Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Neurosci ; 42(20): 4087-4100, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35437280

ABSTRACT

Nogo-66 receptors (NgR1-3) are glycosylphosphatidyl inositol-linked proteins that belong to the leucine-rich repeat superfamily. Through binding to myelin-associated inhibitors, NgRs contribute to the inhibition of axonal regeneration after spinal cord injury. Their role in limiting synaptic plasticity and axonal outgrowth in the adult CNS has been described previously, but not much is known about their role during the development of the nervous system. Here, we show that NgR1 and NgR3 mRNAs are expressed during spinal cord development of the chicken embryo. In particular, they are expressed in the dI1 subpopulation of commissural neurons during the time when their axons navigate toward and across the floorplate, the ventral midline of the spinal cord. To assess a potential role of NgR1 and NgR3 in axon guidance, we downregulated them using in ovo RNAi and analyzed the trajectory of commissural axons by tracing them in open-book preparations of spinal cords. Our results show that loss of either NgR1 or NgR3 causes axons to stall in the midline area and to interfere with the rostral turn of postcrossing axons. In addition, we also show that NgR1, but not NgR3, requires neuronal PlexinA2 for the regulation of commissural axon guidance.SIGNIFICANCE STATEMENT Over the last decades, many studies have focused on the role of NgRs, particularly NgR1, in axonal regeneration in the injured adult CNS. Here, we show a physiological role of NgRs in guiding commissural axons during early development of the chicken spinal cord in vivo Both NgR1 and NgR3 are required for midline crossing and subsequent turning of postcrossing axons into the longitudinal axis of the spinal cord. NgR1, but not NgR3, forms a receptor complex with PlexinA2 during axon guidance. Overall, these findings provide a link between neural regenerative mechanisms and developmental processes.


Subject(s)
Axon Guidance , Receptors, Cell Surface , Animals , Axons/physiology , Chick Embryo , Nogo Receptor 1/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Spinal Cord/metabolism
2.
J Neurosci ; 34(39): 13222-33, 2014 Sep 24.
Article in English | MEDLINE | ID: mdl-25253866

ABSTRACT

After nerve injury, adult sensory neurons can regenerate peripheral axons and reconnect with their target tissue. Initiation of outgrowth, as well as elongation of neurites over long distances, depends on the signaling of receptors for neurotrophic growth factors. Here, we investigated the importance of gp130, the signaling subunit of neuropoietic cytokine receptors in peripheral nerve regeneration. After sciatic nerve crush, functional recovery in vivo was retarded in SNS-gp130(-/-) mice, which specifically lack gp130 in sensory neurons. Correspondingly, a significantly reduced number of free nerve endings was detected in glabrous skin from SNS-gp130(-/-) compared with control mice after nerve crush. Neurite outgrowth and STAT3 activation in vitro were severely reduced in cultures in gp130-deficient cultured neurons. Surprisingly, in neurons obtained from SNS-gp130(-/-) mice the increase in neurite length was reduced not only in response to neuropoietic cytokine ligands of gp130 but also to nerve growth factor (NGF), which does not bind to gp130-containing receptors. Neurite outgrowth in the absence of neurotrophic factors was partially rescued in gp130-deficient neurons by leptin, which activates STAT3 downstream of leptic receptor and independent of gp130. The neurite outgrowth response of gp130-deficient neurons to NGF was fully restored in the presence of leptin. Based on these findings, gp130 signaling via STAT3 activation is suggested not only to be an important regulator of peripheral nerve regeneration in vitro and in vivo, but as determining factor for the growth promoting action of NGF in adult sensory neurons.


Subject(s)
Cytokine Receptor gp130/metabolism , Nerve Regeneration , Neurites/metabolism , STAT3 Transcription Factor/metabolism , Sciatic Nerve/physiology , Sensory Receptor Cells/metabolism , Animals , Cell Growth Processes , Cells, Cultured , Cytokine Receptor gp130/genetics , Leptin/pharmacology , Mice , Mice, Inbred C57BL , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/physiology , Phosphorylation , STAT3 Transcription Factor/genetics , Sciatic Nerve/cytology , Sciatic Nerve/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology
3.
J Neurosci ; 34(5): 1633-46, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24478347

