Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
2.
Oncogene ; 37(2): 241-254, 2018 01 11.
Article in English | MEDLINE | ID: mdl-28925399

ABSTRACT

Glioblastomas (GBM) are lethal primitive brain tumours characterized by a strong intra-tumour heterogeneity. We observed in GBM tissues the coexistence of functionally divergent micro-territories either enriched in more differentiated and non-mitotic cells or in mitotic undifferentiated OLIG2 positive cells while sharing similar genomic abnormalities. Understanding the formation of such functionally divergent micro-territories in glioblastomas (GBM) is essential to comprehend GBM biogenesis, plasticity and to develop therapies. Here we report an unexpected anti-proliferative role of beta-catenin in non-mitotic differentiated GBM cells. By cell type specific stimulation of miR-302, which directly represses cyclin D1 and stemness features, beta-catenin is capable to change its known proliferative function. Nuclear beta-catenin accumulation in non-mitotic cells is due to a feed forward mechanism between DOCK4 and beta-catenin, allowed by increased GSK3-beta activity. DOCK4 over expression suppresses selfrenewal and tumorigenicity of GBM stem-like cells. Accordingly in the frame of GBM median of survival, increased level of DOCK4 predicts improved patient survival.


Subject(s)
GTPase-Activating Proteins/metabolism , Glioblastoma/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , beta Catenin/metabolism , Adult , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain/pathology , Cell Nucleus/metabolism , Cell Proliferation , Feedback, Physiological , GTPase-Activating Proteins/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Mice , Mice, Inbred NOD , MicroRNAs/genetics , Mitosis , Neoplastic Stem Cells/cytology , Oligodendrocyte Transcription Factor 2/metabolism , Primary Cell Culture , RNA, Small Interfering/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult , beta Catenin/genetics
3.
Cell Death Dis ; 5: e1023, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24457967

ABSTRACT

Connexin43 (Cx43), the main gap junction channel-forming protein in astrocytes, is downregulated in malignant gliomas. These tumors are composed of a heterogeneous population of cells that include many with stem-cell-like properties, called glioma stem cells (GSCs), which are highly tumorigenic and lack Cx43 expression. Interestingly, restoring Cx43 reverses GSC phenotype and consequently reduces their tumorigenicity. In this study, we investigated the mechanism by which Cx43 exerts its antitumorigenic effects on GSCs. We have focused on the tyrosine kinase c-Src, which interacts with the intracellular carboxy tail of Cx43. We found that Cx43 regulates c-Src activity and proliferation in human GSCs expanded in adherent culture. Thus, restoring Cx43 in GSCs inhibited c-Src activity, which in turn promoted the downregulation of the inhibitor of differentiation Id1. Id1 sustains stem cell phenotype as it controls the expression of Sox2, responsible for stem cell self-renewal, and promotes cadherin switching, which has been associated to epithelial-mesenchymal transition. Our results show that both the ectopic expression of Cx43 and the inhibition of c-Src reduced Id1, Sox2 expression and promoted the switch from N- to E-cadherin, suggesting that Cx43, by inhibiting c-Src, downregulates Id1 with the subsequent changes in stem cell phenotype. On the basis of this mechanism, we found that a cell-penetrating peptide, containing the region of Cx43 that interacts with c-Src, mimics the effect of Cx43 on GSC phenotype, confirming the relevance of the interaction between Cx43 and c-Src in the regulation of the malignant phenotype and pinpointing this interaction as a promising therapeutic target.


