Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 109
1.
Int J Biol Sci ; 17(12): 3145-3157, 2021.
Article En | MEDLINE | ID: mdl-34421356

Gastric Cancer (GC) is one of the main causes leading to death. PMP22, as a member of the GAS3 family of tetraspan proteins, it is associated with a variety of neurological diseases. Recently, more and more studies have shown that PMP22 play a great role in the physiological processes such as cells adhesion, migration, proliferation and tumorigenesis, but the involvement and functional mechanisms of PMP22 in Gastric carcinoma are not investigated clearly. In this study, we found that the PMP22 was overexpressed in the GC cells and tissue. Knockdown of PMP22 inhibits cell growth. Over-expressed PMP22 inhibits the etoposide-induced apoptosis, meanwhile knockdown of PMP22 promotes the etoposide-induced proliferation suppression, and increases cell apoptosis in GC cells. Furthermore, PMP22 enhanced the inhibition of the p53 transcriptional activities and down-regulated the p53 targeting genes, including p21, BAX and PUMA with or without treatment of etoposide. Finally, our results showed that PMP22 reduced the etoposide-induced tumor growth suppression in nude mice. Taken together, our research provided an anti-apoptotic properties alternative mechanism for PMP22 in gastric carcinoma and suggested PMP22 can be a potential target for the treatment of gastric cancer.


Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Etoposide/pharmacology , Myelin Proteins/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/physiology , Humans , Lentivirus/genetics , Male , Mice , Middle Aged , Myelin Proteins/genetics , Plasmids , RNA Interference , Real-Time Polymerase Chain Reaction , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transfection , Xenograft Model Antitumor Assays
2.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article En | MEDLINE | ID: mdl-34205075

Inherited neuropathies known as Charcot-Marie-Tooth (CMT) disease are genetically heterogeneous disorders affecting the peripheral nerves, causing significant and slowly progressive disability over the lifespan. The discovery of their diverse molecular genetic mechanisms over the past three decades has provided the basis for developing a wide range of therapeutics, leading to an exciting era of finding treatments for this, until now, incurable group of diseases. Many treatment approaches, including gene silencing and gene replacement therapies, as well as small molecule treatments are currently in preclinical testing while several have also reached clinical trial stage. Some of the treatment approaches are disease-specific targeted to the unique disease mechanism of each CMT form, while other therapeutics target common pathways shared by several or all CMT types. As promising treatments reach the stage of clinical translation, optimal outcome measures, novel biomarkers and appropriate trial designs are crucial in order to facilitate successful testing and validation of novel treatments for CMT patients.


Charcot-Marie-Tooth Disease/therapy , Genetic Therapy , Myelin P0 Protein/genetics , Myelin Proteins/genetics , Charcot-Marie-Tooth Disease/genetics , Gene Silencing , Humans , Mutation/genetics , Myelin P0 Protein/antagonists & inhibitors , Myelin Proteins/antagonists & inhibitors , Peripheral Nerves/metabolism , Peripheral Nerves/pathology
3.
Nat Commun ; 12(1): 2356, 2021 04 21.
Article En | MEDLINE | ID: mdl-33883545

Charcot-Marie-Tooth disease 1 A (CMT1A) results from a duplication of the PMP22 gene in Schwann cells and a deficit of myelination in peripheral nerves. Patients with CMT1A have reduced nerve conduction velocity, muscle wasting, hand and foot deformations and foot drop walking. Here, we evaluate the safety and efficacy of recombinant adeno-associated viral vector serotype 9 (AAV2/9) expressing GFP and shRNAs targeting Pmp22 mRNA in animal models of Charcot-Marie-Tooth disease 1 A. Intra-nerve delivery of AAV2/9 in the sciatic nerve allowed widespread transgene expression in resident myelinating Schwann cells in mice, rats and non-human primates. A bilateral treatment restore expression levels of PMP22 comparable to wild-type conditions, resulting in increased myelination and prevention of motor and sensory impairments over a twelve-months period in a rat model of CMT1A. We observed limited off-target transduction and immune response using the intra-nerve delivery route. A combination of previously characterized human skin biomarkers is able to discriminate between treated and untreated animals, indicating their potential use as part of outcome measures.


Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/therapy , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/genetics , Animals , Charcot-Marie-Tooth Disease/pathology , Dependovirus/genetics , Disease Models, Animal , Female , Gene Silencing , Genetic Therapy/methods , Genetic Vectors , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Rats , Rats, Mutant Strains , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
4.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article En | MEDLINE | ID: mdl-32751444

Nogo-A, a glycoprotein expressed in oligodendrocytes and central nervous system myelin, inhibits regeneration after injury. Antibodies against Nogo-A neutralize this inhibitory activity, improve locomotor recovery in spinal cord-injured adult mammals, and promote regrowth/sprouting/saving of damaged axons beyond the lesion site. Nogo-A is also expressed by neurons. Complete ablation of Nogo-A in all cell types expressing it has been found to lead to recovery in some studies but not in others. Neuronal ablation of Nogo-A reduces axonal regrowth after injury. In view of these findings, we hypothesized that, in addition to neutralizing Nogo-A in oligodendrocytes and myelin, Nogo-A antibodies may act directly on neuronal Nogo-A to trigger neurite outgrowth and neuronal survival. Here, we show that polyclonal and monoclonal antibodies against Nogo-A enhance neurite growth and survival of cultured cerebellar granule neurons and increase expression of the neurite outgrowth-promoting L1 cell adhesion molecule and polysialic acid. Application of inhibitors of signal transducing molecules, such as c-src, c-fyn, protein kinase A, and casein kinase II reduce antibody-triggered neurite outgrowth. These observations indicate that the recovery-promoting functions of antibodies against Nogo-A may not only be due to neutralizing Nogo-A in oligodendrocytes and myelin, but also to their interactions with Nogo-A on neurons.


Antibodies, Monoclonal/metabolism , Cell Survival , Myelin Proteins/metabolism , Neurites/metabolism , Neuronal Outgrowth , Oligodendroglia/metabolism , Peptide Fragments/metabolism , Animals , Cells, Cultured , Central Nervous System/injuries , Female , Male , Mice , Myelin Proteins/antagonists & inhibitors , Nerve Regeneration , Neurites/pathology , Oligodendroglia/pathology , Peptide Fragments/antagonists & inhibitors
5.
Mar Biotechnol (NY) ; 21(1): 52-64, 2019 Feb.
Article En | MEDLINE | ID: mdl-30443836

The protein level of muscle-specific human NogoA is abnormally upregulated in amyotrophic lateral sclerosis (ALS) mice and patients. On the other hand, while the presence of miR-206 in muscle cells delays onset and death in ALS, the relationship between these two phenomena remains unclear. Mammalian NogoA protein, also known as Reticulon 4a (Rtn4a), plays an important role in inhibiting the outgrowth of motor neurons. Our group previously identified zebrafish rtn4al as the target gene of miR-206 and found that knockdown of miR-206 increases rtn4al mRNA and Rtn4al protein in zebrafish embryos. It can be concluded from these results that neurite outgrowth of motor neurons is inhibited by Rtn4a1, which is entirely consistent with overexpression of either human NogoA or zebrafish homolog Rtn4al. Since an animal model able to express NogoA/rtn4al at the mature stage is unavailable, we generated a zebrafish transgenic line, Tg(Zα:TetON-Rtn4al), which conditionally and specifically overexpresses Rtn4al in the muscle tissue. After doxycycline induction, adult zebrafish displayed denervation at neuromuscular junction during the first week, then muscle disintegration and split myofibers during the third week, and, finally, significant weight loss in the sixth week. These results suggest that this zebrafish transgenic line, representing the inducible overexpression of Rtn4a1 in muscle, may provide an alternative animal model with which to study ALS because it exhibits ALS-like phenotype.


Amyotrophic Lateral Sclerosis/genetics , Disease Models, Animal , Motor Neurons/metabolism , Myelin Proteins/genetics , Neuromuscular Junction/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Animals, Genetically Modified , Doxycycline/pharmacology , Embryo, Nonmammalian , Gene Expression Regulation/drug effects , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Morpholinos/genetics , Morpholinos/metabolism , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myelin Proteins/agonists , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/metabolism , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Nogo Proteins/agonists , Nogo Proteins/genetics , Nogo Proteins/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Phenotype , Plasmids/chemistry , Plasmids/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/agonists , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
6.
J Clin Invest ; 128(1): 359-368, 2018 01 02.
Article En | MEDLINE | ID: mdl-29202483