ABSTRACT

Primary sensory afferents of the dorsal root ganglion (DRG) that innervate the skin detect a wide range of stimuli, such as touch, temperature, pain, and itch. Different functional classes of nociceptors project their axons to distinct target zones within the developing skin, but the molecular mechanisms that regulate target innervation are less clear. Here we report that the Nogo66 receptor homolog NgR2 is essential for proper cutaneous innervation. NgR2(-/-) mice display increased density of nonpeptidergic nociceptors in the footpad and exhibit enhanced sensitivity to mechanical force and innocuous cold temperatures. These sensory deficits are not associated with any abnormality in morphology or density of DRG neurons. However, deletion of NgR2 renders nociceptive nonpeptidergic sensory neurons insensitive to the outgrowth repulsive activity of skin-derived Versican. Biochemical evidence shows that NgR2 specifically interacts with the G3 domain of Versican. The data suggest that Versican/NgR2 signaling at the dermo-epidermal junction acts in vivo as a local suppressor of axonal plasticity to control proper density of epidermal sensory fiber innervation. Our findings not only reveal the existence of a novel and unsuspected mechanism regulating epidermal target innervation, but also provide the first evidence for a physiological role of NgR2 in the peripheral nervous system.


Subject(s)
Epidermis/innervation , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/genetics , Receptors, Cell Surface/metabolism , Sensory Receptor Cells/metabolism , Versicans/metabolism , Animals , Animals, Newborn , CHO Cells , Calcitonin Gene-Related Peptide/metabolism , Cricetulus , F-Box Proteins , Glycoproteins/metabolism , Hyperalgesia/physiopathology , Mice , Mice, Knockout , Neurofilament Proteins/metabolism , Nociceptors/metabolism , Nogo Receptor 2 , Pain Threshold/physiology , Physical Stimulation/adverse effects , Protein Binding/genetics , Receptors, Cell Surface/genetics , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/metabolism , Sensory Receptor Cells/classification , Sensory Receptor Cells/cytology , TRPV Cation Channels/metabolism , Tubulin/metabolism , Versicans/chemistry , Versicans/genetics
4.
Biochim Biophys Acta ; 1833(6): 1421-33, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23454728

ABSTRACT

RTN1A is a reticulon protein with predominant localization in the endoplasmic reticulum (ER). It was previously shown that RTN1A is expressed in neurons of the mammalian central nervous system but functional information remains sparse. To elucidate the neuronal function of RTN1A, we chose to focus our investigation on identifying possible novel binding partners specifically interacting with the unique N-terminus of RTN1A. Using a nonbiased approach involving GST pull-downs and MS analysis, we identified the intracellular calcium release channel ryanodine receptor 2 (RyR2) as a direct binding partner of RTN1A. The RyR2 binding site was localized to a highly conserved 150-amino acid residue region. RTN1A displays high preference for RyR2 binding in vitro and in vivo and both proteins colocalize in hippocampal neurons and Purkinje cells. Moreover, we demonstrate the precise subcellular localization of RTN1A in Purkinje cells and show that RTN1A inhibits RyR channels in [(3)H]ryanodine binding studies on brain synaptosomes. In a functional assay, RTN1A significantly reduced RyR2-mediated Ca(2+) oscillations. Thus, RTN1A and RyR2 might act as functional partners in the regulation of cytosolic Ca(2+) dynamics the in neurons.


Subject(s)
Calcium/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Binding Sites , Blotting, Western , Cells, Cultured , Cytosol/metabolism , Hippocampus/cytology , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Neurons/cytology , Protein Binding , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ryanodine/metabolism , Tandem Mass Spectrometry
5.
Biochem J ; 455(2): 217-27, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23909438

