Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 1037, 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38310100

ABSTRACT

Liver failure causes breakdown of the Blood CNS Barrier (BCB) leading to damages of the Central-Nervous-System (CNS), however the mechanisms whereby the liver influences BCB-integrity remain elusive. One possibility is that the liver secretes an as-yet to be identified molecule(s) that circulate in the serum to directly promote BCB-integrity. To study BCB-integrity, we developed light-sheet imaging for three-dimensional analysis. We show that liver- or muscle-specific knockout of Hfe2/Rgmc induces BCB-breakdown, leading to accumulation of toxic-blood-derived fibrinogen in the brain, lower cortical neuron numbers, and behavioral deficits in mice. Soluble HFE2 competes with its homologue RGMa for binding to Neogenin, thereby blocking RGMa-induced downregulation of PDGF-B and Claudin-5 in endothelial cells, triggering BCB-disruption. HFE2 administration in female mice with experimental autoimmune encephalomyelitis, a model for multiple sclerosis, prevented paralysis and immune cell infiltration by inhibiting RGMa-mediated BCB alteration. This study has implications for the pathogenesis and potential treatment of diseases associated with BCB-dysfunction.


Subject(s)
Blood-Brain Barrier , Encephalomyelitis, Autoimmune, Experimental , Animals , Female , Mice , Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Endothelial Cells/metabolism , Liver/metabolism , Muscles/metabolism
2.
Int J Mol Sci ; 25(3)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38338800

ABSTRACT

Degenerative retinal diseases associated with photoreceptor loss are a leading cause of visual impairment worldwide, with limited treatment options. Phenotypic profiling coupled with medicinal chemistry were used to develop a small molecule with proliferative effects on retinal stem/progenitor cells, as assessed in vitro in a neurosphere assay and in vivo by measuring Msx1-positive ciliary body cell proliferation. The compound was identified as having kinase inhibitory activity and was subjected to cellular pathway analysis in non-retinal human primary cell systems. When tested in a disease-relevant murine model of adult retinal degeneration (MNU-induced retinal degeneration), we observed that four repeat intravitreal injections of the compound improved the thickness of the outer nuclear layer along with the regeneration of the visual function, as measured with ERG, visual acuity, and contrast sensitivity tests. This serves as a proof of concept for the use of a small molecule to promote endogenous regeneration in the eye.


Subject(s)
Retinal Degeneration , Humans , Mice , Animals , Retinal Degeneration/metabolism , Methylnitrosourea , Retina/metabolism , Photoreceptor Cells , Regeneration , Disease Models, Animal , Mammals
4.
Cell Rep ; 42(8): 112925, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37552601

ABSTRACT

The neddylation inhibitor MLN4924/Pevonedistat is in clinical trials for multiple cancers. Efficacy is generally attributed to cullin RING ligase (CRL) inhibition, but the contribution of non-CRL targets is unknown. Here, CRISPR screens map MLN4924-monotherapy sensitivity in retinoblastoma to a classic DNA damage-induced p53/E2F3/BAX-dependent death effector network, which synergizes with Nutlin3a or Navitoclax. In monotherapy-resistant cells, MLN4924 plus standard-of-care topotecan overcomes resistance, but reduces DNA damage, instead harnessing ribosomal protein nucleolar-expulsion to engage an RPL11/p21/MYCN/E2F3/p53/BAX synergy network that exhibits extensive cross-regulation. Strikingly, unneddylatable RPL11 substitutes for MLN4924 to perturb nucleolar function and enhance topotecan efficacy. Orthotopic tumors exhibit complete responses while preserving visual function. Moreover, MLN4924 plus melphalan deploy this DNA damage-independent strategy to synergistically kill multiple myeloma cells. Thus, MLN4924 synergizes with standard-of-care drugs to unlock a nucleolar death effector network across cancer types implying broad therapeutic relevance.