Subject(s)
Brain Neoplasms/metabolism , Cell-Penetrating Peptides/metabolism , Connexin 43/metabolism , Glioma/metabolism , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , src-Family Kinases/metabolism , Amino Acid Motifs , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/physiopathology , Cell Differentiation , Connexin 43/chemistry , Connexin 43/genetics , Glioma/enzymology , Glioma/genetics , Glioma/physiopathology , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/enzymology , Phenotype , Protein Binding , Rats , Rats, Wistar , SOXB1 Transcription Factors/genetics , src-Family Kinases/genetics
4.
Cell Death Dis ; 4: e767, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23949216

ABSTRACT

Glioblastomas (GBMs) are devastating tumors of the central nervous system, with a poor prognosis of 1-year survival. This results from a high resistance of GBM tumor cells to current therapeutic options, including etoposide (VP-16). Understanding resistance mechanisms may thus open new therapeutic avenues. VP-16 is a topoisomerase inhibitor that causes replication fork stalling and, ultimately, the formation of DNA double-strand breaks and apoptotic cell death. Autophagy has been identified as a VP-16 treatment resistance mechanism in tumor cells. Retinoblastoma protein (RB) is a classical tumor suppressor owing to its role in G1/S cell cycle checkpoint, but recent data have shown RB participation in many other cellular functions, including, counterintuitively, negative regulation of apoptosis. As GBMs usually display an amplification of the EGFR signaling involving the RB protein pathway, we questioned whether RB might be involved in mechanisms of resistance of GBM cells to VP-16. We observed that RB silencing increased VP-16-induced DNA double-strand breaks and p53 activation. Moreover, RB knockdown increased VP-16-induced apoptosis in GBM cell lines and cancer stem cells, the latter being now recognized essential to resistance to treatments and recurrence. We also showed that VP-16 treatment induced autophagy, and that RB silencing impaired this process by inhibiting the fusion of autophagosomes with lysosomes. Taken together, our data suggest that RB silencing causes a blockage on the VP-16-induced autophagic flux, which is followed by apoptosis in GBM cell lines and in cancer stem cells. Therefore, we show here, for the first time, that RB represents a molecular link between autophagy and apoptosis, and a resistance marker in GBM, a discovery with potential importance for anticancer treatment.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Etoposide/pharmacology , Etoposide/therapeutic use , Glioblastoma/pathology , Retinoblastoma Protein/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/ultrastructure , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , Gene Knockdown Techniques , Glioblastoma/drug therapy , Glioblastoma/ultrastructure , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , RNA Interference/drug effects , Tumor Suppressor Protein p53/metabolism
5.
Cell Death Differ ; 19(2): 232-44, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21720384

ABSTRACT

Glioblastoma multiforme (GBM) is the most common form of primary brain tumor in adults, often characterized by poor survival. Glioma-initiating cells (GiCs) are defined by their extensive self-renewal, differentiation, and tumor initiation properties. GiCs are known to be involved in tumor growth and recurrence, and in resistance to conventional treatments. One strategy to efficiently target GiCs in GBM consists in suppressing their stemness and consequently their tumorigenic properties. In this study, we show that the miR-302-367 cluster is strongly induced during serum-mediated stemness suppression. Stable miR-302-367 cluster expression is sufficient to suppress the stemness signature, self-renewal, and cell infiltration within a host brain tissue, through inhibition of the CXCR4 pathway. Furthermore, inhibition of CXCR4 leads to the disruption of the sonic hedgehog (SHH)-GLI-NANOG network, which is involved in self-renewal and expression of the embryonic stem cell-like signature. In conclusion, we demonstrated that the miR-302-367 cluster is able to efficiently trigger a cascade of inhibitory events leading to the disruption of GiCs stem-like and tumorigenic properties.


Subject(s)
Glioma/genetics , MicroRNAs/genetics , Multigene Family/genetics , Neoplastic Stem Cells/pathology , Receptors, CXCR4/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cell Lineage , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/pathology , Humans , Mice , Neoplastic Stem Cells/metabolism , Receptors, CXCR4/genetics , Serum
6.
Oncogene ; 27(8): 1155-66, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-17700518