Charcot-Marie-Tooth disease type 1A (CMT1A) is caused by duplication of peripheral myelin protein 22 (PMP22) and is the most common hereditary peripheral neuropathy. CMT1A is characterized by demyelination and axonal loss, which underlie slowed motor nerve conduction velocity (MNCV) and reduced compound muscle action potentials (CMAP) in patients. There is currently no known treatment for this disease. Here, we show that antisense oligonucleotides (ASOs) effectively suppress PMP22 mRNA in affected nerves in 2 murine CMT1A models. Notably, initiation of ASO treatment after disease onset restored myelination, MNCV, and CMAP almost to levels seen in WT animals. In addition to disease-associated gene expression networks that were restored with ASO treatment, we also identified potential disease biomarkers through transcriptomic profiling. Furthermore, we demonstrated that reduction of PMP22 mRNA in skin biopsies from ASO-treated rats is a suitable biomarker for evaluating target engagement in response to ASO therapy. These results support the use of ASOs as a potential treatment for CMT1A and elucidate potential disease and target engagement biomarkers for use in future clinical trials.


Action Potentials/drug effects , Charcot-Marie-Tooth Disease/drug therapy , Motor Neurons/metabolism , Myelin Proteins/antagonists & inhibitors , Oligodeoxyribonucleotides, Antisense/pharmacology , Skin/metabolism , Action Potentials/genetics , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Motor Neurons/pathology , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Skin/pathology
7.
Mol Cancer Ther ; 16(6): 1187-1198, 2017 06.
Article En | MEDLINE | ID: mdl-28336807

Cancer stem cells possess self-renewal and chemoresistance activities. However, the manner in which these features are maintained remains obscure. We sought to identify cell surface protein(s) that mark self-renewing and chemoresistant gastric cancer cells using the explorer antibody microarray. We identified PMP22, a target gene of the Wnt/ß-catenin pathway, as the most upregulated cell surface protein in gastric cancer xenografts exposed to cisplatin (DDP). PMP22 expression was markedly upregulated in tumorspheric cells and declined with differentiation. Infecting gastric cancer cells with lentivirus expressing PMP22 shRNAs reduced proliferation, tumorsphere formation, and chemoresistance to cisplatin in vitro and in NOD/SCID mice. When combined with bortezomib, a PMP22 inhibitor, the chemotherapeutic sensitivity to cisplatin treatment was dramatically increased by inducing cell apoptosis in cultured cells and xenograft mouse models. Finally, mRNA expression levels of PMP22 were detected in 38 tumor specimens from patients who received six cycles of perioperative chemotherapy. A strong correlation between PMP22 level and tumor recurrence was revealed, thus showing a pivotal role of PMP22 in the clinical chemoresistance of gastric cancer. Our study is the first to show the role of PMP22 in gastric cancer stemness and chemoresistance and reveals a potential new target for the diagnosis and treatment of recurrent gastric cancer. Mol Cancer Ther; 16(6); 1187-98. ©2017 AACR.


Antineoplastic Agents/pharmacology , Cell Self Renewal/genetics , Drug Resistance, Neoplasm/genetics , Myelin Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Stomach Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Humans , Male , Mice , Molecular Targeted Therapy , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/metabolism , Neoplasm Staging , RNA Interference , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Tumor Burden , Xenograft Model Antitumor Assays
8.
Mol Cell Neurosci ; 71: 80-91, 2016 Mar.
Article En | MEDLINE | ID: mdl-26711806

The Nogo-66 receptor (NgR1), a receptor for Nogo-A, contributes to the inhibition of axonal regeneration in the adult central nervous system after traumatic injuries. Thus, NgR1 has been considered a critical target in axon regeneration therapy. Here, we identified a specific NgR1 antagonist peptide (HIYTALV, named NAP2) which promotes neurite regeneration in vitro from a phage display heptapeptide library. NAP2 was co-localized with NgR1 on the surface of PC12 cells and cerebellar granule cells (CGCs) by immunofluorescence assay. Horseradish peroxidase (HRP)-streptavidin-biotin assay further showed that NAP2 binds to NgR1 and the dissociation constant (Kd) was 0.45 µM Functional analyses indicated that NAP2 could reduce the inhibitory effects of Nogo-66 on neurite outgrowth in differentiated PC12 cells and CGCs by blocking the Nogo-66-induced activation of Rho-associated coiled coil-containing protein kinase (ROCK), collapsin response mediator protein 2 (CRMP2) and myosin light chain (MLC). Taken together, the small molecule NgR1 antagonist peptide NAP2 (MW: 815.98Da) has a potential ability in crossing blood brain barrier and will be a promising therapeutic agent for the treatment of spinal cord injury and neurodegenerative diseases.