Subject(s)
Topotecan , Tumor Suppressor Protein p53 , bcl-2-Associated X Protein , Cell Line, Tumor , Cyclopentanes/pharmacology , Ribosomal Proteins , Apoptosis , NEDD8 Protein
5.
Int J Dev Biol ; 67(2): 49-56, 2023.
Article in English | MEDLINE | ID: mdl-37410671

ABSTRACT

The gene KIAA0319-Like (KIAA0319L) is thought to confer susceptibility for developmental dyslexia. Dyslexia may be caused by alterations in neuronal migration, and in utero knockdown of KIAA0319L in rats indicated migration errors. However, studies carried out with KIAA0319L knockout mice did not reveal an altered neuronal migration phenotype. Gene knockout may activate compensatory mechanisms to buffer against genetic mutations during development. Here we assessed the role of KIAA0319L on migrating neurons in the chick developing tectum. Whole mount in situ hybridization was performed for KIAA0319L on embryonic day (E)3 - E5 chick embryos and in situ hybridization on sections was performed at later stages. The specificity and efficiency of engineered microRNA (miRNA) constructs targeting KIAA0319L for knocking down KIAA0319L were verified. miRNAs were electroporated into E5 chick optic tecta. Our studies demonstrate that KIAA0319L is expressed in the developing chick visual system, as well as in the otic vesicles. Knockdown of KIAA0319L in the optic tectum results in abnormal neuronal migration, strengthening the argument that KIAA0319L is involved in this developmental process.


Subject(s)
Dyslexia , Chick Embryo , Mice , Animals , Rats , Dyslexia/genetics , Neurons/physiology , Neurogenesis/physiology , Mice, Knockout
6.
Neurobiol Dis ; 150: 105259, 2021 03.
Article in English | MEDLINE | ID: mdl-33434618

ABSTRACT

Neuronal regeneration in the injured central nervous system is hampered by multiple extracellular proteins. These proteins exert their inhibitory action through interactions with receptors that are located in cholesterol rich compartments of the membrane termed lipid rafts. Here we show that cholesterol-synthesis inhibition prevents the association of the Neogenin receptor with lipid rafts. Furthermore, we show that cholesterol-synthesis inhibition enhances axonal growth both on inhibitory -myelin and -RGMa substrates. Following optic nerve injury, lowering cholesterol synthesis with both drugs and siRNA-strategies allows for robust axonal regeneration and promotes neuronal survival. Cholesterol inhibition also enhanced photoreceptor survival in a model of Retinitis Pigmentosa. Our data reveal that Lovastatin leads to several opposing effects on regenerating axons: cholesterol synthesis inhibition promotes regeneration whereas altered prenylation impairs regeneration. We also show that the lactone prodrug form of lovastatin has differing effects on regeneration when compared to the ring-open hydroxy-acid form. Thus the association of cell surface receptors with lipid rafts contributes to axonal regeneration inhibition, and blocking cholesterol synthesis provides a potential therapeutic approach to promote neuronal regeneration and survival in the diseased Central Nervous System. SIGNIFICANCE STATEMENT: Statins have been intensively used to treat high levels of cholesterol in humans. However, the effect of cholesterol inhibition in both the healthy and the diseased brain remains controversial. In particular, it is unclear whether cholesterol inhibition with statins can promote regeneration and survival following injuries. Here we show that late stage cholesterol inhibition promotes robust axonal regeneration following optic nerve injury. We identified distinct mechanisms of action for activated vs non-activated Lovastatin that may account for discrepancies found in the literature. We show that late stage cholesterol synthesis inhibition alters Neogenin association with lipid rafts, thereby i) neutralizing the inhibitory function of its ligand and ii) offering a novel opportunity to promote CNS regeneration and survival following injuries.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Nerve Regeneration/drug effects , Neurons/drug effects , Optic Nerve/drug effects , Animals , Anticholesteremic Agents/pharmacology , Axons/drug effects , Axons/pathology , Cell Survival , Chick Embryo , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Myelin Sheath , Neurons/metabolism , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Photoreceptor Cells , Prenylation , Prodrugs , Rats , Retina , Retinitis Pigmentosa , trans-1,4-Bis(2-chlorobenzaminomethyl)cyclohexane Dihydrochloride/pharmacology
7.
Sci Rep ; 10(1): 21269, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33277529