ABSTRACT

Nogo-A is the largest isoform of the Nogo/RTN4 (reticulon 4) proteins and has been characterized as a major myelin-associated inhibitor of regenerative nerve growth in the adult CNS (central nervous system). Apart from the myelin sheath, Nogo-A is expressed at high levels in principal neurons of the CNS. The specificity of Nogo-A resides in its central domain, NiG. We identified Apg-1, a member of the stress-induced Hsp110 (heat-shock protein of 110 kDa) family, as a novel interactor of NiG/Nogo-A. The interaction is selective because Apg-1 interacts with Nogo-A/RTN4-A, but not with RTN1-A, the closest paralogue of Nogo-A. Conversely, Nogo-A binds to Apg-1, but not to Apg-2 or Hsp105, two other members of the Hsp110 family. We characterized the Nogo-A-Apg-1 interaction by affinity precipitation, co-immunoprecipitation and proximity ligation assay, using primary hippocampal neurons derived from Nogo-deficient mice. Under conditions of hypoxic and oxidative stress we found that Nogo-A and Apg-1 were tightly co-regulated in hippocampal neurons. Although both proteins were up-regulated under hypoxic conditions, their expression levels were reduced upon the addition of hydrogen peroxide. Taken together, we suggest that Nogo-A is closely involved in the neuronal response to hypoxic and oxidative stress, an observation that may be of relevance not only in stroke-induced ischaemia, but also in neuroblastoma formation.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Myelin Proteins/metabolism , Oxidative Stress , Animals , CHO Cells , Cell Hypoxia/genetics , Cricetulus , Down-Regulation , HSP70 Heat-Shock Proteins/genetics , Hippocampus/metabolism , Mice , Mice, Inbred Strains , Myelin Proteins/genetics , Myelin Sheath/metabolism , Neurons/metabolism , Nogo Proteins
6.
Cell Tissue Res ; 349(1): 105-17, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22311207

ABSTRACT

The Nogo-66 receptor family (NgR) consists in three glycophosphatidylinositol (GPI)-anchored receptors (NgR1, NgR2 and NgR3), which are primarily expressed by neurons in the central and peripheral mammalian nervous system. NgR1 was identified as serving as a high affinity binding protein for the three classical myelin-associated inhibitors (MAIs) Nogo-A, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp), which limit axon regeneration and sprouting in the injured brain. Recent studies suggest that NgR signaling may also play an essential role in the intact adult CNS in restricting axonal and synaptic plasticity and are involved in neurodegenerative diseases, particularly in Alzheimer's disease pathology through modulation of ß-secretase cleavage. Here, we outline the biochemical properties of NgRs and their functional roles in the intact and diseased CNS.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/pathology , Multigene Family , Receptors, Cell Surface/metabolism , Animals , Axons/metabolism , Humans , Neuronal Plasticity/physiology , Signal Transduction
7.
Sci Rep ; 12(1): 18879, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36344556

ABSTRACT

Androgen deprivation therapies aimed to target prostate cancer (PrCa) are only partially successful given the occurrence of neuroendocrine PrCa (NEPrCa), a highly aggressive and highly metastatic form of PrCa, for which there is no effective therapeutic approach. Our group has demonstrated that while absent in prostate adenocarcinoma, the αVß3 integrin expression is increased during PrCa progression toward NEPrCa. Here, we show a novel pathway activated by αVß3 that promotes NE differentiation (NED). This novel pathway requires the expression of a GPI-linked surface molecule, NgR2, also known as Nogo-66 receptor homolog 1. We show here that NgR2 is upregulated by αVß3, to which it associates; we also show that it promotes NED and anchorage-independent growth, as well as a motile phenotype of PrCa cells. Given our observations that high levels of αVß3 and, as shown here, of NgR2 are detected in human and mouse NEPrCa, our findings appear to be highly relevant to this aggressive and metastatic subtype of PrCa. This study is novel because NgR2 role has only minimally been investigated in cancer and has instead predominantly been analyzed in neurons. These data thus pave new avenues toward a comprehensive mechanistic understanding of integrin-directed signaling during PrCa progression toward a NE phenotype.


Subject(s)
Carcinoma, Neuroendocrine , Nogo Receptor 2 , Prostatic Neoplasms , Animals , Humans , Male , Mice , Androgen Antagonists , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Integrins , Prostatic Neoplasms/pathology , Nogo Receptor 2/metabolism
8.
J Neurosci ; 30(40): 13220-34, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20926648

