Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 303
Filter
1.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Article in English | MEDLINE | ID: mdl-33893233

ABSTRACT

Peripheral myelin protein (PMP22) is an integral membrane protein that traffics inefficiently even in wild-type (WT) form, with only 20% of the WT protein reaching its final plasma membrane destination in myelinating Schwann cells. Misfolding of PMP22 has been identified as a key factor in multiple peripheral neuropathies, including Charcot-Marie-Tooth disease and Dejerine-Sottas syndrome. While biophysical analyses of disease-associated PMP22 mutants show altered protein stabilities, leading to reduced surface trafficking and loss of PMP22 function, it remains unclear how destabilization of PMP22 mutations causes mistrafficking. Here, native ion mobility-mass spectrometry (IM-MS) is used to compare the gas phase stabilities and abundances for an array of mutant PM22 complexes. We find key differences in the PMP22 mutant stabilities and propensities to form homodimeric complexes. Of particular note, we observe that severely destabilized forms of PMP22 exhibit a higher propensity to dimerize than WT PMP22. Furthermore, we employ lipid raft-mimicking SCOR bicelles to study PMP22 mutants, and find that the differences in dimer abundances are amplified in this medium when compared to micelle-based data, with disease mutants exhibiting up to 4 times more dimer than WT when liberated from SCOR bicelles. We combine our findings with previous cellular data to propose that the formation of PMP22 dimers from destabilized monomers is a key element of PMP22 mistrafficking.


Subject(s)
Myelin Proteins/metabolism , Peripheral Nervous System Diseases/physiopathology , Protein Transport/physiology , Cell Membrane/metabolism , Humans , Ion Mobility Spectrometry/methods , Mass Spectrometry/methods , Membrane Proteins/metabolism , Myelin Proteins/genetics , Myelin Proteins/physiology , Peripheral Nervous System Diseases/diagnostic imaging , Peripheral Nervous System Diseases/metabolism , Protein Folding , Protein Stability , Schwann Cells/metabolism
2.
J Neuroimmunol ; 333: 476953, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31108399

ABSTRACT

Specific neutralization of the pathogenic autoimmune cells is the ultimate goal in therapy of Multiple Sclerosis (MS). However, the pathogenic autoimmunity in MS, can be directed against several major target antigens, and therefore targeting pathogenic T-cells directed against a single target antigen is unlikely to be effective. To overcome this multiplicity and the potential complexity of pathogenic autoreactivities in MS, we have put forward the concept of concomitant multi-antigen/multi-epitope targeting as, a conceivably more effective approach to immunotherapy of MS. We constructed an (Experimental Autoimmune Encephalomeylitis (EAE)/MS-related synthetic human Target Autoantigen Gene (MS-shMultiTAG) designed to encode in tandem only EAE/MS related epitopes of all known encephalitogenic proteins. The MS-related protein product (designated Y-MSPc) was immunofunctional and upon tolerogenic administration, it effectively suppressed and reversed EAE induced by a single encephalitogenic protein. Furthermore, Y-MSPc also fully abrogated the development of "complex EAE" induced by a mixture of five encephalitogenic T-cell lines, each specific for a different encephalitogenic epitope of MBP, MOG, PLP, MOBP and OSP. Strikingly, Y-MSPc was consistently more effective than treatment with the single disease-specific peptide or with the peptide cocktail, both in suppressing the development of "classical" or "complex" EAE and in ameliorating ongoing disease. Overall, the modulation of EAE by Y-MSPc was associated with anergizing the pathogenic autoreactive T-cells, downregulation of Th1/Th17 cytokine secretion and upregulation of TGF-ß secretion. Moreover, we show that both suppression and treatment of ongoing EAE by tolerogenic administration of Y-MSPc is associated also with a remarkable increase in a unique subset of dendritic-cells (DCs), CD11c+CD11b+Gr1+-myeloid derived DCs in both spleen and CNS of treated mice. These DCs, which are with strong immunoregulatory characteristics and are functional in down-modulation of MS-like-disease displayed increased production of IL-4, IL-10 and TGF-ß and low IL-12. Functionally, these myeloid DCs suppress the in-vitro proliferation of myelin-specific T-cells and more importantly, the cells were functional in-vivo, as their adoptive transfer into EAE induced mice resulted in strong suppression of the disease, associated with a remarkable induction of CD4 + FoxP3+ regulatory cells. These results, which highlight the efficacy of "multi-epitope-targeting" agent in induction of functional regulatory CD11c+CD11b+Gr1+myeloid DCs, further indicate the potential role of these DCs in maintaining peripheral tolerance and their involvement in downregulation of MS-like-disease.