ABSTRACT

Leber congenital amaurosis (LCA), a form of autosomal recessive severe early-onset retinal degeneration, is an important cause of childhood blindness. This may be associated with systemic features or not. Here we identified COG5 compound-heterozygous variants in patients affected with a complex LCA phenotype associated with microcephaly and skeletal dysplasia. COG5 is a component of the COG complex, which facilitates retrograde Golgi trafficking; if disrupted this can result in protein misfolding. To date, variants in COG5 have been associated with a distinct congenital disorder of glycosylation (type IIi) and with a variant of Friedreich's ataxia. We show that COG5 variants can also result in fragmentation of the Golgi apparatus and upregulation of the UPR modulator, PKR-like endoplasmic reticulum kinase (PERK). In addition, upregulation of PERK induces DNA damage in cultured cells and in murine retina. This study identifies a novel role for COG5 in maintaining ER protein homeostasis and that disruption of that role results in activation of PERK and early-onset retinal degeneration, microcephaly and skeletal dysplasia. These results also highlight the importance of the UPR pathway in early-onset retinal dystrophy and as potential therapeutic targets for patients.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Leber Congenital Amaurosis/genetics , eIF-2 Kinase/metabolism , Bone Diseases, Developmental/genetics , DNA Damage , Endoplasmic Reticulum Stress , Female , Humans , Leber Congenital Amaurosis/metabolism , Male , Microcephaly/genetics , Retina/metabolism , Retinal Degeneration/genetics , Unfolded Protein Response , Whole Genome Sequencing
8.
Dev Growth Differ ; 62(6): 391-397, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32279322

ABSTRACT

Current models of axon guidance within the central nervous system (CNS) involve the presentation of environmental cues to navigating growth cones. The surrounding and target tissues present a variety of ligands that either restrict or promote growth, thus providing pathfinding instructions to developing axons. Recent findings show that RGMb, a GPI anchored extracellular protein present on retinal ganglion cells, down-regulates Wnt3a signaling by lowering LRP5 levels at the membrane surface. When RGMb is phosphorylated by the extracellular tyrosine kinase VLK, phosphorylated RGMb (p-RGMb) is internalized and carries LRP5 towards intracellular compartments. In the eye, a dorsal-high ventral-low gradient of VLK generates a dorsal-low ventral-high gradient of LRP5 that modulates Wnt3a signaling. These molecules, which are all expressed by individual RGCs, generate Wnt-signal gradients along the dorso-ventral axis of the retina, resulting in differential axon growth which in turn regulates proper retino-tectal/collicular map formation. This pathway represents a regulatory mechanism whereby extracellular phosphorylation generates what may be the first example of a unique self-guiding mechanism that affects neuronal-target connections independent of paracrine signals from the surrounding target tissue.


Subject(s)
Axon Guidance , Retinal Ganglion Cells/metabolism , Animals , Humans , Phosphorylation
9.
J Clin Invest ; 130(4): 2054-2068, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32175920

ABSTRACT

Inherited retinal degenerations (IRDs) are characterized by the progressive loss of photoreceptors and represent one of the most prevalent causes of blindness among working-age populations. Cyclic nucleotide dysregulation is a common pathological feature linked to numerous forms of IRD, yet the precise mechanisms through which this contributes to photoreceptor death remain elusive. Here we demonstrate that cAMP induced upregulation of the dependence receptor neogenin in the retina. Neogenin levels were also elevated in both human and murine degenerating photoreceptors. We found that overexpressing neogenin in mouse photoreceptors was sufficient to induce cell death, whereas silencing neogenin in degenerating murine photoreceptors promoted survival, thus identifying a pro-death signal in IRDs. A possible treatment strategy is modeled whereby peptide neutralization of neogenin in Rd1, Rd10, and Rho P23H-knockin mice promotes rod and cone survival and rescues visual function as measured by light-evoked retinal ganglion cell recordings, scotopic/photopic electroretinogram recordings, and visual acuity tests. These results expose neogenin as a critical link between cAMP and photoreceptor death, and identify a druggable target for the treatment of retinal degeneration.