ABSTRACT

Although the role of myelin-derived Nogo-A as an inhibitor of axonal regeneration after CNS injury has been thoroughly described, its physiological function in the adult, uninjured CNS is less well known. We address this question in the hippocampus, where Nogo-A is expressed by neurons as well as oligodendrocytes. We used 21 d in vitro slice cultures of neonatal hippocampus where we applied different approaches to interfere with Nogo-A signaling and expression and analyze their effects on the dendritic and axonal architecture of pyramidal cells. Neutralization of Nogo-A by function-blocking antibodies induced a major alteration in the dendrite structure of hippocampal pyramidal neurons. Although spine density was not influenced by Nogo-A neutralization, spine type distribution was shifted toward a more immature phenotype. Axonal complexity and length were greatly increased. Nogo-A KO mice revealed a weak dendritic phenotype resembling the effect of the antibody treatment. To discriminate a possible cell-autonomous role of Nogo-A from an environmental, receptor-mediated function, we studied the effects of short hairpin RNA-induced knockdown of Nogo-A or NgR1, a prominent Nogo-A receptor, within individual neurons. Knockdown of Nogo-A reproduced part of the dendritic and none of the spine or axon alterations. However, downregulation of NgR1 replicated the dendritic, the axonal, and the spine alterations observed after Nogo-A neutralization. Together, our results demonstrate that Nogo-A plays a major role in stabilizing and maintaining the architecture of hippocampal pyramidal neurons. Mechanistically, although the majority of the activity of Nogo-A relies on a receptor-mediated mechanism involving NgR1, its cell-autonomous function plays a minor role.


Subject(s)
Hippocampus/cytology , Hippocampus/growth & development , Myelin Proteins/physiology , Animals , Cell Differentiation/genetics , Cell Shape/genetics , Cells, Cultured , Conditioning, Operant/physiology , Dendrites/metabolism , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/metabolism , Nogo Proteins , Organ Culture Techniques , Protein Stability , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Synaptic Potentials/genetics
9.
J Neuroinflammation ; 8: 113, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21906273

ABSTRACT

BACKGROUND: Nogo-66 receptor NgR1 and its structural homologue NgR2 are binding proteins for a number of myelin-associated inhibitory factors. After neuronal injury, these inhibitory factors are responsible for preventing axonal outgrowth via their interactions with NgR1 and NgR2 expressed on neurons. In vitro, cells expressing NgR1/2 are inhibited from adhering to and spreading on a myelin substrate. Neuronal injury also results in the presence of dendritic cells (DCs) in the central nervous system, where they can come into contact with myelin debris. The exact mechanisms of interaction of immune cells with CNS myelin are, however, poorly understood. METHODS: Human DCs were differentiated from peripheral blood monocytes and mouse DCs were differentiated from wild type and NgR1/NgR2 double knockout bone marrow precursors. NgR1 and NgR2 expression were determined with quantitative real time PCR and immunoblot, and adhesion of cells to myelin was quantified. RESULTS: We demonstrate that human immature myeloid DCs express NgR1 and NgR2, which are then down-regulated upon maturation. Human mature DCs also adhere to a much higher extent to a myelin substrate than immature DCs. We observe the same effect when the cells are plated on Nogo-66-His (binding peptide for NgR1), but not on control proteins. Mature DCs taken from Ngr1/2 knockout mice adhere to a much higher extent to myelin compared to wild type mouse DCs. In addition, Ngr1/2 knockout had no effect on in vitro DC differentiation or phenotype. CONCLUSIONS: These results indicate that a lack of NgR1/2 expression promotes the adhesion of DCs to myelin. This interaction could be important in neuroinflammatory disorders such as multiple sclerosis in which peripheral immune cells come into contact with myelin debris.


Subject(s)
Cell Adhesion/physiology , Dendritic Cells/metabolism , Myelin Proteins/metabolism , Myelin Sheath/metabolism , Protein Isoforms/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Differentiation , Cytokines/metabolism , Dendritic Cells/cytology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Lymphocyte Subsets , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/physiology , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Proteins/genetics , Myelin Sheath/genetics , Nogo Proteins , Nogo Receptor 1 , Nogo Receptor 2 , Nogo Receptors , Protein Isoforms/genetics , Receptors, Cell Surface/genetics
10.
Front Cell Neurosci ; 13: 332, 2019.
Article in English | MEDLINE | ID: mdl-31396054