Myelin Proteins/antagonists & inhibitors , Nerve Regeneration , Neurites/drug effects , Oligopeptides/pharmacology , Animals , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Myelin Proteins/metabolism , Myosin Light Chains/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurites/physiology , Nogo Proteins , PC12 Cells , Protein Binding , Rats , Rats, Sprague-Dawley , rho-Associated Kinases/metabolism
9.
Expert Rev Neurother ; 16(2): 173-86, 2016.
Article En | MEDLINE | ID: mdl-26689223

Over recent decades, experimental and clinical stroke studies have identified a number of neurorestorative treatments that stimulate neural plasticity and promote functional recovery. In contrast to the acute stroke treatments thrombolysis and endovascular thrombectomy, neurorestorative treatments are still effective when initiated days after stroke onset, which makes them applicable to virtually all stroke patients. In this article, selected physical, pharmacological and cell-based neurorestorative therapies are discussed, with special emphasis on interventions that have already been transferred from the laboratory to the clinical setting. We explain molecular and structural processes that promote neural plasticity, discuss potential limitations of neurorestorative treatments, and offer a speculative viewpoint on how neurorestorative treatments will evolve.


Brain Ischemia/rehabilitation , Neurological Rehabilitation/methods , Neuronal Plasticity , Nootropic Agents/therapeutic use , Regeneration , Selective Serotonin Reuptake Inhibitors/therapeutic use , Stem Cell Transplantation/methods , Stroke Rehabilitation , Axons , Brain/physiology , Brain/physiopathology , Brain Ischemia/complications , Cytidine Diphosphate Choline/therapeutic use , Erythropoietin/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Myelin Proteins/antagonists & inhibitors , Neovascularization, Physiologic , Neurogenesis , Nogo Proteins , Phosphodiesterase 5 Inhibitors/therapeutic use , Recovery of Function , Stroke/etiology , Stroke/physiopathology
10.
Biomed Res Int ; 2015: 817914, 2015.
Article En | MEDLINE | ID: mdl-26583134

Parkinson's disease (PD) is a common degenerative disease that lacks efficient treatment. Myelin-associated neurite outgrowth inhibitor A (Nogo-A) is relevant with inhibition of nerve regeneration and may play vital role in pathogenesis of PD. The study aimed to establish the shRNA expression plasmids of Nogo-A gene and explore the regulatory effects of Nogo-A silencing on the expression of inflammation factor tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) as well as tyrosine hydroxylase (TH) in lipopolysaccharide- (LPS-) stimulated rat PC12 cells. The results showed that both mRNA and protein levels of Nogo-A in pGenesil-nogoA-shRNA group were downregulated. The viabilities of PC12 cells decreased with increase of LPS concentrations. LPS significantly increased the supernatant TNF-alpha and IL-6 concentrations and reduced TH protein expression in PC12 cells, while silencing Nogo-A could block these effects. These results suggested that LPS can activate PC12 cells to secrete inflammatory cytokines and lower the TH expression, which can be regulated by Nogo-A gene silencing. Nogo-A silencing might provide new ideas for PD treatment in the future.


Inflammation/genetics , Interleukin-6/metabolism , Myelin Proteins/genetics , Parkinson Disease/genetics , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/biosynthesis , Animals , Gene Expression Regulation/drug effects , Gene Silencing , Humans , Inflammation/chemically induced , Inflammation/pathology , Interleukin-6/genetics , Lipopolysaccharides/toxicity , Myelin Proteins/antagonists & inhibitors , Nerve Regeneration/genetics , Nogo Proteins , PC12 Cells , Parkinson Disease/pathology , Parkinson Disease/therapy , RNA, Messenger/biosynthesis , Rats , Tumor Necrosis Factor-alpha/genetics , Tyrosine 3-Monooxygenase/genetics
11.
Sci Rep ; 5: 12061, 2015 Jul 15.
Article En | MEDLINE | ID: mdl-26174362

Nogo-B, a member of the reticulon 4 protein family, plays a critical role in tissue repair and acute inflammation. Its role in acute lung injury (ALI) remains unclear. Here, we assessed the function of Nogo-B during tissue injury in a lipopolysaccharide (LPS)-induced ALI mouse model. We found that pulmonary Nogo-B was significantly repressed after LPS instillation in C57BL/6 mice. Over-expression of pulmonary Nogo-B using an adenovirus vector carrying the Nogo-B-RFP-3flag gene (Ad-Nogo-B) significantly prolonged the survival of mice challenged with a lethal dose of LPS. The Ad-Nogo-B-treated mice also had less severe lung injury, less alveolar protein exudation, and a higher number of macrophages but less neutrophil infiltration compared with Ad-RFP-treated mice. Interestingly, microarray analysis showed that the Ad-Nogo-B-treated mice had different gene expression profiles compared with the controls and the prominent expression of genes related to wound healing and the humoral immune response after LPS induction. Of the 49 differently expressed genes, we found that the expression of PTX3 was significantly up-regulated following Nogo-B over-expression as observed in lung tissues and RAW264.7 cells. In conclusion, Nogo-B plays a protective role against LPS-induced ALI, and this effect might be exerted through the modulation of alveolar macrophage recruitment and PTX3 production.


Acute Lung Injury/pathology , Lipopolysaccharides/toxicity , Myelin Proteins/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/mortality , Adenoviridae/genetics , Animals , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Cell Line , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/metabolism , Immunity, Humoral , Lipopolysaccharides/immunology , Lung/metabolism , Lung/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/genetics , Nogo Proteins , RNA Interference , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/metabolism , Survival Rate , Transcriptome
12.
Neural Dev ; 10: 6, 2015 Mar 20.
Article En | MEDLINE | ID: mdl-25888884

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.


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
14.
Cell Physiol Biochem ; 35(2): 616-26, 2015.
Article En | MEDLINE | ID: mdl-25612921

BACKGROUND: Nogo-A, a major myelin-associated inhibitor, can inhibit injured optic nerve regeneration. However, whether Amino-Nogo is the most important functional domain of Nogo-A remains unknown. This study aimed to identify the role of Amino-Nogo following optic nerve injury, and the mechanism of the Amino-Nogo-integrin αv signaling pathway in vivo. METHODS: Sprague-Dawley rats with optic nerve crush injury were injected with Nogo-A siRNA (Nogo-A-siRNA), the Nogo-66 functional domain antagonist peptide of Nogo-A (Nep1-40) or a recombinant rat Amino-Nogo-A protein (∆20) into the vitreous cavity to knock down Nogo-A, inhibit Nogo-66 or activate the Amino-Nogo, resparately. Retinal ganglion cell (RGC) density, axon regeneration and the pattern of NPN of visual electrophysiology (flash visual evoked potentials [F-VEP]) at different times post-injury were investigated. RESULTS: Our study revealed a lower RGC survival rate; shorter axonal outgrowth; longer N1, P1 and N2 waves latencies; and lower N1-P1 and P1-N2 amplitudes in the Δ20 group, and Δ20 treatment significantly attenuated integrin αv expression and phosphorylated focal adhesion kinase (p-FAK) levels. In the Nep1-40 and Nogo-A siRNA groups, there were higher RGC survival rates, longer axonal outgrowth, shorter N1 and P1 wave latencies, and higher N1-P1 and P1-N2amplitudes. Nogo-A siRNA treatment significantly increased integrin αv expression and p-FAK levels. Nepl-40 treatment did not alter integrin αv expression. In addition, there was no significant change in integrin α5 in any group. CONCLUSION: These results suggest that the integrin signaling pathway is regulated by Amino-Nogo, which inhibits optic nerve regeneration and functional recovery, and that the integrin subunit involved might be integrin αv but not integrin α5.