ABSTRACT

PEA-15 (phosphoprotein enriched in astrocytes 15 kDa) is a death effector domain-containing protein, which is involved in the regulation of apoptotic cell death. Since PEA-15 is highly expressed in cells of glial origin, we studied the role of PEA-15 in human malignant brain tumors. Immunohistochemical analysis of PEA-15 expression shows strong immunoreactivity in astrocytomas and glioblastomas. Phosphorylation of PEA-15 at Ser(116) is found in vivo in perinecrotic areas in glioblastomas and in vitro after glucose deprivation of glioblastoma cells. Overexpression of PEA-15 induces a marked resistance against glucose deprivation-induced apoptosis, whereas small interfering RNA (siRNA)-mediated downregulation of endogenous PEA-15 results in the sensitization to glucose withdrawal-mediated cell death. This antiapoptotic activity of PEA-15 under low glucose conditions depends on its phosphorylation at Ser(116). Moreover, siRNA-mediated knockdown of PEA-15 abolishes the tumorigenicity of U87MG glioblastoma cells in vivo. PEA-15 regulates the level of phosphorylated extracellular-regulated kinase (ERK)1/2 in glioblastoma cells and the PEA-15-dependent protection from glucose deprivation-induced cell death requires ERK1/2 signaling. PEA-15 transcriptionally upregulates the Glucose Transporter 3, which is abrogated by the inhibition of ERK1/2 phosphorylation. Taken together, our findings suggest that Ser(116)-phosphorylated PEA-15 renders glioma cells resistant to glucose deprivation-mediated cell death as encountered in poor microenvironments, for example in perinecrotic areas of glioblastomas.


Subject(s)
Apoptosis/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Glioblastoma/enzymology , Glucose/deficiency , Intracellular Signaling Peptides and Proteins/physiology , MAP Kinase Signaling System/physiology , Phosphoproteins/physiology , Animals , Apoptosis Regulatory Proteins , Brain Neoplasms/enzymology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Glioblastoma/metabolism , Glioblastoma/pathology , Glucose/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Nude , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphorylation
7.
Oncogene ; 26(19): 2695-706, 2007 Apr 26.
Article in English | MEDLINE | ID: mdl-17057735

ABSTRACT

An instability of the mature cell phenotype is thought to participate to the formation of gliomas, primary brain tumors deriving from astrocytes and/or neural stem cells. Transforming growth factor alpha (TGFalpha) is an erbB1 ligand overexpressed in the earliest stages of gliomas, and exerts trophic effects on gliomal cells and astrocytes. Here, we questioned whether prolonged TGFalpha exposure affects the stability of the normal mature astrocyte phenotype. We first developed astrocyte cultures devoid of residual neural stem cells or progenitors. We demonstrate that days of TGFalpha treatment result in the functional conversion of a population of mature astrocytes into radial glial cells, a population of neural progenitors. TGFalpha-generated radial glial cells support embryonic neurons migration, and give birth to cells of the neuronal lineage, expressing neuronal markers and the electrophysiological properties of neuroblasts. Lengthening TGFalpha treatment to months results in the delayed appearance of cells with neural stem cells properties: they form floating cellular spheres that are self-renewing, can be clonally derived from a single cell and differentiated into cells of the neuronal lineage. This study uncovers a novel population of mature astrocytes capable, in response to a single epigenetic factor, to regress progressively into a neural stem-like cell stage via an intermediate progenitor stage.


Subject(s)
Astrocytes/cytology , Cell Differentiation/drug effects , Neurons/metabolism , Stem Cells/cytology , Transforming Growth Factor alpha/pharmacology , Animals , Astrocytes/metabolism , Cell Lineage , Cell Movement , Cells, Cultured , Electrophysiology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , ErbB Receptors/metabolism , Female , Fetus/cytology , Fetus/metabolism , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Neuroglia/cytology , Neurons/cytology , Receptor, ErbB-2/metabolism , Recombinant Proteins/pharmacology , Stem Cells/metabolism
8.
Oncogene ; 25(29): 4076-85, 2006 Jul 06.
Article in English | MEDLINE | ID: mdl-16532035

