Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Brain Dev ; 42(2): 192-198, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31706665

ABSTRACT

BACKGROUND: Coffin-Siris syndrome (CSS) is a neurodevelopmental disorder characterized by somatic dysmorphic features, developmental and speech delay. It is due to mutations in many different genes, belonging to BAF chromatin-remodelling complex. The last gene involved in this complex, recently individuated and related to CSS, was DPF2, although only nine patients have been reported until now. METHOD: Here we report on a boy with a history of developmental delay, especially regarding speech and language, and dysmorphic features resembling a syndromic condition. Array-Comparative Genomic Hybridization (CGH) and a custom Next Generation Sequencing (NGS) panel including developmental delay related genes were executed. RESULTS: Array-CGH was negative while NGS panel revealed a novel mutation in DPF2 gene. CONCLUSIONS: We add the clinical description of another patient with a novel mutation in DPF2, with a mild phenotype, thus trying to contribute to enlarge CSS phenotypic variability. Moreover, we briefly discuss about cohesinopathies and major differential diagnosis among syndromes with phenotypes overlapping to CSS.


Subject(s)
Coffin-Lowry Syndrome/genetics , DNA-Binding Proteins/genetics , Mutation, Missense , Transcription Factors/genetics , Child , Coffin-Lowry Syndrome/diagnosis , Coffin-Lowry Syndrome/metabolism , Comparative Genomic Hybridization/methods , DNA-Binding Proteins/metabolism , Developmental Disabilities/diagnosis , Developmental Disabilities/genetics , Developmental Disabilities/metabolism , Diagnosis, Differential , Epigenesis, Genetic , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Mutation/genetics , Phenotype , Transcription Factors/metabolism
2.
Gene ; 575(1): 42-7, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26297997

ABSTRACT

Coffin-Lowry syndrome (CLS) is an X-linked semi-dominant disorder characterized by diverse phenotypes including intellectual disability, facial and digital anomalies. Loss-of-function mutations in the Ribosomal Protein S6 Kinase Polypeptide 3 (RPS6KA3) gene have been shown to be responsible for CLS. Among the large number of mutations, however, no exonic mutation causing exon skipping has been described. Here, we report a male patient with CLS having a novel mutation at the 3' end of an exon at a splice donor junction. Interestingly, this nucleotide change causes both a novel missense mutation and partial exon skipping leading to a truncated transcript. These two transcripts were identified by cDNA sequencing of RT-PCR products. In the carrier mother, we found only wildtype transcripts suggesting skewed X-inactivation. Methylation studies confirmed X-inactivation was skewed moderately, but not completely, which is consistent with her mild phenotype. Western blot showed that the mutant RSK2 protein in the patient is expressed at similar levels relative to his mother. Protein modeling demonstrated that the missense mutation is damaging and may alter binding to ATP molecules. This is the first report of exon skipping from an exonic mutation of RPS6KA3, demonstrating that a missense mutation and concomitant disruption of normal splicing contribute to the manifestation of CLS.


Subject(s)
Coffin-Lowry Syndrome/genetics , Exons , Mutation, Missense , RNA Splice Sites , RNA Splicing , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Coffin-Lowry Syndrome/metabolism , Female , Humans , Male , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
4.
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
5.
Cell Mol Life Sci ; 70(9): 1543-73, 2013 May.
Article in English | MEDLINE | ID: mdl-23474979

ABSTRACT

Epigenetic mechanisms play an important role in gene regulation during development. DNA methylation, which is probably the most important and best-studied epigenetic mechanism, can be abnormally regulated in common pathologies, but the origin of altered DNA methylation remains unknown. Recent research suggests that these epigenetic alterations could depend, at least in part, on genetic mutations or polymorphisms in DNA methyltransferases and certain genes encoding enzymes of the one-carbon metabolism pathway. Indeed, the de novo methyltransferase 3B (DNMT3B) has been recently found to be mutated in several types of cancer and in the immunodeficiency, centromeric region instability and facial anomalies syndrome (ICF), in which these mutations could be related to the loss of global DNA methylation. In addition, mutations in glycine-N-methyltransferase (GNMT) could be associated with a higher risk of hepatocellular carcinoma and liver disease due to an unbalanced S-adenosylmethionine (SAM)/S-adenosylhomocysteine (SAH) ratio, which leads to aberrant methylation reactions. Also, genetic variants of chromatin remodeling proteins and histone tail modifiers are involved in genetic disorders like α thalassemia X-linked mental retardation syndrome, CHARGE syndrome, Cockayne syndrome, Rett syndrome, systemic lupus erythematous, Rubinstein-Taybi syndrome, Coffin-Lowry syndrome, Sotos syndrome, and facioescapulohumeral syndrome, among others. Here, we review the potential genetic alterations with a possible role on epigenetic factors and discuss their contribution to human disease.


