Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Cell Rep ; 40(13): 111413, 2022 09 27.
Article En | MEDLINE | ID: mdl-36170840

Efficient myelination supports nerve conduction and axonal health throughout life. In the central nervous system, oligodendrocytes (OLs) carry out this demanding anabolic duty in part through biosynthetic pathways controlled by mTOR. We identify Ral GTPases as critical regulators of mouse spinal cord myelination and myelin maintenance. Ablation of Ral GTPases (RalA, RalB) in OL-lineage cells impairs timely onset and radial growth of developmental myelination, accompanied by increased endosomal/lysosomal abundance. Further examinations, including transcriptomic analyses of Ral-deficient OLs, were consistent with mTORC1-related deficits. However, deletion of the mTOR signaling-repressor Pten in Ral-deficient OL-lineage cells is unable to rescue mTORC1 activation or developmental myelination deficiencies. Induced deletion of Ral GTPases in OLs of adult mice results in late-onset myelination defects and tissue degeneration. Together, our data indicate critical roles for Ral GTPases to promote developmental spinal cord myelination, to ensure accurate mTORC1 signaling, and to protect the healthy state of myelin-axon units over time.


Monomeric GTP-Binding Proteins , ral GTP-Binding Proteins , Animals , Homeostasis , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Monomeric GTP-Binding Proteins/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , TOR Serine-Threonine Kinases/metabolism , ral GTP-Binding Proteins/metabolism
2.
J Cell Biol ; 218(7): 2350-2369, 2019 07 01.
Article En | MEDLINE | ID: mdl-31201267

Small GTPases of the Rho and Ras families are important regulators of Schwann cell biology. The Ras-like GTPases RalA and RalB act downstream of Ras in malignant peripheral nerve sheath tumors. However, the physiological role of Ral proteins in Schwann cell development is unknown. Using transgenic mice with ablation of one or both Ral genes, we report that Ral GTPases are crucial for axonal radial sorting. While lack of only one Ral GTPase was dispensable for early peripheral nerve development, ablation of both RalA and RalB resulted in persistent radial sorting defects, associated with hallmarks of deficits in Schwann cell process formation and maintenance. In agreement, ex vivo-cultured Ral-deficient Schwann cells were impaired in process extension and the formation of lamellipodia. Our data indicate further that RalA contributes to Schwann cell process extensions through the exocyst complex, a known effector of Ral GTPases, consistent with an exocyst-mediated function of Ral GTPases in Schwann cells.


Peripheral Nervous System/growth & development , Schwann Cells/metabolism , ral GTP-Binding Proteins/genetics , Animals , Axons/metabolism , Cell Movement/genetics , Cells, Cultured , Exocytosis/genetics , GTP Phosphohydrolases/genetics , Humans , Mice , Mice, Transgenic , Peripheral Nervous System/metabolism , Signal Transduction/genetics
3.
Oncotarget ; 9(45): 27760-27772, 2018 Jun 12.
Article En | MEDLINE | ID: mdl-29963235

INTRODUCTION: Clinical application of antiangiogenic therapy lacks direct visualization of therapy efficacy and vascular resistance. We aimed to establish molecular imaging during treatment with sunitinib using the fibronectin extradomain A specific small immunoprotein(SIP)-F8 in glioma. METHODS: Biodistribution analysis of F8-SIP-Alexa-555 was performed in SF126-glioma bearing or control mice (n = 23 and 7, respectively). Intravital microscopy(IVM) was performed on a microvascular level after 7 days (n = 5 per group) and subsequently after 6 days of sunitinib treatment (n = 4) or without (n = 2).Additionally, near infrared fluorescence(NIRF) imaging was established with F8-SIP-Alexa-750 allowing non-invasive imaging with and without antiangiogenic treatment in orthotopic tumors (n = 38 divided in 4 groups). MRI was used to determine tumor size and served as a reference for NIRF imaging. RESULTS: F8-SIP demonstrated a time and hemodynamic dependent tumor specific accumulation. A significantly higher vascular accumulation occurred with antiangiogenic treatment compared to untreated tumors enabling visualization of resistant tumor vessels by F8-SIP-mediated NIRF imaging. In orthotopic tumors, sunitinib reduced F8-SIP-Alexa-750 enrichment volume but not fluorescence intensity indicative of F8-SIP accumulation in fewer vessels. CONCLUSION: F8-SIP is highly tumor specific with time and hemodynamic dependent biodistribution. The higher vascular accumulation to remaining vessels enables molecular imaging and targeting of therapy resistant tumor vessels.