ABSTRACT

Astrocyte death has been implicated in several neuropathological diseases, but the identification of molecules susceptible of promoting astrocyte survival has been elusive. We investigated whether transforming growth factor alpha (TGFalpha), an erbB1/EGFR ligand, which promotes glioma progression and affects astrocyte metabolism at embryonic and adult stages, regulates astrocyte survival. Primary serum-free astrocyte cultures from post-natal mouse and fetal human cortices were used. Transforming growth factor alpha protected both species of astrocytes from staurosporine-induced apoptosis. In serum-free medium, mouse astrocytes did not survive beyond 2 months while TGFalpha-treated astrocytes survived up to 12 months. Transforming growth factor alpha also promoted long-term survival of human astrocytes. We additionally extended TGFalpha proliferative effects to human astrocytes. After 3 days of permanent application, TGFalpha induced a major downregulation of both erbB1 and erbB2. This downregulation did not impair the functional activation of the receptors, as ascertained by their tyrosine phosphorylation and the continuous stimulation of both ERK/MAPK and PI3K/Akt pathways up to 7 days, the longest time examined. The full cellular effects of TGFalpha required activation of both transduction pathways. Enhanced proliferation and survival thus define TGFalpha as a gliatrophin for mammalian astrocytes. These results demonstrate that in normal, non-transformed astrocytes, sustained and functional erbBs activation is achieved without bypassing ligand-induced receptors downregulation.


Subject(s)
Astrocytes/metabolism , Down-Regulation/drug effects , ErbB Receptors/metabolism , MAP Kinase Signaling System/drug effects , Receptor, ErbB-2/metabolism , Transforming Growth Factor alpha/pharmacology , Aging/metabolism , Animals , Astrocytes/cytology , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/embryology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma/metabolism , Humans , Mice , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Time Factors , Transforming Growth Factor alpha/metabolism
9.
Neuroscience ; 126(2): 263-75, 2004.
Article in English | MEDLINE | ID: mdl-15207344

ABSTRACT

Phosphoprotein enriched in astrocytes of 15 kDa (PEA-15) is an abundant phosphoprotein in primary cultures of mouse brain astrocytes. Its capability to interact with members of the apoptotic and mitogen activated protein (MAP) kinase cascades endows PEA-15 with anti-apoptotic and anti-proliferative properties. We analyzed the in vivo cellular sources of PEA-15 in the normal adult mouse brain using a novel polyclonal antibody. Immunohistochemical assays revealed numerous PEA-15-immunoreactive cells throughout the brain of wild-type adult mice while no immunoreactive signal was observed in the brain of PEA-15 -/- mice. Cell morphology and double immunofluorescent staining showed that both astrocytes and neurons could be cellular sources of PEA-15. Closer examination revealed that in a given area only part of the astrocytes expressed the protein. The hippocampus was the most striking example of this heterogeneity, a spatial segregation restricting PEA-15 positive astrocytes to the CA1 and CA3 regions. A PEA-15 immunoreactive signal was also observed in a few cells within the subventricular zone and the rostral migratory stream. In vivo analysis of an eventual PEA-15 regulation in astrocytes was performed using a model of astrogliosis occurring along motor neurons degeneration, the transgenic mouse expressing the mutant G93A human superoxyde-dismutase-1, a model of amyotrophic lateral sclerosis. We observed a marked up-regulation of PEA-15 in reactive astrocytes that had developed throughout the ventral horn of the lumbar spinal cord of the transgenic mice. The heterogeneous cellular expression of the protein and its increased expression in pathological situations, combined with the known properties of PEA-15, suggest that PEA-15 expression is associated with a particular metabolic status of cells challenged with potentially apoptotic and/or proliferative signals.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Neurons/metabolism , Phosphoproteins/biosynthesis , 3T3 Cells , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Astrocytes/cytology , Brain/cytology , Cells, Cultured , Female , Gene Expression Regulation/physiology , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Neurons/cytology , Phosphoproteins/immunology
10.
Dev Cell ; 1(2): 239-50, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11702783

ABSTRACT

The ERK 1/2 MAP kinase pathway controls cell growth and survival and modulates integrin function. Here, we report that PEA-15, a protein variably expressed in multiple cell types, blocks ERK-dependent transcription and proliferation by binding ERKs and preventing their localization in the nucleus. PEA-15 contains a nuclear export sequence required for its capacity to anchor ERK in the cytoplasm. Genetic deletion of PEA-15 results in increased ERK nuclear localization with consequent increased cFos transcription and cell proliferation. Thus, PEA-15 can redirect the biological outcome of MAP kinase signaling by regulating the subcellular localization of ERK MAP kinase.


