Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Neuropathol ; 138(2): 275-293, 2019 08.
Article in English | MEDLINE | ID: mdl-31062076

ABSTRACT

Glioblastomas strongly invade the brain by infiltrating into the white matter along myelinated nerve fiber tracts even though the myelin protein Nogo-A prevents cell migration by activating inhibitory RhoA signaling. The mechanisms behind this long-known phenomenon remained elusive so far, precluding a targeted therapeutic intervention. This study demonstrates that the prevalent activation of AKT in gliomas increases the ER protein-folding capacity and enables tumor cells to utilize a side effect of RhoA activation: the perturbation of the IRE1α-mediated decay of SPARC mRNA. Once translation is initiated, glioblastoma cells rapidly secrete SPARC to block Nogo-A from inhibiting migration via RhoA. By advanced ultramicroscopy for studying single-cell invasion in whole, undissected mouse brains, we show that gliomas require SPARC for invading into white matter structures. SPARC depletion reduces tumor dissemination that significantly prolongs survival and improves response to cytostatic therapy. Our finding of a novel RhoA-IRE1 axis provides a druggable target for interfering with SPARC production and underscores its therapeutic value.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplasm Proteins/physiology , Nogo Proteins/biosynthesis , Osteonectin/biosynthesis , Protein Biosynthesis , White Matter/pathology , rhoA GTP-Binding Protein/physiology , Animals , Binding, Competitive , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasm Invasiveness , Nogo Proteins/genetics , Osteonectin/genetics , Protein Domains , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Sphingosine-1-Phosphate Receptors/physiology , Tumor Cells, Cultured , White Matter/metabolism
2.
Dev Cell ; 43(1): 24-34.e5, 2017 10 09.
Article in English | MEDLINE | ID: mdl-28943240

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) critically modulate adhesion-, growth-, and migration-related processes. Here, we show that the transmembrane protein, Nogo-A, inhibits neurite outgrowth and cell spreading in neurons and Nogo-A-responsive cell lines via HSPGs. The extracellular, active 180 amino acid Nogo-A region, named Nogo-A-Δ20, binds to heparin and brain-derived heparan sulfate glycosaminoglycans (GAGs) but not to the closely related chondroitin sulfate GAGs. HSPGs are required for Nogo-A-Δ20-induced inhibition of adhesion, cell spreading, and neurite outgrowth, as well as for RhoA activation. Surprisingly, we show that Nogo-A-Δ20 can act via HSPGs independently of its receptor, Sphingosine-1-Phosphate receptor 2 (S1PR2). We thereby identify the HSPG family members syndecan-3 and syndecan-4 as functional receptors for Nogo-A-Δ20. Finally, we show in explant cultures ex vivo that Nogo-A-Δ20 promotes the migration of neuroblasts via HSPGs but not S1PR2.


Subject(s)
Cell Movement/physiology , Cell Shape/physiology , Heparan Sulfate Proteoglycans/metabolism , Neurites/metabolism , Neuronal Outgrowth/physiology , Nogo Proteins/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Heparitin Sulfate/metabolism , Mice , Protein Binding , Proteoglycans/metabolism , Receptors, Lysosphingolipid/metabolism
3.
PLoS One ; 9(1): e86820, 2014.
Article in English | MEDLINE | ID: mdl-24489789

ABSTRACT

BACKGROUND: The protein Nogo-A regulates axon growth in the developing and mature nervous system, and this is carried out by two distinct domains in the protein, Nogo-A-Δ20 and Nogo-66. The differences in the signalling pathways engaged in axon growth cones by these domains are not well characterized, and have been investigated in this study. METHODOLOGY/PRINCIPAL FINDINGS: We analyzed growth cone collapse induced by the Nogo-A domains Nogo-A-Δ20 and Nogo-66 using explanted chick dorsal root ganglion neurons growing on laminin/poly-lysine substratum. Collapse induced by purified Nogo-A-Δ20 peptide is dependent on protein synthesis whereas that induced by Nogo-66 peptide is not. Nogo-A-Δ20-induced collapse is accompanied by a protein synthesis-dependent rise in RhoA expression in the growth cone, but is unaffected by proteasomal catalytic site inhibition. Conversely Nogo-66-induced collapse is inhibited ∼ 50% by proteasomal catalytic site inhibition. CONCLUSION/SIGNIFICANCE: Growth cone collapse induced by the Nogo-A domains Nogo-A-Δ20 and Nogo-66 is mediated by signalling pathways with distinguishable characteristics concerning their dependence on protein synthesis and proteasomal function.