Subject(s)
Dendritic Cells/physiology , Encephalomyelitis, Autoimmune, Experimental/therapy , Myelin Proteins/therapeutic use , Myeloid Cells/physiology , Adoptive Transfer , Amino Acid Sequence , Animals , Antigens, Ly/analysis , CD11 Antigens/analysis , CD11b Antigen/analysis , Central Nervous System/immunology , Central Nervous System/pathology , Cytokines/biosynthesis , Cytokines/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Epitopes/immunology , Female , Immune Tolerance/drug effects , Mice , Mice, Inbred Strains , Myelin Proteins/immunology , Myelin Proteins/physiology , Peptide Fragments/immunology , Recombinant Proteins/therapeutic use , Spleen/immunology , Spleen/pathology , T-Cell Antigen Receptor Specificity , T-Lymphocytes, Cytotoxic/immunology
4.
Neurochem Res ; 40(9): 1767-85, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26266872

ABSTRACT

Nogos have become a hot topic for its well-known number Nogo-A's big role in clinical matters. It has been recognized that the expression of Nogo-A and the receptor NgR1 inhibit the neuron's growth after CNS injuries or the onset of the MS. The piling evidence supports the notion that the Nogo-A is also involved in the synaptic plasticity, which was shown to negatively regulate the strength of synaptic transmission. The occurrence of significant schizophrenia-like behavioral phenotypes in Nogo-A KO rats also added strong proof to this conclusion. This review mainly focuses on the structure of Nogo-A and its corresponding receptor-NgR1, its intra- and extra-cellular signaling, together with its major physiological functions such as regulation of migration and distribution and its related diseases like stroke, AD, ALS and so on.


Subject(s)
Central Nervous System Diseases/physiopathology , Central Nervous System/physiology , Myelin Proteins/physiology , Animals , Animals, Genetically Modified , Neuronal Plasticity , Nogo Proteins , Rats , Signal Transduction
5.
Nat Med ; 21(9): 1028-1037, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26301690

ABSTRACT

Endothelial dysfunction is a critical factor in many cardiovascular diseases, including hypertension. Although lipid signaling has been implicated in endothelial dysfunction and cardiovascular disease, specific molecular mechanisms are poorly understood. Here we report that Nogo-B, a membrane protein of the endoplasmic reticulum, regulates endothelial sphingolipid biosynthesis with direct effects on vascular function and blood pressure. Nogo-B inhibits serine palmitoyltransferase, the rate-limiting enzyme of the de novo sphingolipid biosynthetic pathway, thereby controlling production of endothelial sphingosine 1-phosphate and autocrine, G protein-coupled receptor-dependent signaling by this metabolite. Mice lacking Nogo-B either systemically or specifically in endothelial cells are hypotensive, resistant to angiotensin II-induced hypertension and have preserved endothelial function and nitric oxide release. In mice that lack Nogo-B, pharmacological inhibition of serine palmitoyltransferase with myriocin reinstates endothelial dysfunction and angiotensin II-induced hypertension. Our study identifies Nogo-B as a key inhibitor of local sphingolipid synthesis and shows that autocrine sphingolipid signaling within the endothelium is critical for vascular function and blood pressure homeostasis.