Subject(s)
Cytoplasm/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Phosphoproteins/metabolism , Phosphoproteins/physiology , 3T3 Cells , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Blotting, Northern , CHO Cells , Cell Division , Cell Nucleus/metabolism , Cell Survival , Cricetinae , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Green Fluorescent Proteins , Immunohistochemistry , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Models, Biological , Molecular Sequence Data , Mutation , Phosphoproteins/genetics , Precipitin Tests , Protein Binding , Sequence Homology, Amino Acid , Time Factors , Transcription, Genetic , Transfection , Two-Hybrid System Techniques
12.
Mol Biol Cell ; 11(9): 2863-72, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10982386

ABSTRACT

PEA-15 is a small, death effector-domain (DED)-containing protein that was recently demonstrated to inhibit tumor necrosis factor-alpha-induced apoptosis and to reverse the inhibition of integrin activation due to H-Ras. This led us to investigate the involvement of PEA-15 in Ras signaling. Surprisingly, PEA-15 activates the extracellular signal receptor-activated kinase (ERK) mitogen-activated protein kinase pathway in a Ras-dependent manner. PEA-15 expression in Chinese hamster ovary cells resulted in an increased mitogen-activated protein kinase kinase and ERK activity. Furthermore, PEA-15 expression leads to an increase in Ras guanosine 5'-triphosphate loading. PEA-15 bypasses the anchorage dependence of ERK activation. Finally, the effects of PEA-15 on integrin signaling are separate from those on ERK activation. Heretofore, all known DEDs functioned in the regulation of apoptosis. In contrast, the DED of PEA-15 is essential for its capacity to activate ERK. The ability of PEA-15 to simultaneously inhibit apoptosis and potentiate Ras-to-Erk signaling may be of importance for oncogenic processes.


Subject(s)
MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , Signal Transduction , ras Proteins/metabolism , 3T3 Cells , Animals , Apoptosis Regulatory Proteins , CHO Cells , Cell Adhesion , Cell Line , Cricetinae , Enzyme Activation , Guanosine Triphosphate/metabolism , Humans , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Mice , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , p38 Mitogen-Activated Protein Kinases
13.
J Biol Chem ; 275(45): 35224-32, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-10926929

ABSTRACT

Phospholipase D (PLD), a signal-transducing membrane-associated enzyme, is implicated in diverse processes including apoptosis, ERK activation, and glucose transport. Prior studies have identified specific PLD activators and repressors that directly regulate its enzymatic activity. Using two-hybrid screens, we have identified PEA-15 as a PLD interactor that unexpectedly functions to alter its level of expression. PEA-15 is a widely expressed death effector domain-containing phosphoprotein involved in signal transduction, apoptosis, ERK activation, and glucose transport. The PLD1-interacting site on PEA-15 consists of part of the death effector domain domain plus additional C-terminal flanking sequences, whereas the PEA-15-interacting site on PLD1 overlaps the previously identified RhoA-interacting site. PEA-15 did not affect basal or stimulated in vitro PLD1 enzymatic activation. However, co-expression of PEA-15 increased levels of PLD1 activity. This increased activation correlated with higher PLD1 protein expression levels, as marked by faster accumulation and longer persistence of PLD1 when PEA-15 was present. PEA-15 similarly increased protein expressions level of PLD2 and co-immunoprecipitated with it. These results suggest that PEA-15 may stabilize PLD or act as a PLD chaperone. The common involvement of PEA-15 and PLD in apoptosis, ERK activation, and glucose transport additionally suggests functional significance.