Subject(s)
Membrane Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Animals , Cell Line, Tumor , Cyclic AMP/genetics , Cyclic AMP/metabolism , Disease Models, Animal , Gene Knock-In Techniques , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred ICR , Mice, Transgenic , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Ganglion Cells/pathology
10.
Adv Exp Med Biol ; 1185: 301-304, 2019.
Article in English | MEDLINE | ID: mdl-31884628

ABSTRACT

Inherited retinal degenerations (IRDs) are a genetically heterogeneous group of disorders characterized by the progressive loss of photoreceptor cells. Despite this heterogeneity in the disease-causing mutation, common underlying mechanisms promoting photoreceptor cell death may be present. Dysregulation of photoreceptor cyclic nucleotide signaling may be one such common feature differentiating healthy from diseased photoreceptors. Here we review evidence that elevated retinal cAMP levels promote photoreceptor death and are a common feature of numerous animal models of IRDs. Improving our understanding of how cAMP levels become elevated and identifying downstream effectors may prove important for the development of therapeutics that will be applicable to multiple forms of the disease.


Subject(s)
Cell Death , Cyclic AMP/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/pathology , Animals , Disease Models, Animal , Retina/metabolism , Signal Transduction
11.
Nat Chem Biol ; 15(11): 1035-1042, 2019 11.
Article in English | MEDLINE | ID: mdl-31451763

ABSTRACT

Until recently, the existence of extracellular kinase activity was questioned. Many proteins of the central nervous system are targeted, but it remains unknown whether, or how, extracellular phosphorylation influences brain development. Here we show that the tyrosine kinase vertebrate lonesome kinase (VLK), which is secreted by projecting retinal ganglion cells, phosphorylates the extracellular protein repulsive guidance molecule b (RGMb) in a dorsal-ventral descending gradient. Silencing of VLK or RGMb causes aberrant axonal branching and severe axon misguidance in the chick optic tectum. Mice harboring RGMb with a point mutation in the phosphorylation site also display aberrant axonal pathfinding. Mechanistic analyses show that VLK-mediated RGMb phosphorylation modulates Wnt3a activity by regulating LRP5 protein gradients. Thus, the secretion of VLK by projecting neurons provides crucial signals for the accurate formation of nervous system circuitry. The dramatic effect of VLK on RGMb and Wnt3a signaling implies that extracellular phosphorylation likely has broad and profound effects on brain development, function and disease.


Subject(s)
Axon Guidance , Axons/metabolism , Animals , Mice , Nerve Tissue Proteins/metabolism , Phosphorylation
12.
Iran J Basic Med Sci ; 21(10): 1004-1012, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30524673

ABSTRACT

OBJECTIVES: The beneficial outcomes of bone marrow-derived mesenchymal stem cell (BMSC) treatment on functional recovery following stroke has been well established. Furthermore, 5-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors have also been shown to increase neuronal survival and promote the movement of BMSCs towards the sites of inflammation. However, the precise mechanisms mediating the improved neurological functional recovery in stoke models following a combination treatment of Simvastatin and BMSCs still remained poorly understood. MATERIALS AND METHODS: Here, an embolic stroke model was used to experimentally induce a focal ischemic brain injury by inserting a preformed clot into the middle cerebral artery (MCA). Following stroke, animals were treated either with an intraperitoneal injection of Simvastatin, an intravenous injection of 3 ×106 BMSCs, or a combination of these two treatments. RESULTS: Seven days after ischemia, the combination of Simvastatin and BMSCs led to a significant increase in BMSC relocation, endogenous neurogenesis, arteriogenesis and astrocyte activation while also reducing neuronal damage when compared to BMSC treatment alone (P<0.001 for all). In addition, based on western blot analysis, following stroke there was a significant decrease in c-Fos expression (P<0.001) in the combination treatment group. CONCLUSION: These results further demonstrate the synergistic benefits of a combination treatment and help to improve our understanding of the underlying mechanisms mediating this beneficial effect.