Subject(s)
Blood Pressure , Endothelium, Vascular/physiology , Homeostasis , Myelin Proteins/physiology , Sphingolipids/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Lysophospholipids/physiology , Male , Mice , Nitric Oxide Synthase Type III/physiology , Nogo Proteins , Receptors, Lysosphingolipid/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology
6.
Biochemistry ; 54(29): 4443-52, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26158299

ABSTRACT

Zinc finger (ZF) proteins make up a large family of metalloproteins that contain discrete domains with amino acid ligands (cysteine and histidine) that serve to coordinate zinc in a tetrahedral geometry. Upon zinc coordination, the domains adopt three-dimensional structure. The most well-studied ZFs are the "classical" ZFs, which use a Cys2His2 motif to bind zinc and adopt an antiparallel ß sheet/α helical fold. In addition to the classical ZF class, at least 13 other ZF classes, collectively termed nonclassical ZFs, have been identified. These other classes are distinguished by the combination and order of the cysteine and histidine ligands within each domain, the spacing between each ligand (i.e., number and type of amino acid), and the structural architecture that the domain adopts in the presence of zinc. One class of nonclassical ZFs, the neural zinc finger/myelin transcription factor (NZF/MyT) class, contains ZF domains with a Cys2His2Cys ligand set, adopts a fold that consists of a series of loops in the presence of zinc, and functions as transcription factors by binding to and regulating genes that are critical for the development of the central nervous system. This Current Topic focuses on our understanding of the NZF/MyT class of nonclassical ZFs and presents current hypotheses regarding this class' unique mechanism of metal-mediated folding and function.


Subject(s)
Nerve Tissue Proteins/physiology , Transcription Factors/physiology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cobalt/chemistry , Conserved Sequence , Humans , Molecular Sequence Data , Myelin Proteins/chemistry , Myelin Proteins/physiology , Nerve Tissue Proteins/chemistry , Protein Binding , Transcription Factors/chemistry , Zinc/chemistry , Zinc Fingers
7.
Zhongguo Gu Shang ; 28(3): 235-9, 2015 Mar.
Article in Chinese | MEDLINE | ID: mdl-25936193

ABSTRACT

OBJECTIVE: To study the effects of Jisuikang (Chinese characters) on Nogo-NgR gene expression, and to explore the protective effects and mechanism of Jisuikang (Chinese characters) on spinal cord injury in rats. METHODS: One hundred eighty female rats were randomly assigned to 6 groups(30 rats per group). Sham group: T10 lamina was resected only and spinal cord was untreated. Model group: spine cord injury (SCI) was created with a modified impinger of Allen's by impacting on the T10 spinal cord. Prednisolone group: Prednisolone (0.06 g/kg) was given by intragastric administration at a time interval of 24 hours after operation. The Jisuikang (Chinese characters) high, moderate and low dose groups: Jisuikang (Chinese characters) was supplied with different dose (50 g/kg, 25 g/kg, 12.5 g/kg) by intragastric administration in rats after operation,for the first time at 30 min after surgery. Animals were killed 3, 7, 14 days after surgery. The expression levels of Nogo-A and NgR were observed by Western Blot and Real-time PCR. RESULTS: The expression of Nogo-A and NgR was at the basic level at all time points in sham group. Compared with model group, the protein expression levels of Nogo-A and NgR in sham, prednisolone, Jisuikang (Chinese characters) moderate dose groups were statistically significant at all time points (P < 0.05). No difference was found in Jisuikang (Chinese characters) high and low dose groups (P > 0.05). Three days after surgery, the mRNA levels of Nogo-A and NgR in treatment group were significantly lower than that in model group (P < 0.01); 7 days after surgery,Nogo-A and NgR mRNA expression were dramatically upregulated and peaked; 14 days after operation, the expression was decreased, but still significantly higher than that in other treatment groups (P < 0.01). Prednisolone and Jisuikang (Chinese characters) moderate dose groups showed the most significant effects among all groups,but there was no statistically significant difference between two groups (P > 0.05). CONCLUSION: The decoction Jisuikang (Chinese characters) can promote the nerve cell regeneration by regulating Nogo-A and NgR gene expression, activating Nogo- NgR signaling pathways after acute spinal cord injury.


