK2 kinase activity in cells. Both properties were lost in a mutant Tat protein (F38A) that is deficient in HIV transactivation. Our data identify a novel reciprocal regulation of Tat and RSK2 function, which might serve to induce early changes in the chromatin organization of the HIV LTR.


Assuntos
Produtos do Gene tat/metabolismo , HIV-1/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Linhagem Celular , Síndrome de Coffin-Lowry/enzimologia , Síndrome de Coffin-Lowry/genética , Produtos do Gene tat/genética , Infecções por HIV/genética , HIV-1/genética , Histonas/metabolismo , Humanos , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Sequências Repetidas Terminais/genética , Ativação Transcricional , Replicação Viral/genética
10.
Nucleic Acids Res ; 32(3): 1214-23, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14973203

RESUMO

Coffin-Lowry syndrome (CLS) is caused by mutations in the RSK2 gene encoding a protein kinase of the Ras signalling pathway. We have studied two point mutations which cause aberrant splicing but do not concern the invariant GT or AG nucleotides of splice sites. The first, an A-->G transition at position +3 of the 5' splice site of exon 6, results in vivo and in vitro in exon skipping and premature translation termination. The natural 5' splice site, although intrinsically weak, is not transactivated under normal conditions. Consequently, replacement of an A/U by a G/U base pairing with U1 snRNA reduces its strength below a critical threshold. The second mutation, an A-->G transition 11 nt upstream of exon 5, creates a new AG near the natural 3' splice site. In vitro this synthetic 3' AG is used exclusively by the splicing machinery. In vivo this splicing event is also observed, but is underestimated because the resulting RSK2 mRNA contains premature stop codons which trigger the nonsense-mediated decay process. We show that a particular mechanism is involved in the aberrant splicing of exon 5, implying involvement of the natural 3' AG during the first catalytic step and the new 3' AG during the second step. Thus, our results explain how these mutations cause severe forms of CLS.


Assuntos
Síndrome de Coffin-Lowry/genética , Íntrons , Mutação Puntual , Splicing de RNA , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Sequência de Bases , Linhagem Celular , Síndrome de Coffin-Lowry/enzimologia , Células HeLa , Humanos , Sítios de Splice de RNA , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA