Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Virol ; 96(8): e0005522, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35353001

RESUMEN

Engagement of host receptors is essential for viruses to enter target cells and initiate infection. Expression patterns of receptors in turn dictate host range, tissue tropism, and disease pathogenesis during infection. Mammalian orthoreovirus (reovirus) displays serotype-dependent patterns of tropism in the murine central nervous system (CNS) that are dictated by the viral attachment protein σ1. However, the receptor that mediates reovirus CNS tropism is unknown. Two proteinaceous receptors have been identified for reovirus, junctional adhesion molecule A (JAM-A) and Nogo-66 receptor 1 (NgR1). Engagement of JAM-A is required for reovirus hematogenous dissemination but is dispensable for neural spread and infection of the CNS. To determine whether NgR1 functions in reovirus neuropathogenesis, we compared virus replication and disease in wild-type (WT) and NgR1-/- mice. Genetic ablation of NgR1 did not alter reovirus replication in the intestine or transmission to the brain following peroral inoculation. Viral titers in neural tissues following intramuscular inoculation, which provides access to neural dissemination routes, also were comparable in WT and NgR1-/- mice, suggesting that NgR1 is dispensable for reovirus neural spread to the CNS. The absence of NgR1 also did not alter reovirus replication, neural tropism, and virulence following direct intracranial inoculation. In agreement with these findings, we found that the human but not the murine homolog of NgR1 functions as a receptor and confers efficient reovirus binding and infection of nonsusceptible cells in vitro. Thus, neither JAM-A nor NgR1 is required for reovirus CNS tropism in mice, suggesting that other unidentified receptors support this function. IMPORTANCE Viruses engage diverse molecules on host cell surfaces to navigate barriers, gain cell entry, and establish infection. Despite discovery of several reovirus receptors, host factors responsible for reovirus neurotropism are unknown. Human NgR1 functions as a reovirus receptor in vitro and is expressed in CNS neurons in a pattern overlapping reovirus tropism. We used mice lacking NgR1 to test whether NgR1 functions as a reovirus neural receptor. Following different routes of inoculation, we found that murine NgR1 is dispensable for reovirus dissemination to the CNS, tropism and replication in the brain, and resultant disease. Concordantly, expression of human but not murine NgR1 confers reovirus binding and infection of nonsusceptible cells in vitro. These results highlight species-specific use of alternate receptors by reovirus. A detailed understanding of species- and tissue-specific factors that dictate viral tropism will inform development of antiviral interventions and targeted gene delivery and therapeutic viral vectors.


Asunto(s)
Receptor Nogo 1 , Reoviridae , Animales , Molécula A de Adhesión de Unión/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Reoviridae/metabolismo , Infecciones por Reoviridae/virología
2.
J Ethnopharmacol ; 289: 115018, 2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-35092824

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Gastrodia elata Blume (G. elata), a traditional Chinese herb, known as "Tian Ma", is widely used as a common medicine and diet ingredient for treating or preventing neurological disorders for thousands of years in China. However, the anti-depressant effect of G. elata and the underlying mechanism have not been fully evaluated. AIM OF THE STUDY: The study is aimed to investigate the anti-depressant effect and the molecular mechanism of G. elata in vitro and in vivo using PC12 cells and zebrafish model, respectively. MATERIAL AND METHODS: Network pharmacology was performed to explore the potential active ingredients and action targets of G. elata Blume extracts (GBE) against depression. The cell viability and proliferation were determined by MTT and EdU assay, respectively. TUNEL assay was used to examine the anti-apoptotic effect of GBE. Immunofluorescence and Western blot were used to detect the protein expression level. In addition, novel tank diving test was used to investigate the anti-depressant effect in zebrafish depression model. RT-PCR was used to analyze the mRNA expression levels of genes. RESULTS: G. elata against depression on the reticulon 4 receptors (RTN4R) and apoptosis-related targets, which were predicted by network pharmacology. Furthermore, GBE enhanced cell viability and inhibited the apoptosis in PC12 cells against CORT treatment. GBE relieved depression-like symptoms in adult zebrafish, included increase of exploratory behavior and regulation of depression related genes. Mechanism studies showed that the GBE inhibited the expression of RTN4R-related and apoptosis-related genes. CONCLUSION: Our studies show the ameliorative effect of G. elata against depression. The mechanism may be associated with the inhibition of RTN4R-related and apoptosis pathways.


Asunto(s)
Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Extractos Vegetales/farmacología , Animales , Antidepresivos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Gastrodia , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Farmacología en Red , Receptor Nogo 1/genética , Células PC12 , Ratas , Pez Cebra
3.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34848542

RESUMEN

Normally, dendritic size is established prior to adolescence and then remains relatively constant into adulthood due to a homeostatic balance between growth and retraction pathways. However, schizophrenia is characterized by accelerated reductions of cerebral cortex gray matter volume and onset of clinical symptoms during adolescence, with reductions in layer 3 pyramidal neuron dendritic length, complexity, and spine density identified in multiple cortical regions postmortem. Nogo receptor 1 (NGR1) activation of the GTPase RhoA is a major pathway restricting dendritic growth in the cerebral cortex. We show that the NGR1 pathway is stimulated by OMGp and requires the Rho guanine nucleotide exchange factor Kalirin-9 (KAL9). Using a genetically encoded RhoA sensor, we demonstrate that a naturally occurring missense mutation in Kalrn, KAL-PT, that was identified in a schizophrenia cohort, confers enhanced RhoA activitation in neuronal dendrites compared to wild-type KAL. In mice containing this missense mutation at the endogenous locus, there is an adolescent-onset reduction in dendritic length and complexity of layer 3 pyramidal neurons in the primary auditory cortex. Spine density per unit length of dendrite is unaffected. Early adult mice with these structural deficits exhibited impaired detection of short gap durations. These findings provide a neuropsychiatric model of disease capturing how a mild genetic vulnerability may interact with normal developmental processes such that pathology only emerges around adolescence. This interplay between genetic susceptibility and normal adolescent development, both of which possess inherent individual variability, may contribute to heterogeneity seen in phenotypes in human neuropsychiatric disease.


Asunto(s)
Corteza Cerebral/citología , Dendritas/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neuronas/fisiología , Transducción de Señal/fisiología , Animales , Sistemas CRISPR-Cas , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Genotipo , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ratones , Ratones Transgénicos , Mutación Missense , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Maduración Sexual
4.
Mol Biol Rep ; 48(8): 5955-5964, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34350550

RESUMEN

BACKGROUND: Vitiligo is a complex disease in which patchy depigmentation is the result of an autoimmune-induced loss of melanocytes in affected regions. On the basis of a genome-wide linkage analysis of vitiligo in the Chinese Han population, we previously showed significant evidence of a linkage between 22q12 and vitiligo. Our aim in the current study was to identify vitiligo susceptibility variants within an expanded region of the 22q12 locus. METHODS AND RESULTS: An in-depth analysis of the expanded region of the 22q12 locus was performed by imputation using a large GWAS dataset consisting of 1117 cases and 1701 controls. Eight nominal SNPs were selected and genotyped in an independent cohort of Chinese Han individuals (2069 patients and 1370 control individuals) by using the Sequenom MassArray iPLEX1 system. The data were analyzed with PLINK 1.07 software. The C allele of rs730669 located in ZDHHC8/RTN4R showed a strong association with vitiligo (P = 3.25 × 10-8, OR = 0.81). The C allele of rs4820338 located in VPREB1 and the A allele of rs2051582 (a SNP reported in our previous study) located in IL2RB showed a suggestive association with vitiligo (P = 1.04 × 10-5, OR = 0.86; P = 1.78 × 10-6, OR = 1.27). The three identified SNPs showed independent associations with vitiligo in a conditional logistic regression analysis (all P < 1.0 × 10-5; all D' < 0.05 and r2 < 1.0 × 10-4). CONCLUSIONS: The study reveals that two novel variants rs730669 (ZDHHC8/RTN4R) and rs4820338 (VPREB1) on 22q11.2 might confer susceptibility to vitiligo and affect disease subphenotypes. The presence of multiple independent variants emphasizes their important roles in the genetic pathogenesis of disease.


Asunto(s)
Cromosomas Humanos Par 22/genética , Vitíligo/genética , Aciltransferasas/genética , Adolescente , Adulto , Alelos , Pueblo Asiatico/genética , Estudios de Casos y Controles , China/epidemiología , Estudios de Cohortes , Etnicidad/genética , Femenino , Frecuencia de los Genes/genética , Ligamiento Genético/genética , Predisposición Genética a la Enfermedad , Variación Genética/genética , Estudio de Asociación del Genoma Completo/métodos , Genotipo , Humanos , Inmunoglobulina de Cadenas Ligeras Subrogadas/genética , Masculino , Proteínas de la Membrana/genética , Receptor Nogo 1/genética , Polimorfismo de Nucleótido Simple/genética , Adulto Joven
5.
Sci Rep ; 11(1): 2890, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536561

RESUMEN

We have previously reported evidence that Nogo-A activation of Nogo-receptor 1 (NgR1) can drive axonal dystrophy during the neurological progression of experimental autoimmune encephalomyelitis (EAE). However, the B-cell activating factor (BAFF/BlyS) may also be an important ligand of NgR during neuroinflammation. In the current study we define that NgR1 and its homologs may contribute to immune cell signaling during EAE. Meningeal B-cells expressing NgR1 and NgR3 were identified within the lumbosacral spinal cords of ngr1+/+ EAE-induced mice at clinical score 1. Furthermore, increased secretion of immunoglobulins that bound to central nervous system myelin were shown to be generated from isolated NgR1- and NgR3-expressing B-cells of ngr1+/+ EAE-induced mice. In vitro BAFF stimulation of NgR1- and NgR3-expressing B cells, directed them into the cell cycle DNA synthesis phase. However, when we antagonized BAFF signaling by co-incubation with recombinant BAFF-R, NgR1-Fc, or NgR3 peptides, the B cells remained in the G0/G1 phase. The data suggest that B cells express NgR1 and NgR3 during EAE, being localized to infiltrates of the meninges and that their regulation is governed by BAFF signaling.


Asunto(s)
Factor Activador de Células B/metabolismo , Linfocitos B/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Meninges/patología , Esclerosis Múltiple/inmunología , Animales , Linfocitos B/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Meninges/inmunología , Ratones , Ratones Noqueados , Esclerosis Múltiple/patología , Proteínas Nogo/metabolismo , Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Receptores Nogo/metabolismo
6.
Mol Med Rep ; 23(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33495810

RESUMEN

The generation of ß­amyloid protein (Aß) is considered a key step in the pathogenesis of Alzheimer's disease (AD) and the regulation of its production is an important therapeutic strategy. It was hypothesized in the present study that Nogo­A may be involved in AD and may regulate the generation of Aß. Nogo­A is known to act as a major inhibitor of neuron regeneration in the adult central nervous system. A recent study indicated that Nogo­A is associated with AD; however, the underlying effect and molecular mechanisms remain largely elusive. In the present study, the potential effects of Nogo­A on AD were investigated. ELISA was used to detect the levels of Aß, enzymatic activity detection kits were used to determine the activity of secretase enzymes in amyloid precursor protein (APP) metabolism, and western blot analysis was used to detect the expression levels of proteins associated with the APP processing and Nogo­A/Nogo­66 receptor (NgR) signaling pathways. The results revealed that Nogo­66, the major inhibitory region of Nogo­A, promoted neuronal Aß secretion by increasing the activity of ß­secretase 1 via the NgR/Rho­associated coiled­coil containing kinases pathway in a dose­dependent manner. The present data suggested that Nogo­A may facilitate the onset and development of AD by promoting Aß secretion, providing information on a potential novel target for AD therapy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Proteínas Nogo/metabolismo , Receptor Nogo 1/metabolismo , Transducción de Señal , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Neuronas/metabolismo , Neuronas/patología , Proteínas Nogo/genética , Receptor Nogo 1/genética , Ratas , Ratas Sprague-Dawley , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
7.
Curr Biol ; 30(15): 2962-2973.e5, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32589913

RESUMEN

Disrupting binocular vision during a developmental critical period can yield enduring changes to ocular dominance (OD) in primary visual cortex (V1). Here we investigated how this experience-dependent plasticity is coordinated within the laminar circuitry of V1 by deleting separately in each cortical layer (L) a gene required to close the critical period, nogo-66 receptor (ngr1). Deleting ngr1 in excitatory neurons in L4, but not in L2/3, L5, or L6, prevented closure of the critical period, and adult mice remained sensitive to brief monocular deprivation. Intracortical disinhibition, but not thalamocortical disinhibition, accompanied this OD plasticity. Both juvenile wild-type mice and adult mice lacking ngr1 in L4 displayed OD plasticity that advanced more rapidly L4 than L2/3 or L5. Interestingly, blocking OD plasticity in L2/3 with the drug AM-251 did not impair OD plasticity in L5. We propose that L4 restricts disinhibition and gates OD plasticity independent of a canonical cortical microcircuit.


Asunto(s)
Plasticidad Neuronal/fisiología , Receptor Nogo 1/genética , Receptor Nogo 1/fisiología , Células Receptoras Sensoriales/fisiología , Corteza Visual/fisiología , Animales , Predominio Ocular , Eliminación de Gen , Ratones , Visión Binocular/fisiología
8.
Brain ; 143(6): 1697-1713, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32375169

RESUMEN

After CNS trauma such as spinal cord injury, the ability of surviving neural elements to sprout axons, reorganize neural networks and support recovery of function is severely restricted, contributing to chronic neurological deficits. Among limitations on neural recovery are myelin-associated inhibitors functioning as ligands for neuronal Nogo receptor 1 (NgR1). A soluble decoy (NgR1-Fc, AXER-204) blocks these ligands and provides a means to promote recovery of function in multiple preclinical rodent models of spinal cord injury. However, the safety and efficacy of this reagent in non-human primate spinal cord injury and its toxicological profile have not been described. Here, we provide evidence that chronic intrathecal and intravenous administration of NgR1-Fc to cynomolgus monkey and to rat are without evident toxicity at doses of 20 mg and greater every other day (≥2.0 mg/kg/day), and far greater than the projected human dose. Adult female African green monkeys underwent right C5/6 lateral hemisection with evidence of persistent disuse of the right forelimb during feeding and right hindlimb during locomotion. At 1 month post-injury, the animals were randomized to treatment with vehicle (n = 6) or 0.10-0.17 mg/kg/day of NgR1-Fc (n = 8) delivered via intrathecal lumbar catheter and osmotic minipump for 4 months. One animal was removed from the study because of surgical complications of the catheter, but no treatment-related adverse events were noted in either group. Animal behaviour was evaluated at 6-7 months post-injury, i.e. 1-2 months after treatment cessation. The use of the impaired forelimb during spontaneous feeding and the impaired hindlimb during locomotion were both significantly greater in the treatment group. Tissue collected at 7-12 months post-injury showed no significant differences in lesion size, fibrotic scar, gliosis or neuroinflammation between groups. Serotoninergic raphespinal fibres below the lesion showed no deficit, with equal density on the lesioned and intact side below the level of the injury in both groups. Corticospinal axons traced from biotin-dextran-amine injections in the left motor cortex were equally labelled across groups and reduced caudal to the injury. The NgR1-Fc group tissue exhibited a significant 2-3-fold increased corticospinal axon density in the cervical cord below the level of the injury relative to the vehicle group. The data show that NgR1-Fc does not have preclinical toxicological issues in healthy animals or safety concerns in spinal cord injury animals. Thus, it presents as a potential therapeutic for spinal cord injury with evidence for behavioural improvement and growth of injured pathways in non-human primate spinal cord injury.


Asunto(s)
Receptor Nogo 1/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Axones/patología , Médula Cervical/patología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Masculino , Actividad Motora/fisiología , Proteínas de la Mielina/metabolismo , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Neuronas/metabolismo , Neuronas/patología , Receptor Nogo 1/genética , Tractos Piramidales/patología , Ratas , Receptores Fc/genética , Receptores Fc/metabolismo , Recuperación de la Función , Médula Espinal/metabolismo , Médula Espinal/patología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
9.
Protein Cell ; 11(6): 417-432, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32350740

RESUMEN

Vision formation is classically based on projections from retinal ganglion cells (RGC) to the lateral geniculate nucleus (LGN) and the primary visual cortex (V1). Neurons in the mouse V1 are tuned to light stimuli. Although the cellular information of the retina and the LGN has been widely studied, the transcriptome profiles of single light-stimulated neuron in V1 remain unknown. In our study, in vivo calcium imaging and whole-cell electrophysiological patch-clamp recording were utilized to identify 53 individual cells from layer 2/3 of V1 as light-sensitive (LS) or non-light-sensitive (NS) by single-cell light-evoked calcium evaluation and action potential spiking. The contents of each cell after functional tests were aspirated in vivo through a patch-clamp pipette for mRNA sequencing. Moreover, the three-dimensional (3-D) morphological characterizations of the neurons were reconstructed in a live mouse after the whole-cell recordings. Our sequencing results indicated that V1 neurons with a high expression of genes related to transmission regulation, such as Rtn4r and Rgs7, and genes involved in membrane transport, such as Na+/K+ ATPase and NMDA-type glutamatergic receptors, preferentially responded to light stimulation. Furthermore, an antagonist that blocks Rtn4r signals could inactivate the neuronal responses to light stimulation in live mice. In conclusion, our findings of the vivo-seq analysis indicate the key role of the strength of synaptic transmission possesses neurons in V1 of light sensory.


Asunto(s)
Luz , Neuronas/metabolismo , Neuronas/efectos de la radiación , Técnicas de Placa-Clamp , RNA-Seq , Análisis de la Célula Individual , Animales , Calcio/metabolismo , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Corteza Visual/citología , Corteza Visual/metabolismo , Corteza Visual/efectos de la radiación
10.
J Neuroimmunol ; 341: 577172, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32028123

RESUMEN

Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system. The animal model of MS, experimental autoimmune encephalomyelitis (EAE), is commonly used for studies of human inflammatory demyelinating diseases and has been shown to be suitable for studying the effects of exercise on MS pathophysiology. The present study was conducted to determine the impact of forced swimming and voluntary running wheel exercises before and after the induction of EAE on expression of Nogo-A, NgR, and ROCK genes in the brain tissue. A total of 96 C57BL/6 mice were randomly divided into two groups, namely exercises before (EXb, n = 48) and after (EXa, n = 48) induction of EAE. Each group was divided into four subgroups: Forced Swimming + EAE (n = 12), Voluntary Running Wheel + EAE (n = 12), NoEX-EAE (n = 12), and Control group (n = 12). Animals performed either swimming exercise for 30 min per day or running wheel for one hour per day, five days per week for four weeks. Results of Luxal Fast Blue (LFB) staining demonstrated that the degree of demyelination was significantly less in the experimental exercised compared to NoEX-EAE groups (P < .05). Amazingly, both modes of exercise reduced the severity of MS symptoms in mice exposed to swimming and wheel running, evaluated as body weight, clinical scores, degree of demyelination, and gene expressions, regardless of whether the exercise was performed before or after EAE induction.


Asunto(s)
Encéfalo/metabolismo , Encefalomielitis Autoinmune Experimental/terapia , Terapia por Ejercicio , Esclerosis Múltiple/terapia , Proteínas del Tejido Nervioso/biosíntesis , Condicionamiento Físico Animal , Animales , Peso Corporal , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nogo/biosíntesis , Proteínas Nogo/genética , Receptor Nogo 1/biosíntesis , Receptor Nogo 1/genética , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Carrera , Natación , Volición , Quinasas Asociadas a rho/biosíntesis , Quinasas Asociadas a rho/genética
11.
Eur Rev Med Pharmacol Sci ; 24(1): 295-303, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31957843

RESUMEN

OBJECTIVE: The aim of this study was to investigate the effect and the mechanism of the NogoA/NgR1/RhoA signaling pathway on the apoptosis of neurons in cerebral infarction (CI) rats. Our findings might provide references for clinical prevention and treatment of CI. MATERIALS AND METHODS: A total of 60 adult male Wistar rats were randomly divided into 3 groups, including: Sham operation group (Sham group), CI group, and CI + NogoA gene knockout group (CI + NogoA KO group) using a random number table. The model of CI was successfully constructed using suture method in rats of CI group and CI + NogoA KO group. Only blood vessels were exposed in Sham group. At 2 days after CI operation, the rats were killed, and brain tissues were collected. Reverse Transcription-Polymerase Chain Reaction (RT-PCR) and Western blotting were used to detect the messenger ribonucleic acid (mRNA) and protein expression levels of NogoA/NgR1/RhoA in brain lesion tissues of rats in the three groups, respectively. Subsequently, the pathological damage of brain tissues was detected via hematoxylin and eosin (H&E) staining. TTC staining was carried out to evaluate the infarction area in each group. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining was conducted to measure the apoptosis level of neurons in brain tissues of rats in each group. Additionally, the level of Nissl's body in brain tissues of each group was examined by Nissl staining. Furthermore, the expression level of the platelet-derived growth factor (PDGF) in brain tissues of rats in the three groups was measured via immunohistochemistry. RESULTS: The mRNA and protein expression levels of the NogoA/NgR1/RhoA signaling pathway in brain tissues of rats in CI group increased significantly (p<0.05). NogoA KO could significantly reduce the infarction area of brain tissues in rats (p<0.05). H&E staining and Nissl's body staining revealed that neurons in the brain tissues of rats showed evident edema and disordered arrangement after CI. Meanwhile, the number of Nissl's body was remarkably reduced. However, after KO of NogoA, brain tissue damage was significantly alleviated in rats, and the number of Nissl's body increased remarkably at the same time (p<0.05). According to TUNEL staining results, inhibiting NogoA could notably reverse CI-induced apoptosis of neurons in brain tissues of rats (p<0.05). Immunohistochemical staining results demonstrated that the expression of PDGF in brain tissues of rats in CI group decreased markedly, whereas was significantly elevated in rats of CI + NogoA KO group (p<0.05). CONCLUSIONS: The expression of the NogoA/NgR1/RhoA signaling pathway was significantly elevated in brain tissues of CI rats. Furthermore, suppressing the NogoA/NgR1/RhoA signaling pathway could reduce CI-induced apoptosis of neurons in rats.


Asunto(s)
Corteza Cerebral/metabolismo , Infarto Cerebral/metabolismo , Neuronas/metabolismo , Receptor Nogo 1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Apoptosis , Corteza Cerebral/patología , Infarto Cerebral/patología , Modelos Animales de Enfermedad , Masculino , Neuronas/patología , Receptor Nogo 1/deficiencia , Receptor Nogo 1/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/genética , Proteínas de Unión al GTP rho/genética
12.
Exp Mol Med ; 51(10): 1-15, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31649250