Subject(s)
Medicine, Chinese Traditional , Myelin Proteins/genetics , Receptors, Cell Surface/genetics , Spinal Cord Injuries/drug therapy , Animals , Female , GPI-Linked Proteins/analysis , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Myelin Proteins/analysis , Myelin Proteins/physiology , Nerve Regeneration/drug effects , Nogo Proteins , Nogo Receptor 1 , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/analysis , Receptors, Cell Surface/physiology , Signal Transduction/drug effects , Spinal Cord Injuries/metabolism
8.
J Neurosci ; 35(16): 6413-28, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904793

ABSTRACT

Axons in the adult CNS have poor ability to grow after injury, impeding functional recovery in patients of spinal cord injury. This has been attributed to both a developmental decline in neuron-intrinsic growth ability and the presence of extrinsic growth inhibitors. We previously showed that genetic deletion of Nogo, an extrinsic inhibitor, promoted axonal sprouting from uninjured corticospinal tract (CST) neurons but not regeneration from injured CST neurons, whereas genetic deletion of PTEN, an intrinsic inhibitor, promoted both CST sprouting and regeneration. Here we test the hypothesis that combining an elevation of neuron-intrinsic growth ability and a reduction of extrinsic growth inhibition by genetic codeletion of PTEN and Nogo may further improve injury-induced axonal growth. In an apparent paradox, additionally deleting Nogo further enhanced CST regeneration but not sprouting in PTEN-deleted mice. Enhanced CST regeneration and sprouting in PTEN and PTEN/Nogo-deleted mice were associated with no or only temporary improvement in functional recovery. Our data illustrate that neuron-intrinsic and -extrinsic factors regulate axon regeneration and sprouting in complex ways and provide proof-of-principle evidence that targeting both can further improve regeneration. Neuron-intrinsic growth ability is an important determinant of neuronal responsiveness to changes in extrinsic growth inhibition, such that an elevated intrinsic growth state is a prerequisite for reducing extrinsic inhibition to take effect on CST regeneration. Meanwhile, additional strategies are required to unleash the full potential for functional recovery with enhanced axon regeneration and/or sprouting.


Subject(s)
Axons/physiology , Myelin Proteins/deficiency , Nerve Regeneration/physiology , PTEN Phosphohydrolase/deficiency , Pyramidal Tracts/physiology , Animals , Behavior, Animal/physiology , Mice , Mice, Mutant Strains , Myelin Proteins/genetics , Myelin Proteins/physiology , Nerve Regeneration/genetics , Nogo Proteins , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Recovery of Function/genetics , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology
9.
Neural Dev ; 10: 6, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25888884

ABSTRACT

BACKGROUND: In contrast to mammals, zebrafish successfully regenerate retinal ganglion cell (RGC) axons after optic nerve section (ONS). This difference is explained on the one hand by neurite growth inhibitors in mammals (including Nogo-A), as opposed to growth-promoting glial cells in the fish visual pathway, and on the other hand by the neuron-intrinsic properties allowing the upregulation of growth-associated proteins in fish RGCs but not in mammals. RESULTS: Here, we report that Rtn4b, the zebrafish homologue of mammalian Nogo-A/RTN4-A, is upregulated in axotomized zebrafish RGCs and is primarily associated with the endoplasmic reticulum (ER). Rtn4b functions as a neuron-intrinsic determinant for axon regeneration, as was shown by downregulating Rtn4b through retrogradely transported morpholinos (MOs), applied to the optic nerve at the time of ONS. MO1 and MO2 reduced the number of axons from retina explants in a concentration-dependent manner. With MO1, the reduction was 55% (70 µM MO1) and 74% (140 µM MO1), respectively, with MO2: 59% (70 µM MO2) and 73% (140 µM MO2), respectively (compared to the control MO-treated side). Moreover, regenerating axons 7d after ONS and MO1 or MO2 application were labeled by Alexa488, applied distal to the first lesion. The number of Alexa488 labeled RGCs, containing the Rtn4b MO1 or MO2, was reduced by 54% and 62%, respectively, over control MO. CONCLUSIONS: Thus, Rtn4b is an important neuron-intrinsic component and required for the success of axon regeneration in the zebrafish visual system. The spontaneous lesion-induced upregulation of Rtn4b in fish correlates with an increase in ER, soma size, biosynthetic activity, and thus growth and predicts that mammalian neurons require the same upregulation in order to successfully regenerate RGC axons.