ABSTRACT

Restoration of neuronal connectivity after lesion of the central nervous system, such as spinal cord injury, is one of the biggest challenges in modern medicine. In particular, the accumulation of axon growth inhibitory factors at the site of injury constitutes a major obstacle to structural and thus functional repair. We previously investigated a group of prenylflavonoids derived from hops for their capacity to promote neuroregeneration. We identified a molecule called ENDF1 that was very potent to enhance regrowth and branching of neurites from dorsal root ganglion neurons in culture on growth promoting substrates. In the present study, we investigated ENDF1's capacity to promote regeneration of rat dorsal root ganglion neurons in vitro in the presence of three main components of the extracellular matrix acting as axon growth inhibitors: Semaphorin 3A, Ephrin A4 and mixed chondroitin sulfate proteoglycans. We report that ENDF1 application significantly promoted the percentages of sensory neurons able to regrow their neurites regardless of the presence of those inhibitors, and this to an extent similar to the one obtained after NGF treatment. Moreover, ENDF1 strongly enhanced the total neurite length and the complexity of neurites extending from neurons challenged with axon growth inhibitors. Although the impact of NGF and ENDF1 on the regeneration of neurons was similar, the activity of ENDF1 was not mediated by signaling through the TrkA receptor, indicating that each molecule act through different signaling pathways. In addition, ENDF1 did not decrease the phosphorylation of cofilin, a downstream effector of the regeneration-associated RhoA/ROCK signaling pathway. Hence, ENDF1 is a potent pro-neuroregenerative factors that could help in identifying new efficient targets for regenerative therapies of the nervous system.

11.
Front Mol Neurosci ; 10: 317, 2017.
Article in English | MEDLINE | ID: mdl-29066950

ABSTRACT

The bioactive lipid sphingosine-1-phosphate (S1P) is an important regulator in the nervous system. Here, we explored the role of S1P and its receptors in vitro and in preclinical models of peripheral nerve regeneration. Adult sensory neurons and motor neuron-like cells were exposed to S1P in an in vitro assay, and virtually all neurons responded with a rapid retraction of neurites and growth cone collapse which were associated with RhoA and ROCK activation. The S1P1 receptor agonist SEW2871 neither activated RhoA or neurite retraction, nor was S1P-induced neurite retraction mitigated in S1P1-deficient neurons. Depletion of S1P3 receptors however resulted in a dramatic inhibition of S1P-induced neurite retraction and was on the contrary associated with a significant elongation of neuronal processes in response to S1P. Opposing responses to S1P could be observed in the same neuron population, where S1P could activate S1P1 receptors to stimulate elongation or S1P3 receptors and retraction. S1P was, for the first time in sensory neurons, linked to the phosphorylation of collapsin response-mediated protein-2 (CRMP2), which was inhibited by ROCK inhibition. The improved sensory recovery after crush injury further supported the relevance of a critical role for S1P and receptors in fine-tuning axonal outgrowth in peripheral neurons.

12.
J Neurotrauma ; 23(3-4): 384-96, 2006.
Article in English | MEDLINE | ID: mdl-16629624

ABSTRACT

Myelin of the adult mammalian central nervous system (CNS) has been attributed to suppress structural plasticity and to impede regenerating nerve fibers. Nogo-A is possibly the best characterized of a variety of neurite growth inhibitors present in CNS myelin. Neutralizing its activity results in improved axon regrowth and functional recovery in experimental CNS lesion models of adult rodents and primates. While Nogo-A has become a major target for therapeutic intervention to promote axon regeneration in the CNS, it is realized that such an approach will likely have to be combined with other therapeutic strategies to maximize functional recovery after spinal cord injury (SCI).


Subject(s)
Myelin Proteins/physiology , Spinal Cord Injuries/physiopathology , Animals , Humans , Neurites/physiology , Nogo Proteins , Signal Transduction/physiology , Spinal Cord Injuries/metabolism
13.
J Neurosci ; 22(23): 10368-76, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12451136

ABSTRACT

The adult mammalian CNS has a limited capacity for nerve regeneration and structural plasticity. The presence of glia-derived inhibitory factors myelin-associated glycoprotein (MAG) and Nogo-A have been suggested to provide a nonpermissive environment for elongating nerve fibers. In particular, Nogo-A, an integral membrane protein predominantly expressed by oligodendrocytes, has been demonstrated to impair neurite growth in vitro and in vivo. Structure function analysis revealed that Nogo-A protein contains at least two active domains, NiG and Nogo-66, with diverse effects on neurite outgrowth and cell spreading. We now provide evidence that these inhibitory domains mediate their effects via an antagonistic regulation of the small GTPases RhoA and Rac1, resulting in activation of RhoA and suppression of Rac1. By inactivating RhoA with C3 transferase or the downstream effector Rho-kinase ROCK with, the inhibitory effects of both Nogo-A fragments and MAG on neurite outgrowth and oligodendrocyte-mediated growth cone collapse were abolished. Furthermore, we show that the recently cloned receptor for Nogo-66 and MAG, NgR, is not necessary for either NiG- or MAG-induced RhoA activation.