RESUMEN

Nogo receptor (NgR) has been shown to inhibit the migration and invasion of human glioma cells. However, little is known regarding the regulatory mechanisms of NgR in glioblastoma multiforme (GBM). In this study, we propose a novel mechanism that regulates the maturation process of NgR through an interaction with vimentin. The inhibition of TGFß1 activity by LY2109761 attenuated the migration/invasion of GBM cells by upregulating cell-surface NgR. Conversely, the treatment of GBM cells with TGFß1 suppressed NgR maturation. We showed that NgR and vimentin interact, which could be a possible mechanism for the suppression of NgR maturation. The knockdown of vimentin suppressed the migration/invasion of GBM cells through the increased maturation of NgR. Finally, TCGA (The Cancer Genome Atlas) analysis also supported the association of NgR and vimentin. The maturation of NgR is regulated by the interaction of vimentin and NgR, which attenuates the invasive activity of GBM, and might be a potential therapeutic target for brain cancer.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Glioblastoma/genética , Receptor Nogo 1/genética , Factor de Crecimiento Transformador beta/genética , Vimentina/genética , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/patología , Humanos , Invasividad Neoplásica/genética
13.
J Neurosci ; 39(28): 5562-5580, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31061088

RESUMEN

We previously identified that ngr1 allele deletion limits the severity of experimental autoimmune encephalomyelitis (EAE) by preserving axonal integrity. However, whether this favorable outcome observed in EAE is a consequence of an abrogated neuronal-specific pathophysiological mechanism, is yet to be defined. Here we show that, Cre-loxP-mediated neuron-specific deletion of ngr1 preserved axonal integrity, whereas its re-expression in ngr1-/- female mice potentiated EAE-axonopathy. As a corollary, myelin integrity was preserved under Cre deletion in ngr1flx/flx , retinal ganglion cell axons whereas, significant demyelination occurred in the ngr1-/- optic nerves following the re-introduction of NgR1. Moreover, Cre-loxP-mediated axon-specific deletion of ngr1 in ngr1flx/flx mice also demonstrated efficient anterograde transport of fluorescently-labeled ChTxß in the optic nerves of EAE-induced mice. However, the anterograde transport of ChTxß displayed accumulation in optic nerve degenerative axons of EAE-induced ngr1-/- mice, when NgR1 was reintroduced but was shown to be transported efficiently in the contralateral non- recombinant adeno-associated virus serotype 2-transduced optic nerves of these mutant mice. We further identified that the interaction between the axonal motor protein, Kinesin-1 and collapsin response mediator protein 2 (CRMP2) was unchanged upon Cre deletion of ngr1 Whereas, this Kinesin-1/CRMP2 association was reduced when NgR1 was re-expressed in the ngr1-/- optic nerves. Our data suggest that NgR1 governs axonal degeneration in the context of inflammatory-mediated demyelination through the phosphorylation of CRMP2 by stalling axonal vesicular transport. Moreover, axon-specific deletion of ngr1 preserves axonal transport mechanisms, blunting the induction of inflammatory demyelination and limiting the severity of EAE.SIGNIFICANCE STATEMENT Multiple sclerosis (MS) is commonly induced by aberrant immune-mediated destruction of the protective sheath of nerve fibers (known as myelin). However, it has been shown that MS lesions do not only consist of this disease pattern, exhibiting heterogeneity with continual destruction of axons. Here we investigate how neuronal NgR1 can drive inflammatory-mediated axonal degeneration and demyelination within the optic nerve by analyzing its downstream signaling events that govern axonal vesicular transport. We identify that abrogating the NgR1/pCRMP2 signaling cascade can maintain Kinesin-1-dependent anterograde axonal transport to limit inflammatory-mediated axonopathy and demyelination. The ability to differentiate between primary and secondary mechanisms of axonal degeneration may uncover therapeutic strategies to limit axonal damage and progressive MS.