Subject(s)
Myelin Proteins/physiology , Nerve Regeneration/physiology , Optic Nerve Injuries/genetics , Optic Nerve/physiology , Retinal Ganglion Cells/metabolism , Zebrafish Proteins/physiology , Animals , Axonal Transport , Axotomy , Endoplasmic Reticulum/metabolism , Morpholinos/pharmacology , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , Optic Nerve Injuries/metabolism , Up-Regulation , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics
10.
Acta Neurol Belg ; 115(4): 527-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25567550

ABSTRACT

For the most part, the central nervous system is unable to regenerate. The majority of injuries of vascular, inflammatory, degenerative and traumatic aetiology lead to an irreversible loss of central nervous system function. The paper presents the role of Nogo-A, MAG and OMgp proteins in the inhibition of central nervous system regeneration, and their potential clinical significance.


Subject(s)
Central Nervous System Diseases/physiopathology , Central Nervous System Diseases/therapy , Myelin Proteins/physiology , Nerve Regeneration/physiology , Animals , Axons/pathology , Axons/physiology , Humans , Nogo Proteins
11.
J Neurosci ; 34(48): 16140-52, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25429154

ABSTRACT

Haploinsufficiency of peripheral myelin protein 22 (PMP22) causes hereditary neuropathy with liability to pressure palsies, a peripheral nerve lesion induced by minimal trauma or compression. As PMP22 is localized to cholesterol-enriched membrane domains that are closely linked with the underlying actin network, we asked whether the myelin instability associated with PMP22 deficiency could be mediated by involvement of the protein in actin-dependent cellular functions and/or lipid raft integrity. In peripheral nerves and cells from mice with PMP22 deletion, we assessed the organization of filamentous actin (F-actin), and actin-dependent cellular functions. Using in vitro models, we discovered that, in the absence of PMP22, the migration and adhesion capacity of Schwann cells and fibroblasts are similarly impaired. Furthermore, PMP22-deficient Schwann cells produce shortened myelin internodes, and display compressed axial cell length and collapsed lamellipodia. During early postnatal development, F-actin-enriched Schmidt-Lanterman incisures do not form properly in nerves from PMP22(-/-) mice, and the expression and localization of molecules associated with uncompacted myelin domains and lipid rafts, including flotillin-1, cholesterol, and GM1 ganglioside, are altered. In addition, we identified changes in the levels and distribution of cholesterol and ApoE when PMP22 is absent. Significantly, cholesterol supplementation of the culture medium corrects the elongation and migration deficits of PMP22(-/-) Schwann cells, suggesting that the observed functional impairments are directly linked with cholesterol deficiency of the plasma membrane. Our findings support a novel role for PMP22 in the linkage of the actin cytoskeleton with the plasma membrane, likely through regulating the cholesterol content of lipid rafts.


Subject(s)
Actins/physiology , Cell Movement/physiology , Membrane Microdomains/physiology , Myelin Proteins/physiology , Animals , Cattle , Cells, Cultured , Female , Ganglia, Spinal/physiology , Male , Mice , Mice, Knockout
12.
Annu Rev Cell Dev Biol ; 30: 503-33, 2014.
Article in English | MEDLINE | ID: mdl-25288117

ABSTRACT

Myelination of axons in the nervous system of vertebrates enables fast, saltatory impulse propagation, one of the best-understood concepts in neurophysiology. However, it took a long while to recognize the mechanistic complexity both of myelination by oligodendrocytes and Schwann cells and of their cellular interactions. In this review, we highlight recent advances in our understanding of myelin biogenesis, its lifelong plasticity, and the reciprocal interactions of myelinating glia with the axons they ensheath. In the central nervous system, myelination is also stimulated by axonal activity and astrocytes, whereas myelin clearance involves microglia/macrophages. Once myelinated, the long-term integrity of axons depends on glial supply of metabolites and neurotrophic factors. The relevance of this axoglial symbiosis is illustrated in normal brain aging and human myelin diseases, which can be studied in corresponding mouse models. Thus, myelinating cells serve a key role in preserving the connectivity and functions of a healthy nervous system.


Subject(s)
Myelin Sheath/physiology , Adenosine Triphosphate/metabolism , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Axons/physiology , Central Nervous System/metabolism , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Cytoskeleton/ultrastructure , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Glucose/metabolism , Humans , Inflammation , Leukoencephalopathies/metabolism , Leukoencephalopathies/pathology , Mice , Microscopy, Electron , Myelin Proteins/physiology , Neuronal Plasticity , Oligodendroglia/physiology , Peripheral Nervous System/metabolism , Schwann Cells/physiology , Synaptic Transmission/physiology
13.
Neuroscience ; 283: 17-25, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25168730

ABSTRACT

Nogo-A interaction with its different receptors (Nogo receptor 1 (NgR1), S1P receptor 2 (S1PR2), paired immunoglobulin-like receptor B (PirB)) restricts plasticity and growth-dependent processes leading, via the activation of different signaling pathway to the stabilization of the neuronal networks (either developmentally or during processes of memory consolation in the mature nervous system). Taking away these molecular brakes might allow for the induction of extensive structural and functional rearrangements and might promote compensatory growth processes after an injury of the CNS, in cortical structures as well as in the spinal cord. However, it is important to keep in mind that this could as well be a dangerous endeavor, since it might facilitate unwanted and unnecessary (and probably even maladaptive) neuronal connections.


Subject(s)
Central Nervous System/physiology , Memory/physiology , Myelin Proteins/physiology , Neuronal Plasticity/physiology , Animals , Central Nervous System/cytology , Humans , Nerve Net/physiology , Nogo Proteins , Signal Transduction/physiology
14.
Anticancer Res ; 34(8): 4059-68, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25075030

ABSTRACT

BACKGROUND: Nogo-B was recently shown to be involved in proliferation, apoptosis and invasiveness of cancer cells, whereas its specific receptor (NgBR) was found to be up-regulated in estrogen receptor-α positive breast cancer. No data are currently available concerning their expression in non-small cell lung carcinomas (NSCLC). MATERIALS AND METHODS: Expression of Nogo isoforms and NgBR was studied in 191 NSCLC. RESULTS: Higher Nogo-A/B immunoreactivity was noted in cancer cells of squamous cell carcinomas (SQC) compared to adenocarcinomas (p<0.001). Stage II-IV tumors had the lowest Nogo-A/B expression (p<0.0001) compared to stage I cases. Nogo-A/B expression decreased with increasing SQC malignancy grade (p=0.026). Significant NgBR mRNA down-regulation was associated with larger primary tumor size (p=0.039), lymph node involvement (p=0.039) and advancement stage (p=0.0054). Low NgBR mRNA expression predicted poor patients outcome (p=0.029). CONCLUSION: The current data may point to the involvement of Nogo isoforms and NgBR in the pathogenesis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/chemistry , Lung Neoplasms/chemistry , Myelin Proteins/physiology , Receptors, Cell Surface/physiology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Immunohistochemistry , Lung Neoplasms/etiology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Myelin Proteins/analysis , Myelin Proteins/genetics , Neoplasm Staging , Nogo Proteins , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA, Messenger/analysis , Receptors, Cell Surface/analysis , Receptors, Cell Surface/genetics
15.
Neural Dev ; 9: 8, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24755266