Subject(s)
DNA/genetics , Epigenesis, Genetic , Mutation , Animals , CHARGE Syndrome/genetics , CHARGE Syndrome/metabolism , Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , Coffin-Lowry Syndrome/genetics , Coffin-Lowry Syndrome/metabolism , DNA/metabolism , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Rett Syndrome/genetics , Rett Syndrome/metabolism , Rubinstein-Taybi Syndrome/genetics , Rubinstein-Taybi Syndrome/metabolism , Sotos Syndrome/genetics , Sotos Syndrome/metabolism , alpha-Thalassemia/genetics , alpha-Thalassemia/metabolism
6.
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
7.
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
8.
J Neurochem ; 103(1): 388-99, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17645456

ABSTRACT

Ribosomal S6 kinase2 (RSK2) is known to take part in several signal transduction cascades including Mitogen Activated Protein Kinase/Extracellular Regulated Kinase (MAPK/ERK). Following our recent observation that ERK can serve as a coincidence detector for fast and slow neurotransmission in the hippocampus, we analyzed the status of RSK2 phosphorylation subsequent to application of NMDA, dopamine, or both to preparations of mature hippocampal slices in Sprague-Dawley rats. RSK2 was indeed phosphorylated; however, in addition, the amount of RSK2 protein (60%) was induced within 10 min following stimulation. Moreover, the induced expression of RSK2 could be detected in both the cell body layer and the dendrites of hippocampal CA1 cells. Pharmacological analysis showed that RSK2 induction was MAPK ERK Kinase (MEK)-ERK independent, but mammalian Target of Rapamycin (mTOR) and translation dependent. We suggest that the fast kinetics of RSK2 translation that follows physiological stimulations, together with recent observations that its over-expression is vital for the attenuation of major signal transduction cascades, indicate an expanded physiological function of RSK2 in neurons, and sheds new light on the role of RSK2 in the Coffin-Lowry syndrome.


Subject(s)
Coffin-Lowry Syndrome/metabolism , Dopamine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , N-Methylaspartate/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/biosynthesis , Animals , Enzyme Induction/drug effects , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Organ Culture Techniques , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases
9.
Cell Metab ; 4(6): 419-20, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17141624
10.
Cell Metab ; 4(6): 441-51, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17141628

ABSTRACT

The transcription factor ATF4 enhances bone formation by favoring amino acid import and collagen synthesis in osteoblasts, a function requiring its phosphorylation by RSK2, the kinase inactivated in Coffin-Lowry Syndrome. Here, we show that in contrast, RSK2 activity, ATF4-dependent collagen synthesis, and bone formation are increased in mice lacking neurofibromin in osteoblasts (Nf1(ob)(-/-) mice). Independently of RSK2, ATF4 phosphorylation by PKA is enhanced in Nf1(ob)(-/-) mice, thereby increasing Rankl expression, osteoclast differentiation, and bone resorption. In agreement with ATF4 function in amino acid transport, a low-protein diet decreased bone protein synthesis and normalized bone formation and bone mass in Nf1(ob)(-/-) mice without affecting other organ weight, while a high-protein diet overcame Atf4(-/-) and Rsk2(-/-) mice developmental defects, perinatal lethality, and low bone mass. By showing that ATF4-dependent skeletal dysplasiae are treatable by dietary manipulations, this study reveals a molecular connection between nutrition and skeletal development.


Subject(s)
Activating Transcription Factor 4/metabolism , Bone Diseases, Developmental/diet therapy , Bone Diseases, Developmental/metabolism , Dietary Proteins/therapeutic use , Neurofibromin 1/metabolism , Osteoblasts/metabolism , Amino Acids/metabolism , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/genetics , Bone Diseases, Developmental/congenital , Bone Diseases, Developmental/pathology , Bone Resorption/diet therapy , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Coffin-Lowry Syndrome/genetics , Coffin-Lowry Syndrome/metabolism , Coffin-Lowry Syndrome/pathology , Collagen/biosynthesis , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Mice , Mice, Knockout , Neurofibromin 1/deficiency , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/genetics , RANK Ligand/biosynthesis , RANK Ligand/genetics , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
11.
Mol Interv ; 2(6): 376-91, 339, 2002 Oct.
Article in English | MEDLINE | ID: mdl-14993414

ABSTRACT

Our understanding of the molecular underpinnings of human cognition has been greatly aided by the convergent synergy of clinical, genetic, and signaling research. By identifying the mutated genes that give rise to syndromes of mental retardation or cognitive defects in patients, and by placing the associated gene products within signaling networks, researchers are piecing together how learning occurs and how memories are formed and sustained.


Subject(s)
Cognition/physiology , Genetics, Medical , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Animals , Coffin-Lowry Syndrome/genetics , Coffin-Lowry Syndrome/metabolism , Dendrites/pathology , Down Syndrome/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Humans , Memory/physiology , Mice , Mutation , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Neurofibromatoses/genetics , Neurofibromatoses/metabolism , Rett Syndrome/genetics , Rett Syndrome/metabolism , Rubinstein-Taybi Syndrome/genetics , Rubinstein-Taybi Syndrome/metabolism , Williams Syndrome/genetics , Williams Syndrome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...