Subject(s)
Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/metabolism , Neurites/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Amides/pharmacology , Animals , CHO Cells , Cell Communication/drug effects , Cell Communication/physiology , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Cricetinae , Enzyme Inhibitors/pharmacology , GPI-Linked Proteins , Gene Expression Regulation/physiology , Growth Cones/drug effects , Growth Cones/physiology , Humans , Intracellular Signaling Peptides and Proteins , Kidney/cytology , Kidney/metabolism , Microscopy, Video , Myelin Proteins/chemistry , Myelin Proteins/genetics , Myelin Proteins/pharmacology , Myelin-Associated Glycoprotein/pharmacology , Neurites/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nogo Proteins , Nogo Receptor 1 , Oligodendroglia/cytology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Structure, Tertiary/physiology , Pyridines/pharmacology , Rats , Receptors, Cell Surface/metabolism , rho-Associated Kinases , rhoA GTP-Binding Protein/antagonists & inhibitors
14.
J Neurosci ; 23(13): 5393-406, 2003 Jul 02.
Article in English | MEDLINE | ID: mdl-12843238

ABSTRACT

Nogo-A is a potent neurite growth inhibitor in vitro and plays a role both in the restriction of axonal regeneration after injury and in structural plasticity in the CNS of higher vertebrates. The regions that mediate inhibition and the topology of the molecule in the plasma membrane have to be defined. Here we demonstrate the presence of three different active sites: (1) an N-terminal region involved in the inhibition of fibroblast spreading, (2) a stretch encoded by the Nogo-A-specific exon that restricts neurite outgrowth and cell spreading and induces growth cone collapse, and (3) a C-terminal region (Nogo-66) with growth cone collapsing function. We show that Nogo-A-specific active fragments bind to the cell surface of responsive cells and to rat brain cortical membranes, suggesting the existence of specific binding partners or receptors. Several antibodies against different epitopes on the Nogo-A-specific part of the protein as well as antisera against the 66 aa loop in the C-terminus stain the cell surface of living cultured oligodendrocytes. Nogo-A is also labeled by nonmembrane-permeable biotin derivatives applied to living oligodendrocyte cultures. Immunofluorescent staining of intracellular, endoplasmic reticulum-associated Nogo-A in cells after selective permeabilization of the plasma membrane reveals that the epitopes of Nogo-A, shown to be accessible at the cell surface, are exposed to the cytoplasm. This suggests that Nogo-A could have a second membrane topology. The two proposed topological variants may have different intracellular as well as extracellular functions.


Subject(s)
Myelin Proteins/physiology , Neurites/physiology , 3T3 Cells , Animals , Axons/drug effects , Axons/physiology , Binding Sites/physiology , Biotinylation , Brain Chemistry , CHO Cells , Cell Adhesion , Cell Membrane/chemistry , Cell Membrane/metabolism , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Chick Embryo , Cricetinae , Fibroblasts/metabolism , GPI-Linked Proteins , Mice , Molecular Sequence Data , Myelin Proteins/genetics , Myelin Proteins/metabolism , Nogo Proteins , Nogo Receptor 1 , Oligodendroglia/metabolism , Protein Binding/physiology , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Rats , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Deletion
15.
Sci STKE ; 2004(235): pe24, 2004 May 25.
Article in English | MEDLINE | ID: mdl-15173600

ABSTRACT

In the adult mammalian central nervous system (CNS), growth of neuronal fibers is actively inhibited by myelin. The proteins myelin-associated glycoprotein (MAG), oligodendrocyte myelin glycoprotein (OMgP), and Nogo-66 have been identified as inhibitory components present in CNS myelin. All three proteins exert their inhibitory activity by binding to a neuronal receptor complex containing the Nogo-66 receptor (NgR) and the neurotrophin (NT) receptor p75NTR. In their recent publication, Mi et al. identify the novel protein Lingo-1 as an interactor of p75NTR and NgR. The Lingo-1-NgR-p75NTR complex is shown to confer the inhibitory effects on nerve cell regeneration of Nogo-66, OMgP, and MAG by activating the small guanosine triphosphatase (GTPase) RhoA. Together with the recent finding that p75NTR interacts with the transmembrane protein sortilin to form a different receptor complex with cell death-promoting activity, the results of Mi et al. indicate that p75NTR exerts its diverse cellular functions by associating with function-specific co-receptors.


Subject(s)
Nerve Regeneration/physiology , Neurons/physiology , Protein Subunits/physiology , Receptors, Nerve Growth Factor/physiology , Cell Death/physiology , Receptor, Nerve Growth Factor , Signal Transduction/physiology
16.
Eur J Cell Biol ; 83(2): 51-4, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15146976

ABSTRACT

Mammalian neurons require a constant supply of oxygen to maintain adequate cellular functions and survival. Following sustained hypoxia during ischemic events in brain, the energy status of neurons and glia is compromised, which may subsequently lead to cell death by apoptosis and necrosis. Concomitant with energy depletion is the formation of the purine nucleoside adenosine, a powerful endogenous neuroprotectant. In this paper the effect of chemical hypoxia on cell survival and neurite outgrowth of primary cerebellar granule cells was investigated. Rotenone, a mitochondrial complex I inhibitor, induced a 30.4 +/- 3.6% loss of viable cells and a 35.0 +/- 4.4% loss of neurite formation of cerebellar granule cells, which was partially restored by the addition of purine nucleosides adenosine, inosine and guanosine. Inosine had the most striking effect of 37.7 +/- 2.9% rescue of viability and 71.2 +/- 18.4% rescue of neurite outgrowth. Data confirm the suggested role of purine nucleosides for the neuronal regeneration of primary brain cells following hypoxic insult.


Subject(s)
Cell Hypoxia/drug effects , Cerebellum/cytology , Neurites/drug effects , Neuroprotective Agents/pharmacology , Purine Nucleosides/pharmacology , Adenosine/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Guanosine/pharmacology , Inosine/pharmacology , Neurites/physiology , Rats , Rats, Sprague-Dawley , Rotenone/pharmacology , Time Factors , Uncoupling Agents/pharmacology
17.
Eur J Neurosci ; 2(1): 69-76, 1990 Jan.
Article in English | MEDLINE | ID: mdl-12106104

ABSTRACT

In order to obtain information on the physiological regulation of NGF-synthesis in the central nervous system (CNS) we investigated the effects of a series of growth factors (known to be present in the CNS) in cultures of purified rat astrocytes and compared these effects with those observed after intraventricular injection of the same molecules. After preliminary experiments had shown that 10% fetal calf serum (FCS) produced a marked increase in NGF-mRNA levels in astrocytes (but neither in microglia nor oligodendrocytes) as demonstrated by Northern blot analysis and in situ hybridization the experiments were performed at low (0.5%) FCS concentrations. Supramaximal concentrations of IL-1 and various growth factors caused a 5- to 7-fold increase in NGF-mRNA after 6 h. By 24 h the NGF-mRNA levels approached control values again, most probably due to inactivation of the added factors since after readdition after 24 h the response was about the same as the initial one. Norepinephrine and 8-bromo-cAMP did not change NGF-mRNA levels. The growth factor-mediated changes in NGF-mRNA levels in astrocyte cultures were not consistently reflected by the changes observed after intraventricular injection. IL-1 produced by far the largest increase in hippocampal NGF-mRNA after intraventricular injection. This large response to IL-1 could result from a positive feedback mechanism, since IL-1beta injection not only increases NGF-mRNA but also IL-1beta-mRNA in the hippocampus. The understanding of the physiological regulation of NGF synthesis in the CNS is the basis for a rational approach to its pharmacological modification. This, in turn, is an attractive alternative to the (long-term) infusion of NGF or the transplantation of NGF-secreting cells with the goal of providing trophic support to the cholinergic neurons of the basal forebrain nuclei. These neurons are consistently affected in the early stages of Alzheimer's disease, their impaired function being essentially responsible for the cognitive deficits.

18.
J Neuroimmunol ; 145(1-2): 139-47, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14644040

ABSTRACT

Nogo-A is a protein associated with central nervous system (CNS) myelin thought to impair regenerative responses and to suppress sprouting and plastic changes of synaptic terminals. In this study, we report that serum IgM autoantibodies to the recombinant large N-terminal inhibitory domain of Nogo-A are a frequent finding in multiple sclerosis (MS) and acute inflammatory (IND) and non-inflammatory neurological diseases (OND), but not in neurodegenerative diseases (ND), systemic inflammatory disease and healthy controls. Furthermore, we demonstrate intrathecal production of anti-Nogo-A antibodies measured by increased IgG indices. Intrathecal anti-Nogo antibodies were significantly more frequent in patients with relapsing-remitting as compared to chronic progressive (CP) MS. We also found a highly significant negative correlation of these antibody responses with age indicating that they are more frequent in younger patients. We finally demonstrate that human anti-Nogo-A antibodies recognize native Nogo-A in brain extracts, oligodendrocytes and cells expressing human Nogo-A.


Subject(s)
Autoantibodies/blood , Autoantibodies/cerebrospinal fluid , Autoantigens/immunology , Multiple Sclerosis/immunology , Myelin Proteins/immunology , Acute Disease , Adult , Aged , Animals , Autoantibodies/biosynthesis , Autoantibodies/metabolism , Brain/immunology , Brain/metabolism , CHO Cells , Cells, Cultured , Central Nervous System Diseases/blood , Central Nervous System Diseases/cerebrospinal fluid , Central Nervous System Diseases/immunology , Cricetinae , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/cerebrospinal fluid , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/cerebrospinal fluid , Nogo Proteins , Oligodendroglia/immunology , Oligodendroglia/metabolism , Rats , Transfection
19.
Exp Gerontol ; 38(1-2): 79-86, 2003.
Article in English | MEDLINE | ID: mdl-12543264

ABSTRACT

Unlike neonatal axons, mammalian adult axons of the CNS do not regenerate after injury. This developmental loss of regenerative capacity, is correlated with the onset of myelination. Likewise, myelin, or myelin-associated components such as Nogo-A and myelin-associated glycoprotein (MAG) inhibit regeneration from older but not younger neurons. Identification of the molecular events responsible for this developmental loss of regenerative capacity is central to devise strategies to encourage regeneration in adults after injury. Endogenous levels of the cyclic nucleotides cAMP and cGMP have been suggested to determine the neuronal responsiveness to various axonal guidance factors. Elevating cAMP concentrations block Nogo-A or MAG induced inhibition of neurite outgrowth in older neurons, whereas suppressing cAMP levels in young neurons renders them susceptible to Nogo-A and MAG. Interestingly, elevated cAMP levels abrogated the Nogo-A and MAG mediated activation of RhoA and down regulation of Rac1 in adult neurons. In contrast, elevation of cAMP leads to the inactivation of RhoA and prevents activation of downstream effector proteins, while Rac is activated. We therefore conclude that the endogenous neuronal cAMP levels determine the neuronal responsiveness to myelin-associated neurite growth inhibitors by regulating rho GTPase activities.


Subject(s)
Carbazoles , Cerebellum/physiology , Growth Inhibitors/pharmacology , Myelin Proteins/pharmacology , Nerve Regeneration/drug effects , Neurites/drug effects , Signal Transduction/drug effects , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Myelin-Associated Glycoprotein/pharmacology , Neurites/metabolism , Neurons/drug effects , Neurons/metabolism , Nogo Proteins , Pyrroles/pharmacology , Rats , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
20.
Front Behav Neurosci ; 8: 175, 2014.
Article in English | MEDLINE | ID: mdl-24860456

ABSTRACT

Molecular mechanisms which stabilize dendrites and dendritic spines are essential for regulation of neuronal plasticity in development and adulthood. The class of Nogo receptor proteins, which are critical for restricting neurite outgrowth inhibition signaling, have been shown to have roles in developmental, experience and activity induced plasticity. Here we investigated the role of the Nogo receptor homolog NgR2 in structural plasticity in a transgenic null mutant for NgR2. Using Golgi-Cox staining to analyze morphology, we show that loss of NgR2 alters spine morphology in adult CA1 pyramidal neurons of the hippocampus, significantly increasing mushroom-type spines, without altering dendritic tree complexity. Furthermore, this shift is specific to apical dendrites in distal CA1 stratum radiatum (SR). Behavioral alterations in NgR2(-/-) mice were investigated using a battery of standardized tests and showed that whilst there were no alterations in learning and memory in NgR2(-/-) mice compared to littermate controls, NgR2(-/-) displayed reduced fear expression in the contextual conditioned fear test, and exhibited reduced anxiety- and depression-related behaviors. This suggests that the loss of NgR2 results in a specific phenotype of reduced emotionality. We conclude that NgR2 has role in maintenance of mature spines and may also regulate fear and anxiety-like behaviors.

SELECTION OF CITATIONS
SEARCH DETAIL