ABSTRACT

BACKGROUND: As a consequence of gene/genome duplication, the RTN4/Nogo gene has two counterparts in zebrafish: rtn4a and rtn4b. The shared presence of four specific amino acid motifs-M1 to M4-in the N-terminal region of mammalian RTN4, and zebrafish Rtn4b suggests that Rtn4b is the closest homologue of mammalian Nogo-A. RESULTS: To explore their combined roles in zebrafish development, we characterized the expression patterns of rtn4a and rtn4b in a comparative manner and performed morpholino-mediated knockdowns. Although both genes were coexpressed in the neural tube and developing brain at early stages, they progressively acquired distinct expression domains such as the spinal cord (rtn4b) and somites (rtn4a). Downregulation of rtn4a and rtn4b caused severe brain abnormalities, with rtn4b knockdown severely affecting the spinal cord and leading to immobility. In addition, the retinotectal projection was severely affected in both morphants, as the retina and optic tectum appeared smaller and only few retinal axons reached the abnormally reduced tectal neuropil. The neuronal defects were more persistent in rtn4b morphants. Moreover, the latter often lacked pectoral fins and lower jaws and had malformed branchial arches. Notably, these defects led to larval death in rtn4b, but not in rtn4a morphants. CONCLUSIONS: In contrast to mammalian Nogo-A, its zebrafish homologues, rtn4a and particularly rtn4b, are essential for embryonic development and patterning of the nervous system.


Subject(s)
Myelin Proteins/physiology , Neurons/metabolism , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Brain/embryology , Down-Regulation , Myelin Proteins/genetics , Myelin Proteins/metabolism , Nogo Proteins , Retina/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
16.
Curr Opin Neurobiol ; 27: 53-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24632308

ABSTRACT

The expression of Nogo-A and the receptor NgR1 limits the recovery of adult mammals from central nervous system injury. Multiple studies have demonstrated efficacy from targeting this pathway for functional recovery and neural repair after spinal cord trauma, ischemic stroke, optic nerve injury and models of multiple sclerosis. Recent molecular studies have added S1PR2 as a receptor for the amino terminal domain of Nogo-A, and have demonstrated shared components for Nogo-A and CSPG signaling as well as novel Nogo antagonists. It has been recognized that neural repair involves plasticity, sprouting and regeneration. A physiologic role for Nogo-A and NgR1 has been documented in the restriction of experience-dependent plasticity with maturity, and the stability of synaptic, dendritic and axonal anatomy.


Subject(s)
Central Nervous System Diseases , Myelin Proteins/physiology , Neuronal Plasticity/physiology , Recovery of Function/physiology , Animals , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Central Nervous System Diseases/physiopathology , Humans , Nogo Proteins , Signal Transduction/physiology
17.
Neuromolecular Med ; 16(1): 3-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24218324

ABSTRACT

Reticulons (RTNs) are a group of membrane-associated proteins mainly responsible for shaping the tubular endoplasmic reticulum network, membrane trafficking, inhibition of axonal growth, and apoptosis. These proteins share a common sequence feature, the reticulon homology domain, which consists of paired hydrophobic stretches that are believed to induce membrane curvature by acting as a wedge in bilayer membranes. RTNs are ubiquitously expressed in all tissues, but each RTN member exhibits a unique expression pattern that prefers certain tissues or even cell types. Recently, accumulated evidence has suggested additional and unexpected roles for RTNs, including those on DNA binding, autophagy, and several inflammatory-related functions. These manifold actions of RTNs account for their ever-growing recognition of their involvement in neurodegenerative diseases like Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis, as well as hereditary spastic paraplegia. This review summarizes the latest discoveries on RTNs in human pathophysiology, and the engagement of these in neurodegeneration, along with the implications of these findings for a better understanding of the molecular events triggered by RTNs and their potential exploitation as next-generation therapeutics.