4.
J Neurosci ; 36(31): 8132-48, 2016 08 03.
Article En | MEDLINE | ID: mdl-27488634

UNLABELLED: The aim of this study was to explore the signaling and neuroprotective effect of transactivator of transcription (TAT) protein transduction of the apoptosis repressor with CARD (ARC) in in vitro and in vivo models of cerebral ischemia in mice. In mice, transient focal cerebral ischemia reduced endogenous ARC protein in neurons in the ischemic striatum at early reperfusion time points, and in primary neuronal cultures, RNA interference resulted in greater neuronal susceptibility to oxygen glucose deprivation (OGD). TAT.ARC protein delivery led to a dose-dependent better survival after OGD. Infarct sizes 72 h after 60 min middle cerebral artery occlusion (MCAo) were on average 30 ± 8% (mean ± SD; p = 0.005; T2-weighted MRI) smaller in TAT.ARC-treated mice (1 µg intraventricularly during MCAo) compared with controls. TAT.ARC-treated mice showed better performance in the pole test compared with TAT.ß-Gal-treated controls. Importantly, post-stroke treatment (3 h after MCAo) was still effective in affording reduced lesion volume by 20 ± 7% (mean ± SD; p < 0.05) and better functional outcome compared with controls. Delayed treatment in mice subjected to 30 min MCAo led to sustained neuroprotection and functional behavior benefits for at least 28 d. Functionally, TAT.ARC treatment inhibited DAXX-ASK1-JNK signaling in the ischemic brain. ARC interacts with DAXX in a CARD-dependent manner to block DAXX trafficking and ASK1-JNK activation. Our work identifies for the first time ARC-DAXX binding to block ASK1-JNK activation as an ARC-specific endogenous mechanism that interferes with neuronal cell death and ischemic brain injury. Delayed delivery of TAT.ARC may present a promising target for stroke therapy. SIGNIFICANCE STATEMENT: Up to now, the only successful pharmacological target of human ischemic stroke is thrombolysis. Neuroprotective pharmacological strategies are needed to accompany therapies aiming to achieve reperfusion. We describe that apoptosis repressor with CARD (ARC) interacts and inhibits DAXX and proximal signals of cell death. In a murine stroke model mimicking human malignant infarction in the territory of the middle cerebral artery, TAT.ARC salvages brain tissue when given during occlusion or 3 h delayed with sustained functional benefits (28 d). This is a promising novel therapeutic approach because it appears to be effective in a model producing severe injury by interfering with an array of proximal signals and effectors of the ischemic cascade, upstream of JNK, caspases, and BIM and BAX activation.


Apoptosis , Brain Ischemia/metabolism , Brain Ischemia/pathology , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Gene Products, tat/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Co-Repressor Proteins , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones , Protein Binding , Protein Interaction Maps
5.
J Cereb Blood Flow Metab ; 33(5): e1-9, 2013 May.
Article En | MEDLINE | ID: mdl-23443176

Intravenous administration of iron oxide nanoparticles during the acute stage of experimental stroke can produce signal intensity changes in the ischemic region. This has been attributed, albeit controversially, to the infiltration of iron-laden blood-borne macrophages. The properties of nanoparticles that render them most suitable for phagocytosis is a matter of debate, as is the most relevant timepoint for administration. Both of these questions are examined in the present study. Imaging experiments were performed in mice with 30 minutes of middle cerebral artery occlusion (MCAO). Iron oxide nanoparticles with different charges and sizes were used, and mice received 300 µmol Fe/kg intravenously: either superparamagnetic iron oxide nanoparticles (SPIOs), ultrasmall SPIOs, or very small SPIOs. The particles were administered 7 days before MCAO, at the time of reperfusion, or 72 hours after MCAO. Interestingly, there was no observable signal change in the ischemic brains that could be attributed to iron. Furthermore, no Prussian blue-positive cells were found in the brains or blood leukocytes, despite intense staining in the livers and spleens. This implies that the nanoparticles selected for this study are not phagocytosed by blood-borne leukocytes and do not enter the ischemic mouse brain.


Brain/blood supply , Brain/pathology , Contrast Media , Infarction, Middle Cerebral Artery/pathology , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Animals , Contrast Media/chemistry , Leukocytes/cytology , Leukocytes/pathology , Magnetite Nanoparticles/chemistry , Male , Mice , Mice, Inbred C57BL , Particle Size , Phagocytosis
6.
J Cereb Blood Flow Metab ; 31(11): 2152-9, 2011 Nov.
Article En | MEDLINE | ID: mdl-21863037

Small ubiquitin-like modifier (SUMO)2/3 but not SUMO1 conjugation is activated after transient cerebral ischemia. To investigate its function, we blocked neuronal SUMO2/3 translation through lentiviral microRNA delivery in primary cortical neurons. Viability was unaffected by SUMO2/3 silencing unless neurons were stressed by transient oxygen-glucose deprivation (OGD). Both 15 and 45 minutes of OGD were tolerated by control microRNA-expressing neurons but damaged >60% of neurons expressing SUMO2/3 microRNA. Damaging OGD (75 minutes) increased neuronal loss to 54% (control microRNA) and to 99% (SUMO2/3 microRNA). This suggests that activation of SUMO2/3 conjugation is an endogenous neuroprotective stress response.


Ischemic Attack, Transient/prevention & control , Neurons/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Culture Techniques , Cell Hypoxia , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Genetic Vectors , Glucose/metabolism , Immunohistochemistry , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Lentivirus/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/administration & dosage , MicroRNAs/genetics , MicroRNAs/pharmacology , Neurons/pathology , Oxygen/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation/drug effects , Sumoylation/genetics , Ubiquitins/genetics
...