13.
Cell Death Differ ; 25(8): 1503-1516, 2018 08.
Article in English | MEDLINE | ID: mdl-29396549

ABSTRACT

In response to stroke, astrocytes become reactive astrogliosis and are a major component of a glial scar. This results in the formation of both a physical and chemical (production of chondroitin sulfate proteoglycans) barrier, which prevent neurite regeneration that, in turn, interferes with functional recovery. However, the mechanisms of reactive astrogliosis and glial scar formation are poorly understood. In this work, we hypothesized that repulsive guidance molecule a (RGMa) regulate reactive astrogliosis and glial scar formation. We first found that RGMa was strongly expressed by reactive astrocytes in the glial scar in a rat model of middle cerebral artery occlusion/reperfusion. Genetic or pharmacologic inhibition of RGMa in vivo resulted in a strong reduction of reactive astrogliosis and glial scarring as well as in a pronounced improvement in functional recovery. Furthermore, we showed that transforming growth factor ß1 (TGFß1) stimulated RGMa expression through TGFß1 receptor activin-like kinase 5 (ALK5) in primary cultured astrocytes. Knockdown of RGMa abrogated key steps of reactive astrogliosis and glial scar formation induced by TGFß1, including cellular hypertrophy, glial fibrillary acidic protein upregulation, cell migration, and CSPGs secretion. Finally, we demonstrated that RGMa co-immunoprecipitated with ALK5 and Smad2/3. TGFß1-induced ALK5-Smad2/3 interaction and subsequent phosphorylation of Smad2/3 were impaired by RGMa knockdown. Taken together, we identified that after stroke, RGMa promotes reactive astrogliosis and glial scar formation by forming a complex with ALK5 and Smad2/3 to promote ALK5-Smad2/3 interaction to facilitate TGFß1/Smad2/3 signaling, thereby inhibiting neurological functional recovery. RGMa may be a new therapeutic target for stroke.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroglia/pathology , Signal Transduction/drug effects , Stroke/pathology , Transforming Growth Factor beta1/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Cicatrix/pathology , Disease Models, Animal , Fibronectin Type III Domain/genetics , GPI-Linked Proteins , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Stroke/metabolism
14.
Cell Rep ; 20(1): 99-111, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28683327

ABSTRACT

Developing strategies that promote axonal regeneration within the injured CNS is a major therapeutic challenge, as axonal outgrowth is potently inhibited by myelin and the glial scar. Although regeneration can be achieved using the genetic deletion of PTEN, a negative regulator of the mTOR pathway, this requires inactivation prior to nerve injury, thus precluding therapeutic application. Here, we show that, remarkably, fibroblast-derived exosomes (FD exosomes) enable neurite growth on CNS inhibitory proteins. Moreover, we demonstrate that, upon treatment with FD exosomes, Wnt10b is recruited toward lipid rafts and activates mTOR via GSK3ß and TSC2. Application of FD exosomes shortly after optic nerve injury promoted robust axonal regeneration, which was strongly reduced in Wnt10b-deleted animals. This work uncovers an intercellular signaling pathway whereby FD exosomes mobilize an autocrine Wnt10b-mTOR pathway, thereby awakening the intrinsic capacity of neurons for regeneration, an important step toward healing the injured CNS.


Subject(s)
Autocrine Communication , Axons/metabolism , Exosomes/metabolism , Nerve Regeneration , Optic Nerve Injuries/metabolism , Wnt Proteins/metabolism , Animals , Axons/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Fibroblasts/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Membrane Microdomains/metabolism , Mice , Optic Nerve/metabolism , Optic Nerve/physiology , PC12 Cells , Rats , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism , Wnt Proteins/genetics
15.
Clin Exp Pharmacol Physiol ; 42(11): 1158-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26218989

ABSTRACT

Despite extensive research over the years, there still exists some debate as to what constitutes the optimal therapeutic strategy to promote recovery following stroke. Due to the complexity of injured brain pathophysiology, treatment approaches should ideally address numerous factors, ultimately aiming to promote tissue protection, axonal regrowth and functional recovery. This study extends the understanding of the effects of bone marrow stromal cell (BMSC) treatment following experimentally induced ischemic stroke in rats. Focal ischemic brain injury was experimentally induced in rats by placing a preformed clot into the middle cerebral artery. Animals were injected intravenously with BMSCs at 24 h after stroke and were killed 7 days post injury. When administered BMSCs following stroke, the neurological outcome was significantly improved relative to controls. There was an increase in the number of BMSCs labelled with BrdU present in the injured hemisphere of the brain compared to the non-injured side. Furthermore, administration of BMSCs also led to increases in astrocytosis, vascularization and endogenous proliferation. These findings provide insight into the mechanisms of action of BMSC treatment and further argue for the therapeutic potential of BMSCs as an effective treatment following cerebral stroke.