Integrin alphaV/metabolism , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/chemistry , Nerve Regeneration , Optic Nerve/physiopathology , Signal Transduction , Animals , Evoked Potentials, Visual , Gene Knockdown Techniques , Myelin Proteins/metabolism , Nogo Proteins , Optic Nerve/cytology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/physiopathology , Peptide Fragments/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/metabolism
16.
J Neurochem ; 132(1): 70-84, 2015 Jan.
Article En | MEDLINE | ID: mdl-25314656

Axonal regeneration after injury to the CNS is hampered by myelin-derived inhibitors, such as Nogo-A. Natural products, such as green tea, which are neuroprotective and safe for long-term therapy, would complement ongoing various pharmacological approaches. In this study, using nerve growth factor-differentiated neuronal-like Neuroscreen-1 cells, we show that extremely low concentrations of unfractionated green tea polyphenol mixture (GTPP) and its active ingredient, epigallocatechin-3-gallate (EGCG), prevent both the neurite outgrowth-inhibiting activity and growth cone-collapsing activity of Nogo-66 (C-terminal domain of Nogo-A). Furthermore, a synergistic interaction was observed among GTPP constituents. This preventive effect was dependent on 67-kDa laminin receptor (67LR) to which EGCG binds with high affinity. The antioxidants N-acetylcysteine and cell-permeable catalase abolished this preventive effect of GTPP and EGCG, suggesting the involvement of sublethal levels of H2 O2 in this process. Accordingly, exogenous sublethal concentrations of H2 O2 , added as a bolus dose (5 µM) or more effectively through a steady-state generation (1-2 µM), mimicked GTPP in counteracting the action of Nogo-66. Exogenous H2 O2 mediated this action by bypassing the requirement of 67LR. Taken together, these results show for the first time that GTPP and EGCG, acting through 67LR and elevating intracellular sublethal levels of H2 O2 , inhibit the antineuritogenic action of Nogo-A. Currently, several agents are being evaluated for overcoming axonal growth inhibitors to promote functional recovery after stroke and spinal cord injury. Epigallocatechin-3-gallate (EGCG), present in green tea polyphenol mixture (GTPP), prevents antineuritogenic activity of Nogo-A, a myelin-derived axonal growth inhibitor. The preventive action of EGCG involves the cell-surface-associated 67-kDa laminin receptor and H2 O2 . GTPP may complement ongoing efforts to treat neuronal injuries.>


Hydrogen Peroxide/pharmacology , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/pharmacology , Neurites/drug effects , Oxidants/pharmacology , Polyphenols/pharmacology , Receptors, Laminin/drug effects , Tea/chemistry , Animals , Cells, Cultured , Growth Cones/drug effects , Mice , Nogo Proteins , Polyphenols/chemistry , Pseudopodia/drug effects
17.
JAMA Neurol ; 72(2): 176-9, 2015 Feb.
Article En | MEDLINE | ID: mdl-25437093

IMPORTANCE: Although multiple sclerosis (MS) is generally considered an autoimmune demyelinating disorder of the central nervous system, axonal degeneration through Nogo receptor-1 signaling was recently recognized as an important pathological feature. Our previous identification of lateral olfactory tract usher substance (LOTUS), an endogenous Nogo receptor-1 antagonist, prompted us to analyze the relationship between LOTUS levels of cerebrospinal fluid and the clinical course of MS to evaluate whether LOTUS could be a useful biomarker for MS. OBJECTIVE: To examine variations in LOTUS concentrations in the cerebrospinal fluid of patients with MS in accordance with their clinical course. DESIGN, SETTING, AND PARTICIPANTS: Cerebrospinal fluid samples were obtained retrospectively from normal controls (NCs; n = 27) and patients with MS (n = 40), amyotrophic lateral sclerosis (n = 22), and multiple system atrophy (n = 10) between January 1, 2008, and January 1, 2014. Patients with MS were divided into relapsing-remitting MS (RRMS; n = 30) and secondary progressive MS (n = 10). Patients with RRMS were further divided into relapse and remission groups. MAIN OUTCOMES AND MEASURES: The LOTUS concentration in cerebropsinal fluid was quantitatively detected by immunoblotting using a specific LOTUS antibody and the concentrations compared in accordance with the patients' clinical course, such as remission and relapse groups in RRMS and secondary progressive MS. RESULTS: The mean (SD) cerebrospinal fluid LOTUS concentration in the relapse group of RRMS (9.3 [3.6] µg/dL) was lower than that of NCs (19.2 [4.7] µg/dL; P < .001) whereas the level in the remission group of RRMS (19.6 [5.8] µg/dL) was similar to that of NCs. The LOTUS concentration in SPMS (6.7 [1.4] µg/dL; P < .001) was lower than that of NCs and the remission group of RRMS. The LOTUS levels in other neurodegenerative diseases, such as amyotrophic lateral sclerosis and multiple system atrophy, were normal. CONCLUSIONS AND RELEVANCE: Variations in LOTUS concentrations were correlated with disease activity in MS. Therefore, LOTUS concentration may be useful as a possible biomarker for MS. Low LOTUS concentrations may be possibly involved in Nogo receptor-1 signaling, which may induce axonal degeneration in the relapse phase of RRMS and secondary progressive MS.


Biomarkers/cerebrospinal fluid , Multiple Sclerosis, Chronic Progressive/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Myelin Proteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Adult , Aged , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Cerebrospinal Fluid/chemistry , GPI-Linked Proteins/antagonists & inhibitors , Humans , Male , Middle Aged , Multiple System Atrophy/cerebrospinal fluid , Nogo Receptor 1
18.
Orphanet J Rare Dis ; 9: 201, 2014 Dec 10.
Article En | MEDLINE | ID: mdl-25491744

Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common inherited sensory and motor peripheral neuropathy. It is caused by PMP22 overexpression which leads to defects of peripheral myelination, loss of long axons, and progressive impairment then disability. There is no treatment available despite observations that monotherapeutic interventions slow progression in rodent models. We thus hypothesized that a polytherapeutic approach using several drugs, previously approved for other diseases, could be beneficial by simultaneously targeting PMP22 and pathways important for myelination and axonal integrity. A combination of drugs for CMT1A polytherapy was chosen from a group of authorised drugs for unrelated diseases using a systems biology approach, followed by pharmacological safety considerations. Testing and proof of synergism of these drugs were performed in a co-culture model of DRG neurons and Schwann cells derived from a Pmp22 transgenic rat model of CMT1A. Their ability to lower Pmp22 mRNA in Schwann cells relative to house-keeping genes or to a second myelin transcript (Mpz) was assessed in a clonal cell line expressing these genes. Finally in vivo efficacy of the combination was tested in two models: CMT1A transgenic rats, and mice that recover from a nerve crush injury, a model to assess neuroprotection and regeneration. Combination of (RS)-baclofen, naltrexone hydrochloride and D-sorbitol, termed PXT3003, improved myelination in the Pmp22 transgenic co-culture cellular model, and moderately down-regulated Pmp22 mRNA expression in Schwannoma cells. In both in vitro systems, the combination of drugs was revealed to possess synergistic effects, which provided the rationale for in vivo clinical testing of rodent models. In Pmp22 transgenic CMT1A rats, PXT3003 down-regulated the Pmp22 to Mpz mRNA ratio, improved myelination of small fibres, increased nerve conduction and ameliorated the clinical phenotype. PXT3003 also improved axonal regeneration and remyelination in the murine nerve crush model. Based on these observations in preclinical models, a clinical trial of PTX3003 in CMT1A, a neglected orphan disease, is warranted. If the efficacy of PTX3003 is confirmed, rational polytherapy based on novel combinations of existing non-toxic drugs with pleiotropic effects may represent a promising approach for rapid drug development.


Axons/metabolism , Charcot-Marie-Tooth Disease/metabolism , Disease Models, Animal , Drug Repositioning/methods , Myelin Proteins/biosynthesis , Nerve Fibers, Myelinated/metabolism , Animals , Axons/drug effects , Axons/pathology , Baclofen/administration & dosage , Charcot-Marie-Tooth Disease/drug therapy , Charcot-Marie-Tooth Disease/pathology , Coculture Techniques , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Therapy, Combination , Female , Gene Expression Regulation , Male , Mice , Myelin Proteins/antagonists & inhibitors , Naltrexone/administration & dosage , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/pathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Sorbitol/administration & dosage
19.
PLoS One ; 9(9): e106378, 2014.
Article En | MEDLINE | ID: mdl-25184636