Subject(s)
Gene Expression Regulation, Enzymologic , Phospholipase D/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Alleles , Amino Acid Sequence , Animals , Apoptosis , Apoptosis Regulatory Proteins , Binding Sites , Biological Transport , Blotting, Western , COS Cells , Cell Line , Enzyme Activation , Glucose/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phospholipase D/genetics , Phosphoproteins/genetics , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction , Time Factors , Two-Hybrid System Techniques
14.
J Neurosci ; 19(19): 8244-51, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10493725

ABSTRACT

Apoptosis is a very general phenomenon, but only a few reports concern astrocytes. Indeed, astrocytes express receptors for tumor necrosis factor (TNF) alpha, a cytokine demonstrated on many cells and tissues to mediate apoptosis after recruitment of adaptor proteins containing a death effector domain (DED). PEA-15 is a DED-containing protein prominently expressed in the CNS and particularly abundant in astrocytes. This led us to investigate if PEA-15 expression could be involved in astrocytic protection against deleterious effects of TNF. In vitro assays evidence that PEA-15 may bind to DED-containing protein FADD and caspase-8 known to be apical adaptors of the TNF apoptotic signaling. After generation of PEA-15 null mutant mice, our results demonstrate that PEA-15 expression increases astrocyte survival after exposure to TNF.


Subject(s)
Apoptosis/physiology , Arabidopsis Proteins , Astrocytes/cytology , Astrocytes/physiology , Corpus Striatum/cytology , Phosphoproteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Astrocytes/drug effects , Caspase 8 , Caspase 9 , Caspases/chemistry , Caspases/metabolism , Cells, Cultured , Corpus Striatum/physiology , Embryo, Mammalian , Fatty Acid Desaturases/chemistry , Fatty Acid Desaturases/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Neuroglia/cytology , Neuroglia/physiology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid
15.
Muscle Nerve ; 22(6): 712-7, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10366224

ABSTRACT

Autosomal dominant cerebellar ataxias (ADCAs) are clinically and genetically heterogeneous neurodegenerative disorders. The aim of this study was to evaluate electrophysiologically peripheral nervous system involvement in each of the groups studied and its correlation with the number of CAG repeats. Forty patients with ADCA were clinically and electrophysiologically investigated. Thirty-five patients belonged to the ADCA type I group (SCA1, 12; SCA2, 10; SCA3, 13) and five to the ADCA type II group. Axonal sensory or sensorimotor polyneuropathy was found in 42% of the SCA1 patients, 80% of the SCA2 patients, and 54% of the SCA3 patients, whereas electrophysiological studies were normal in all those with ADCA type II. The number of CAG repeats was significantly higher in SCA1 patients with polyneuropathy than in those without polyneuropathy (P = 0.01), whereas the reverse was observed in SCA3/MJD (Machado-Joseph disease) patients (P = 0.05). We conclude that axonal polyneuropathy is often associated with ADCA type I, but its frequency varies according to factors such as the locus responsible and the number of CAG repeats.


Subject(s)
Cerebellar Ataxia/genetics , Genes, Dominant , Peripheral Nervous System Diseases/genetics , Adolescent , Adult , Female , Genotype , Humans , Male , Middle Aged , Mutation , Phenotype
16.
J Neurochem ; 71(3): 1307-14, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9721757