Subject(s)
Ganglia, Spinal/metabolism , Growth Cones/metabolism , Myelin Proteins/genetics , Myelin Proteins/pharmacology , Protein Biosynthesis/drug effects , Animals , Anisomycin/pharmacology , Chick Embryo , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Gene Expression Regulation, Developmental , Growth Cones/drug effects , Growth Cones/pathology , Laminin , Leupeptins/pharmacology , Myelin Proteins/metabolism , Nogo Proteins , Polylysine , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Protein Structure, Tertiary , Protein Synthesis Inhibitors/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , Tissue Culture Techniques , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
4.
Neuroscientist ; 20(4): 372-386, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24402613

ABSTRACT

Initially discovered as a potent neurite outgrowth inhibitor in the central nervous system (CNS), Nogo-A has emerged as a multifunctional protein. Involvement of this protein has been demonstrated in numerous developmental processes, ranging from cell migration, axon guidance and fasciculation, dendritic branching and CNS plasticity to oligodendrocyte differentiation and myelination. Although initially necessary and beneficial for shaping and later maintaining CNS structure and functionality, the growth restricting properties of Nogo-A can have negative effects on nervous system injury or disease. Hence, correlating with its various neurobiological roles, Nogo-A was implicated in a range of CNS disturbances, including trauma such as spinal cord injury or stroke, neurodegenerative diseases such as Alzheimer's disease, amyotrophic lateral sclerosis or multiple sclerosis, or in schizophrenia. In this review, we summarize the current state of knowledge for Nogo-A's involvement in these nervous system diseases and perturbations and discuss the possible underlying mechanisms. Furthermore, we provide a comprehensive overview on molecular signaling pathways as well as structural properties identified for Nogo-A and point to open questions in the field.

5.
PLoS One ; 8(10): e78212, 2013.
Article in English | MEDLINE | ID: mdl-24205161

ABSTRACT

BACKGROUND: Adhesion dependent mechanisms are increasingly recognized to be important for a wide range of biological processes, diseases and therapeutics. This has led to a rising demand of pharmaceutical modulators. However, most currently available adhesion assays are time consuming and/or lack sensitivity and reproducibility or depend on specialized and expensive equipment often only available at screening facilities. Thus, rapid and economical high-content screening approaches are urgently needed. RESULTS: We established a fully open source high-content screening method for identifying modulators of adhesion. We successfully used this method to detect small molecules that are able to influence cell adhesion and cell spreading of Swiss-3T3 fibroblasts in general and/or specifically counteract Nogo-A-Δ20-induced inhibition of adhesion and cell spreading. The tricyclic anti-depressant clomipramine hydrochloride was shown to not only inhibit Nogo-A-Δ20-induced cell spreading inhibition in 3T3 fibroblasts but also to promote growth and counteract neurite outgrowth inhibition in highly purified primary neurons isolated from rat cerebellum. CONCLUSIONS: We have developed and validated a high content screening approach that can be used in any ordinarily equipped cell biology laboratory employing exclusively freely available open-source software in order to find novel modulators of adhesion and cell spreading. The versatility and adjustability of the whole screening method will enable not only centers specialized in high-throughput screens but most importantly also labs not routinely employing screens in their daily work routine to investigate the effects of a wide range of different compounds or siRNAs on adhesion and adhesion-modulating molecules.


Subject(s)
Biological Assay/methods , Cell Adhesion/physiology , Myelin Proteins/metabolism , 3T3 Cells , Animals , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cellular Structures/metabolism , Cellular Structures/physiology , Cerebellum/metabolism , Cerebellum/physiology , Fibroblasts/metabolism , Fibroblasts/physiology , Mice , Neurites/metabolism , Neurites/physiology , Nogo Proteins , Rats , Reproducibility of Results , Sensitivity and Specificity
7.
J Cell Biol ; 193(7): 1147-56, 2011 Jun 27.
Article in English | MEDLINE | ID: mdl-21690307

ABSTRACT

After an acute central nervous system injury, axonal regeneration is limited as the result of a lack of neuronal intrinsic competence and the presence of extrinsic inhibitory signals. The injury fragments the myelin neuronal insulating layer, releasing extrinsic inhibitory molecules to signal through the neuronal membrane-bound Nogo receptor (NgR) complex. In this paper, we show that a neuronal transcriptional pathway can interfere with extrinsic inhibitory myelin-dependent signaling, thereby promoting neurite outgrowth. Specifically, retinoic acid (RA), acting through the RA receptor ß (RAR-ß), inhibited myelin-activated NgR signaling through the transcriptional repression of the NgR complex member Lingo-1. We show that suppression of Lingo-1 was required for RA-RAR-ß to counteract extrinsic inhibition of neurite outgrowth. Furthermore, we confirm in vivo that RA treatment after a dorsal column overhemisection injury inhibited Lingo-1 expression, specifically through RAR-ß. Our findings identify a novel link between RA-RAR-ß-dependent proaxonal outgrowth and inhibitory NgR complex-dependent signaling, potentially allowing for the development of molecular strategies to enhance axonal regeneration after a central nervous system injury.