BACKGROUND: Inhibitory factors have been implicated in the failure of remyelination in demyelinating diseases. Myelin associated inhibitors act through a common receptor called Nogo receptor (NgR) that plays critical inhibitory roles in CNS plasticity. Here we investigated the effects of abrogating NgR inhibition in a non-immune model of focal demyelination in adult mouse optic chiasm. METHODOLOGY/PRINCIPAL FINDINGS: A focal area of demyelination was induced in adult mouse optic chiasm by microinjection of lysolecithin. To knock down NgR levels, siRNAs against NgR were intracerebroventricularly administered via a permanent cannula over 14 days, Functional changes were monitored by electrophysiological recording of latency of visual evoked potentials (VEPs). Histological analysis was carried out 3, 7 and 14 days post demyelination lesion. To assess the effect of NgR inhibition on precursor cell repopulation, BrdU was administered to the animals prior to the demyelination induction. Inhibition of NgR significantly restored VEPs responses following optic chiasm demyelination. These findings were confirmed histologically by myelin specific staining. siNgR application resulted in a smaller lesion size compared to control. NgR inhibition significantly increased the numbers of BrdU+/Olig2+ progenitor cells in the lesioned area and in the neurogenic zone of the third ventricle. These progenitor cells (Olig2+ or GFAP+) migrated away from this area as a function of time. CONCLUSIONS/SIGNIFICANCE: Our results show that inhibition of NgR facilitate myelin repair in the demyelinated chiasm, with enhanced recruitment of proliferating cells to the lesion site. Thus, antagonizing NgR function could have therapeutic potential for demyelinating disorders such as Multiple Sclerosis.


Demyelinating Diseases/therapy , Myelin Proteins/antagonists & inhibitors , Myelin Sheath/metabolism , Optic Chiasm/metabolism , Receptors, Cell Surface/genetics , Recovery of Function , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodeoxyuridine/administration & dosage , Cell Movement , Cell Proliferation , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Evoked Potentials, Visual , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation , Injections, Intraventricular , Lysophosphatidylcholines , Male , Mice , Mice, Inbred C57BL , Microinjections , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Sheath/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nogo Receptor 1 , Oligodendrocyte Transcription Factor 2 , Optic Chiasm/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Time Factors
20.
Mol Cells ; 37(8): 613-9, 2014 Aug.
Article En | MEDLINE | ID: mdl-25134537

The optic nerve often suffers regenerative failure after injury, leading to serious visual impairment such as glaucoma. The main inhibitory factors, including Nogo-A, oligodendrocyte myelin glycoprotein, and myelin-associated glycoprotein, exert their inhibitory effects on axonal growth through the same receptor, the Nogo-66 receptor (NgR). Oncomodulin (OM), a calcium-binding protein with a molecular weight of an ∼12 kDa, which is secreted from activated macrophages, has been demonstrated to have high and specific affinity for retinal ganglion cells (RGC) and promote greater axonal regeneration than other known polypeptide growth factors. Protamine has been reported to effectively deliver small interference RNA (siRNA) into cells. Accordingly, a fusion protein of OM and truncated protamine (tp) may be used as a vehicle for the delivery of NgR siRNA into RGC for gene therapy. To test this hypothesis, we constructed OM and tp fusion protein (OM/tp) expression vectors. Using the indirect immunofluorescence labeling method, OM/tp fusion proteins were found to have a high affinity for RGC. The gel shift assay showed that the OM/tp fusion proteins retained the capacity to bind to DNA. Using OM/tp fusion proteins as a delivery tool, the siRNA of NgR was effectively transfected into cells and significantly down-regulated NgR expression levels. More importantly, OM/tp-NgR siRNA dramatically promoted axonal growth of RGC compared with the application of OM/tp recombinant protein or NgR siRNA alone in vitro. In addition, OM/tp-NgR siRNA highly elevated intracellular cyclic adenosine monophosphate (cAMP) levels and inhibited activation of the Ras homolog gene family, member A (RhoA). Taken together, our data demonstrated that the recombinant OM/tp fusion proteins retained the functions of both OM and tp, and that OM/tp-NgR siRNA might potentially be used for the treatment of optic nerve injury.


Axons/drug effects , Calcium-Binding Proteins/genetics , Gene Transfer Techniques , Myelin Proteins/antagonists & inhibitors , Protamines/genetics , Receptors, Cell Surface/antagonists & inhibitors , Retinal Ganglion Cells/drug effects , Animals , Axons/metabolism , Calcium-Binding Proteins/metabolism , Cyclic AMP/metabolism , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation , Myelin Proteins/genetics , Myelin Proteins/metabolism , Nerve Regeneration/drug effects , Nogo Receptor 1 , Plasmids/chemistry , Primary Cell Culture , Protamines/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Inbred F344 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Ganglion Cells/metabolism , Signal Transduction , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
...