Subject(s)
Nerve Tissue Proteins/physiology , Neurodegenerative Diseases/metabolism , Animals , Apoptosis/physiology , Autophagy/physiology , Carrier Proteins/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Forecasting , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Membrane Proteins/physiology , Multigene Family , Myelin Proteins/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/classification , Neurogenesis/physiology , Nogo Proteins , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
18.
Kobe J Med Sci ; 60(3): E57-65, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25612671

ABSTRACT

Endoplasmic reticulum (ER) is an organelle that has an elaborate and continuous membrane system composed of sheet-like cisternae and a network of interconnected tubules. The ER tubules are shaped by reticulons, a conserved ER membrane protein family. However, how the membrane-shaping activity is regulated remains to be elucidated. To understand the mode of action of reticulons, we isolated TMEM33, a conserved protein harboring three transmembrane domains, as a reticulon 4C-binding protein by affinity chromatography. In addition to reticulon 4C, TMEM33 binds to reticulon 1A, -2B, -3C and a reticulon homology domain-containing protein Arl6IP1. Exogenously expressed TMEM33 localizes at both the ER membrane and the nuclear envelope. Exogenously expressed TMEM33 co-localizes with exogenously expressed reticulon 4C well at the ER sheets and partially at the ER tubules. Exogenously expressed TMEM33 suppresses the exogenously expressed reticulon 4C-induced tubulation of ER. These results suggest that TMEM33 has a potency to suppress the membrane-shaping activity of reticulons, thereby regulating the tubular structure of ER.


Subject(s)
Carrier Proteins/analysis , Membrane Proteins/analysis , Myelin Proteins/metabolism , Animals , Brain Chemistry , Carrier Proteins/metabolism , Chromatography, Affinity , Cloning, Molecular , Endoplasmic Reticulum/chemistry , Gene Expression , HeLa Cells , Humans , Immunosorbent Techniques , Membrane Proteins/chemistry , Membrane Proteins/genetics , Myelin Proteins/genetics , Myelin Proteins/physiology , Nogo Proteins , Nuclear Envelope/chemistry , Recombinant Fusion Proteins , Swine , Transfection
19.
Sheng Li Xue Bao ; 65(4): 445-50, 2013 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-23963076

ABSTRACT

Nogo-B is a major family member of the reticulon protein family 4. It is widely expressed in the central nervous system and peripheral tissues, and is mainly located in endoplasmic reticulum and cell membrane. Previous studies have revealed that Nogo-B plays a key role in vascular injury, tissue repair and inflammation process. It also may be critical for apoptosis of tumor cells and central diseases. Further investigation of the molecular characteristics and biological function of Nogo-B might be of great help to understand its role in diverse diseases.


Subject(s)
Myelin Proteins/physiology , Animals , Apoptosis , Cell Membrane/physiology , Endoplasmic Reticulum/physiology , Humans , Inflammation , Nogo Proteins
20.
Trends Neurosci ; 36(6): 363-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23642707

ABSTRACT

Growth inhibitory molecules in the adult mammalian central nervous system (CNS) have been implicated in the blocking of axonal sprouting and regeneration following injury. Prominent CNS regeneration inhibitors include Nogo-A, oligodendrocyte myelin glycoprotein (OMgp), and chondroitin sulfate proteoglycans (CSPGs), and a key question concerns their physiological role in the naïve CNS. Emerging evidence suggests novel functions in dendrites and at synapses of glutamatergic neurons. CNS regeneration inhibitors target the neuronal actin cytoskeleton to regulate dendritic spine maturation, long-term synapse stability, and Hebbian forms of synaptic plasticity. This is accomplished in part by antagonizing plasticity-promoting signaling pathways activated by neurotrophic factors. Altered function of CNS regeneration inhibitors is associated with mental illness and loss of long-lasting memory, suggesting unexpected and novel physiological roles for these molecules in brain health.


Subject(s)
Nerve Regeneration/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Amnesia/physiopathology , Animals , Astrocytes/physiology , Brain/growth & development , Brain/physiology , Brain Injuries/physiopathology , Brain Injuries/therapy , Chondroitin Sulfate Proteoglycans/physiology , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Dendrites/physiology , Dendrites/ultrastructure , Dominance, Ocular/physiology , GPI-Linked Proteins/physiology , Glycosaminoglycans/physiology , Humans , Mental Disorders/physiopathology , Mice , Models, Neurological , Myelin Proteins/physiology , Nogo Proteins , Nogo Receptor 1 , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...