Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Glia ; 64(12): 2247-2262, 2016 12.
Article in English | MEDLINE | ID: mdl-27658374

ABSTRACT

Axo-glial interactions are critical for myelination and the domain organization of myelinated fibers. Cell adhesion molecules belonging to the Cadm family, and in particular Cadm3 (axonal) and its heterophilic binding partner Cadm4 (Schwann cell), mediate these interactions along the internode. Using targeted shRNA-mediated knockdown, we show that the removal of axonal Cadm3 promotes Schwann cell myelination in the in vitro DRG neuron/Schwann cell myelinating system. Conversely, over-expressing Cadm3 on the surface of DRG neuron axons results in an almost complete inability by Schwann cells to form myelin segments. Axons of superior cervical ganglion (SCG) neurons, which do not normally support the formation of myelin segments by Schwann cells, express higher levels of Cadm3 compared to DRG neurons. Knocking down Cadm3 in SCG neurons promotes myelination. Finally, the extracellular domain of Cadm3 interferes in a dose-dependent manner with the activation of ErbB3 and of the pro-myelinating PI3K/Akt pathway, but does not interfere with the activation of the Mek/Erk1/2 pathway. While not in direct contradiction, these in vitro results shed lights on the apparent lack of phenotype that was reported from in vivo studies of Cadm3-/- mice. Our results suggest that Cadm3 may act as a negative regulator of PNS myelination, potentially through the selective regulation of the signaling cascades activated in Schwann cells by axonal contact, and in particular by type III Nrg-1. Further analyses of peripheral nerves in the Cadm-/- mice will be needed to determine the exact role of axonal Cadm3 in PNS myelination. GLIA 2016;64:2247-2262.


Subject(s)
Cell Adhesion Molecules/deficiency , Gene Expression Regulation/genetics , Immunoglobulins/deficiency , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Schwann Cells/physiology , Signal Transduction/physiology , Animals , Axons/drug effects , Axons/metabolism , Cell Adhesion Molecules/genetics , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Coculture Techniques , Ganglia, Spinal/cytology , Gene Expression Regulation/physiology , Immunoglobulins/genetics , Immunoprecipitation , Myelin Basic Protein/metabolism , Neurites/metabolism , Neurons/drug effects , Neurons/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Schwann Cells/drug effects , Signal Transduction/drug effects
2.
Cancer Res ; 76(18): 5479-90, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27406828

ABSTRACT

AAC-11 is an antiapoptotic protein that is upregulated in most cancer cells. Increased expression of AAC-11 confers a survival advantage when cancer cells are challenged with various stresses and contributes to tumor invasion and metastases, whereas its deregulation reduces resistance to chemotherapeutic drugs. The antiapoptotic effect of AAC-11 may be clinically relevant as its expression correlates with poor prognosis in several human cancers. Thus, inactivation of AAC-11 might constitute an attractive approach for developing cancer therapeutics. We have developed an AAC-11-derived cell-penetrating peptide, herein named RT53, mimicking in part the heptad leucine repeat region of AAC-11, which functions as a protein-protein interaction module, and that can prevent AAC-11 antiapoptotic properties. In this study, we investigated the anticancer effects of RT53. Our results indicate that RT53 selectively kills cancer cells while sparing normal cells. RT53 selectively inserts into the membranes of cancer cells, where it adopts a punctate distribution and induces membranolysis and release of danger-associated molecular pattern molecules. Systemic administration of RT53 inhibited the growth of preexisting BRAF wild-type and V600E mutant melanoma xenograft tumors through induction of apoptosis and necrosis. Toxicological studies revealed that repetitive injections of RT53 did not produce significant toxicity. Finally, RT53-killed B16F10 cells induced tumor growth inhibition in immunocompetent mice following a rechallenge with live cancer cells of the same type. Collectively, our data demonstrate that RT53 possesses tumor-inhibitory activity with no toxicity in mice, suggesting its potential as a therapeutic agent for the treatment of melanoma and probably other cancers. Cancer Res; 76(18); 5479-90. ©2016 AACR.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis/drug effects , Cell-Penetrating Peptides/pharmacology , Melanoma, Experimental/drug therapy , Nuclear Proteins/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Immunoprecipitation , In Situ Nick-End Labeling , Melanoma, Experimental/pathology , Mice , Mice, Nude , Xenograft Model Antitumor Assays
3.
Cell Cycle ; 14(8): 1242-51, 2015.
Article in English | MEDLINE | ID: mdl-25695197

ABSTRACT

Beside its central role in the mitochondria-dependent cell death pathway, the apoptotic protease activating factor 1 (Apaf-1) is involved in the DNA damage response through cell-cycle arrest induced by genotoxic stress. This non-apoptotic function requires a nuclear translocation of Apaf-1 during the G1-to-S transition. However, the mechanisms that trigger the nuclear accumulation of Apaf-1 upon DNA damage remain to be investigated. Here we show that the main 4 isoforms of Apaf-1 can undergo nuclear translocation and restore Apaf-1 deficient MEFs cell cycle arrest in the S phase following genotoxic stress through activation of Chk-1. Interestingly, DNA damage-dependent nuclear accumulation of Apaf-1 occurs independently of p53 and the retinoblastoma (pRb) pathway. We demonstrated that Apaf-1 associates with the nucleoporin Nup107 and this association is necessary for Apaf-1 nuclear import. The CED-4 domain of Apaf-1 directly binds to the central domain of Nup107 in an ATR-regulated, phosphorylation-dependent manner. Interestingly, expression of the Apaf-1-interacting domain of Nup107 interfered with Apaf-1 nuclear translocation upon genotoxic stress, resulting in a marked reduction of Chk-1 activation and cell cycle arrest. Thus, our results confirm the crucial role of Apaf-1 nuclear relocalization in mediating cell-cycle arrest induced by genotoxic stress and implicate Nup107 as a critical regulator of the DNA damage-induced intra-S phase checkpoint response.


Subject(s)
Apoptotic Protease-Activating Factor 1/metabolism , Cell Nucleus/metabolism , DNA Damage , Nuclear Pore Complex Proteins/metabolism , Animals , Apoptotic Protease-Activating Factor 1/deficiency , Apoptotic Protease-Activating Factor 1/genetics , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Checkpoint Kinase 1 , Cisplatin/toxicity , DNA Damage/drug effects , Humans , Mice , Nuclear Pore Complex Proteins/antagonists & inhibitors , Nuclear Pore Complex Proteins/genetics , Phosphorylation/drug effects , Protein Binding , Protein Isoforms/metabolism , Protein Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...