ABSTRACT

PEA-15 (phosphoprotein enriched in astrocytes, Mr = 15,000) is an acidic serine-phosphorylated protein highly expressed in the CNS, where it can play a protective role against cytokine-induced apoptosis. PEA-15 is a major substrate for protein kinase C. Endothelins, which are known to exert pleiotropic effects on astrocytes, were used to analyze further the processes involved in PEA-15 phosphorylation. Endothelin-1 or endothelin-3 (0.1 microM) induced a robust phosphorylation of PEA-15 that was abolished by the removal of extracellular calcium, but only diminished by inhibitors of protein kinase C. Microsequencing of phosphopeptides generated by digestion of PEA-15 following endothelin-1 treatment identified two phosphorylated residues: Ser104, previously recognized as the protein kinase C site, and a novel phosphoserine, Ser116, located in a consensus motif for either protein kinase casein kinase II or calcium/calmodulin-dependent protein kinase II (CaMKII). Partly purified PEA-15 was a substrate in vitro for CaMKII, but not for casein kinase II. Two-dimensional phosphopeptide mapping demonstrated that the site phosphorylated in vitro by CaMKII was also phosphorylated in intact astrocytes in response to endothelin. CaMKII phosphorylated selectively Ser116 and had no effect on Ser104, but in vitro phosphorylation by CaMKII appeared to facilitate further phosphorylation by protein kinase C. Treatment of intact astrocytes with okadaic acid enhanced the phosphorylation of the CaMKII site. These results demonstrate that PEA-15 is phosphorylated in astrocytes by CaMKII (or a related kinase) and by protein kinase C in response to endothelin.


Subject(s)
Astrocytes/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium/physiology , Endothelins/pharmacology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Casein Kinases , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/drug effects , Enzyme Inhibitors/pharmacology , Mice , Okadaic Acid/pharmacology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphorylation , Protein Kinases/metabolism
17.
Hum Mol Genet ; 6(5): 709-15, 1997 May.
Article in English | MEDLINE | ID: mdl-9158145

ABSTRACT

Spinocerebellar ataxia 2 (SCA2) is caused by the expansion of an unstable CAG repeat encoding a polyglutamine tract. One hundred and eighty four index patients with autosomal dominant cerebellar ataxia type I were screened for this mutation. We found expansion in 109 patients from 30 families of different geographical origins (15%) and in two isolated cases with no known family histories (2%). The SCA2 chromosomes contained from 34 to 57 repeats and consisted of a pure stretch of CAG, whereas all tested normal chromosomes (14-31 repeats), except one with 14 repeats, were interrupted by 1-3 repeats of CAA. As in other diseases caused by unstable mutations, a strong negative correlation was observed between the age at onset and the size of the CAG repeat (r = -0.81). The frequency of several clinical signs such as myoclonus, dystonia and myokymia increased with the number of CAG repeats whereas the frequency of others was related to disease duration. The CAG repeat was highly unstable during transmission with variations ranging from -8 to +12, and a mean increase of +2.2, but there was no significant difference according to the parental sex. This instability was confirmed by the high degree of gonadal mosaicism observed in sperm DNA of one patient.


Subject(s)
Mutation , Proteins/genetics , Spinocerebellar Degenerations/etiology , Trinucleotide Repeats , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Ataxins , Child , Deglutition Disorders/genetics , Dystonia/genetics , Female , Gene Frequency , Gonads/physiology , Humans , Male , Middle Aged , Mosaicism , Nerve Tissue Proteins , Ophthalmoplegia/genetics , Pedigree , Spinocerebellar Degenerations/epidemiology
18.
J Biol Chem ; 271(25): 14800-6, 1996 Jun 21.
Article in English | MEDLINE | ID: mdl-8662970

ABSTRACT

Specific phosphoproteins are targets of numerous extracellular signals received by astrocytes. One such target, which we previously described, is PEA-15, a protein kinase C substrate associated with microtubules. Two cDNAs differing in the length of their 3'-untranslated region (3'UTR) were cloned from a mouse astrocytic library. Accordingly, Northern blots revealed two transcripts (1.7 and 2.5 kilobase pairs) abundant brain regions but also found in peripheral tissues. PEA-15-deduced protein sequence (130 amino acids) shared no similarity with known proteins but is 96% identical to its human counterpart. In addition, several regions of the 3'UTR share more than 90% identity between mouse and human. Different potential regulatory sequences are found in the 3'UTR, which also completely includes the proto-oncogene MAT1. The high level of conservation of both the coding and the untranslated regions and the differential tissular distribution of the two transcripts of this major brain phosphoprotein suggest that not only the protein but also the 3'UTR of PEA-15 mRNA play a role in astrocytic functions.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Phosphoproteins/biosynthesis , RNA, Messenger/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Base Sequence , Blotting, Northern , Cells, Cultured , Cloning, Molecular , Conserved Sequence , Corpus Striatum , DNA Primers , Embryo, Mammalian , Gene Library , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Mice , Microtubules/metabolism , Molecular Sequence Data , Organ Specificity , Phosphoproteins/analysis , Phosphoproteins/chemistry , Polymerase Chain Reaction , Protein Biosynthesis , Proto-Oncogene Mas , RNA, Messenger/chemistry , Recombinant Proteins/biosynthesis
19.
Hum Genet ; 97(5): 671-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8655151