Subject(s)
Brain/pathology , Cell Proliferation , Infarction, Middle Cerebral Artery/surgery , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Neurogenesis , Animals , Arterioles/pathology , Arterioles/physiopathology , Astrocytes/pathology , Behavior, Animal , Biomarkers/metabolism , Brain/metabolism , Brain/physiopathology , Cells, Cultured , Disease Models, Animal , Gliosis , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Mesenchymal Stem Cells/metabolism , Motor Activity , Neovascularization, Physiologic , Neurons/pathology , Phenotype , Rats, Wistar , Recovery of Function , Time Factors
16.
Mol Cell Neurosci ; 51(3-4): 101-11, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22940085

ABSTRACT

Regulation of tissue size is a poorly understood process. Mammalian Staufen 2 (Stau2) is a double-stranded mRNA binding protein known to regulate dendrite formation in vitro as well as cell survival and migration in vivo. Three Stau2 isoforms have been identified in the brain of mammals. Here we show that all these Stau2 isoforms are also expressed in the developing eye of chicken embryos. Strikingly, ectopic expression of Stau2 was sufficient to increase eye size, suggesting a novel biological role of Stau2 in eye morphogenesis. Moreover, down regulation of Stau2 in vivo resulted in a small eye. Microphthalmia was not associated with either increased cell death or differentiation but with reduced cell proliferation. Rescue experiments showed that all three Stau2 isoforms present in the developing eye could prevent microphthalmia. Finally, we showed that Stau2 silencing decreased HES-1 and Sox-2 in the developing eye. These data highlight a new biological function for Stau2 and suggest that translation control of specific Stau2-associated transcripts may be a key regulator of tissue size.


Subject(s)
Eye/growth & development , RNA-Binding Proteins/metabolism , Animals , Cell Proliferation , Chick Embryo , Down-Regulation , Eye/embryology , Eye/metabolism , Gene Expression Regulation, Developmental , Gene Silencing , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Microphthalmos/genetics , Microphthalmos/metabolism , Organ Size , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering , RNA-Binding Proteins/genetics , Rats , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription, Genetic
17.
Dev Cell ; 22(2): 391-402, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22340500

ABSTRACT

The nervous system is enormously complex, yet the number of cues that control axonal growth is surprisingly meager. Posttranslational modifications amplify diversity, but the degree to which they are employed is unclear. Here, we show that Furin and SKI-1 combine with autocatalytic cleavage and a disulfide bridge to generate four membrane-bound and three soluble forms of the repulsive guidance molecule (RGMa). We provide in vivo evidence that these proprotein convertases are involved in axonal growth and that RGMa cleavage is essential for Neogenin-mediated outgrowth inhibition. Surprisingly, despite no sequence homology, N- and C-RGMa fragments bound the same Fibronectin-like domains in Neogenin and blocked outgrowth. This represents an example in which unrelated fragments from one molecule inhibit outgrowth through a single receptor domain. RGMa is a tethered membrane-bound molecule, and proteolytic processing amplifies RGMa diversity by creating soluble versions with long-range effects as well.


Subject(s)
Axons/physiology , DNA-Binding Proteins/metabolism , Furin/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cloning, Molecular , DNA-Binding Proteins/genetics , Furin/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Silencing , Immunoprecipitation , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics
18.
J Neurosci ; 29(4): 1126-31, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19176821

ABSTRACT

Antibodies are powerful tools for delineating the specific function of protein domains, yet several limitations restrict their in vivo applicability. Here we present a new method to obtain sustained in vivo inhibition of specific protein domains using recombinant antibodies. We show that long term in vivo expression of single-chain Fv (scFv) fragments in the developing CNS can be achieved through retroviral transduction. Moreover, specific scFvs generated against the N- and C-terminal domains of the repulsive guidance molecule, RGMa, prevent proper axon targeting in the visual system. This work reveals a previously unappreciated role for the RGMa N-terminal domain in axon guidance, and provides a novel, broadly applicable and rapid procedure to functionally antagonize any protein domain in vivo.


Subject(s)
Antibodies/chemistry , Axons/drug effects , Central Nervous System/cytology , Immunoglobulin Variable Region/pharmacology , Neurons/cytology , Protein Structure, Tertiary/physiology , Animals , Antibodies/pharmacology , Central Nervous System/embryology , Chick Embryo , Ephrin-A5/chemistry , Ephrin-A5/immunology , Ephrin-A5/metabolism , Neurons/drug effects , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...