Subject(s)
Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neurites/metabolism , Receptors, Retinoic Acid/physiology , Tretinoin/physiology , Animals , Cells, Cultured , Gene Expression Regulation , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Myelin Sheath/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Neurites/ultrastructure , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism
8.
J Neurosci ; 29(48): 15266-76, 2009 Dec 02.
Article in English | MEDLINE | ID: mdl-19955379

ABSTRACT

Rho GTPases are thought to mediate the action of several axonal growth inhibitors in the adult brain and spinal cord. RhoA has been targeted pharmacologically in both humans and animals to promote neurite outgrowth and functional recovery following CNS trauma. However, rat spinal cord injury studies suggest a complicated and partial benefit of inhibiting Rho or its downstream effector, Rho-associated kinase (ROCKII). This limited benefit may reflect inhibition of other kinases, poor access, or a minimal role of ROCKII in vivo. Therefore, we studied ROCKII mutant mice to probe this pathway genetically. ROCKII(-/-) dorsal root ganglion neurons are less sensitive to inhibition by Nogo protein or by chondroitin sulfate proteoglycan in vitro. We examined adult ROCKII(-/-) mice in two injury paradigms, cervical multilevel dorsal rhizotomy and midthoracic dorsal spinal cord hemisection. After dorsal root crush injury, the ROCKII(-/-) mice recovered use of the affected forepaw more quickly than did controls. Moreover, multiple classes of sensory axons regenerated across the dorsal root entry zone into the spinal cord of mice lacking ROCKII. After the spinal cord injury, ROCKII(-/-) mice showed enhanced local growth of raphespinal axons in the caudal spinal cord and corticospinal axons into the lesion site. Improved functional recovery was not observed by Basso Mouse Scale score following dorsal hemisection, likely due to developmental defects in the nervous system. Together, these findings demonstrate that the ROCKII gene product limits axonal growth after CNS trauma.


Subject(s)
Axons/pathology , Axons/physiology , Spinal Cord Injuries/pathology , rho-Associated Kinases/physiology , Amides/pharmacology , Analysis of Variance , Animals , Axons/drug effects , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain Injuries/pathology , Brain Injuries/physiopathology , CA1 Region, Hippocampal/cytology , Cells, Cultured , Cholera Toxin/metabolism , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Median Neuropathy/etiology , Median Neuropathy/pathology , Median Neuropathy/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins/pharmacology , Nerve Regeneration/physiology , Neurons/classification , Neurons/drug effects , Neurons/pathology , Nogo Proteins , Pyridines/pharmacology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Rhizotomy/methods , Spinal Cord Injuries/physiopathology , Time Factors , Versicans/pharmacology , rho-Associated Kinases/deficiency
9.
Neuroscientist ; 13(5): 454-69, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17901255

ABSTRACT

Rho-associated protein kinases (ROCKs) play key roles in mediating the control of the actin cytoskeleton by Rho family GTPases in response to extracellular signals. Such signaling pathways contribute to diverse neuronal functions from cell migration to axonal guidance to dendritic spine morphology to axonal regeneration to cell survival. In this review, the authors summarize biochemical knowledge of ROCK function and categorize neuronal ROCK-dependent signaling pathways. Further study of ROCK signal transduction mechanisms and specificities will enhance our understanding of brain development, plasticity, and repair. The ROCK pathway also provides a potential site for therapeutic intervention to promote neuronal regeneration and to limit degeneration.


Subject(s)
Neurons/enzymology , Neurons/physiology , rho GTP-Binding Proteins/physiology , rho-Associated Kinases/physiology , Animals , Cell Adhesion Molecules/physiology , Central Nervous System/enzymology , Central Nervous System/physiology , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Humans , Lipoproteins/physiology , Myelin Proteins/physiology , Nerve Tissue Proteins/physiology , Nogo Proteins , Protein Processing, Post-Translational/genetics , Receptor Protein-Tyrosine Kinases/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Immunologic/physiology , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Roundabout Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...