ABSTRACT

Autosomal dominant cerebellar ataxias (ADCAs) are a group of neurodegenerative disorders that are clinically and genetically heterogeneous. We report here a genetic linkage study, with five chromosome 12q markers, of three Martinican families with ADCA type 1, for which the spinocerebellar ataxia 1 (SCA1) locus was excluded. Linkage to the SCA2 locus was demonstrated with a maximal lead score of 6.64 at theta = 0.00 with marker D12S354. Recombinational events observed by haplotype reconstruction demonstrated that the SCA2 locus is located in an approximately 7-cM interval flanked by D12S105 and D12S79. Using the z(max)-1 method, multipoint analysis further reduced the candidate interval for SCA2 to a region of 5 cM. Two families shared a common haplotype at loci spanning 7 cM, which suggests a founder effect, whereas a different haplotype segregated with the disease in the third family. Finally, a mean anticipation of 12+/-14 years was found in parent-child couples, with no parental sex effect, suggesting that the disease might be caused by an expanded and unstable triplet repeat.


Subject(s)
Chromosomes, Human, Pair 12 , Genes, Dominant , Spinocerebellar Degenerations/genetics , Adolescent , Adult , Age of Onset , Child , Chromosome Mapping , Family , Female , Genetic Markers , Genotype , Haplotypes , Humans , Lod Score , Male , Martinique , Middle Aged , Pedigree , Recombination, Genetic , Repetitive Sequences, Nucleic Acid
20.
Ann Neurol ; 39(4): 490-9, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8619527

ABSTRACT

Patients with spinocerebellar ataxia 3 (SCA3) and Machado-Joseph disease (MJD) carry an expanded CAG repeat in the MJD1 gene. One hundred twenty families of different geographic origin with autosomal dominant cerebellar ataxia (ADCA) type I were tested. Thirty-four families (126 patients) carried an expanded CAG repeat. The expanded and the normal allele did not overlap and the repeat was unstable during transmission, with variation in the size of the CAG length ranging from -8 to +5 and a mean expansion of 0.86 repeats without differences according to the parental sex. There was a combined effect of the number of CAG repeats of the expanded and normal allele on the age at onset, which accounted for 70% of its variability. The length of the CAG repeat influenced the frequency of clinical signs associated with cerebellar ataxia, such as abnormal tendon reflexes or decreased vibration sense, whereas the interindividual variation of supranuclear ophthalmoplegia, sphincter and swallowing difficulties, and amyotrophy was mostly determined by different disease durations. We compared the clinical profile of 91 SCA3/MJD patients with 51 SCA1 and 32 SCA2 patients. There were striking differences between the SCA3/MJD and SCA2 but not with SCA1 groups of patients. Despite their clinical similarities, distinct neuropathological features were observed in 2 SCA3/MJD and 2 SCA1 patients.


Subject(s)
Machado-Joseph Disease/genetics , Machado-Joseph Disease/physiopathology , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/physiopathology , Adult , Age of Onset , Central Nervous System/pathology , Female , Gene Frequency , Humans , Machado-Joseph Disease/pathology , Male , Middle Aged , Mutation , Repetitive Sequences, Nucleic Acid , Spinocerebellar Degenerations/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...