Asunto(s)
Transporte Axonal , Encefalomielitis Autoinmune Experimental/metabolismo , Vaina de Mielina/metabolismo , Receptor Nogo 1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Axones/metabolismo , Células Cultivadas , Encefalomielitis Autoinmune Experimental/genética , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cinesinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Receptor Nogo 1/genética , Células Ganglionares de la Retina/metabolismo , Transducción de Señal
14.
J Biol Chem ; 294(15): 6042-6053, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30787106

RESUMEN

Oligomeric assemblies of amyloid-ß (Aß) peptide (Aßo) in the brains of individuals with Alzheimer's disease (AD) are toxic to neuronal synapses. More than a dozen Aß receptor candidates have been suggested to be responsible for various aspects of the molecular pathology and memory impairment in mouse models of AD. A lack of consistent experimental design among previous studies of different receptor candidates limits evaluation of the relative roles of these candidates, producing some controversy within the field. Here, using cell-based assays with several Aß species, including Aßo from AD brains obtained by autopsy, we directly compared the Aß-binding capacity of multiple receptor candidates while accounting for variation in expression and confirming cell surface expression. In a survey of 15 reported Aß receptors, only cellular prion protein (PrPC), Nogo receptor 1 (NgR1), and leukocyte immunoglobulin-like receptor subfamily B member 2 (LilrB2) exhibited direct binding to synaptotoxic assemblies of synthetic Aß. Both PrPC and NgR1 preferentially bound synaptotoxic oligomers rather than nontoxic monomers, and the method of oligomer preparation did not significantly alter our binding results. Hippocampal neurons lacking both NgR1 and LilrB2 exhibited a partial reduction of Aßo binding, but this reduction was lower than in neurons lacking PrPC under the same conditions. Finally, binding studies with soluble Aßo from human AD brains revealed a strong affinity for PrPC, weak affinity for NgR1, and no detectable affinity for LilrB2. These findings clarify the relative contributions of previously reported Aß receptors under controlled conditions and highlight the prominence of PrPC as an Aß-binding site.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptor Nogo 1/metabolismo , Proteínas PrPC/metabolismo , Receptores Inmunológicos/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Receptor Nogo 1/genética , Proteínas PrPC/genética , Receptores Inmunológicos/genética
15.
J Neurotrauma ; 36(7): 1054-1059, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30226403

RESUMEN

Mild traumatic brain injury (mTBI) constitutes 75 ∼ 90% of all TBI cases and causes various physical, cognitive, emotional, and other psychological symptoms. Nogo receptor 1 (NgR1) is a regulator of structural brain plasticity during development and in adulthood. Here, we used mice that, in the absence of doxycycline, overexpress NgR1 in forebrain neurons (MemoFlex) to determine the role of NgR1 in recovery from mTBI with respect to balance, cognition, memory, and emotion. We compared wild-type (WT), MemoFlex, and MemoFlex + doxycycline mice to the same three groups subjected to mTBI. mTBI was induced by a controlled 30-g weight drop. We found that inability to downregulate NgR1 significantly impairs recovery from mTBI-induced impairments. When the NgR1 transgene was turned off, recovery was similar to that of WT mice. The results suggest that the ability to regulate NgR1 signaling is needed for optimal recovery of motor coordination and balance, spatial memory, cognition, and emotional functions after mTBI.


Asunto(s)
Conmoción Encefálica/metabolismo , Cognición/fisiología , Emociones/fisiología , Receptor Nogo 1/metabolismo , Equilibrio Postural/fisiología , Recuperación de la Función/fisiología , Animales , Conmoción Encefálica/fisiopatología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Receptor Nogo 1/genética , Prosencéfalo/metabolismo , Memoria Espacial/fisiología
16.
Exp Mol Pathol ; 105(3): 357-363, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30312597

RESUMEN

The present study is aimed to investigate the molecular mechanism of miR-10 targeting NgR to regulate the proliferation and inflammatory cytokine secretion in microglia cells (MCs). The expression levels of NgR, IL-1ß, and TNF-α in rat MCs were detected by QPCR and Immunoblotting assays. The proliferation of MC was detected by MTT and Soft agar clone formation assays. The target gene of miR-10 was verified by dual luciferase reporter gene assay. Overexpressed miR-10 downregulated the expression of NgR, promoted MC proliferation and inhibited inflammatory cytokine secretion. On the other hand, the inhibition of miR-10 upregulated the expression of NgR, inhibited MC proliferation and promoted inflammatory cytokine secretion. It was found that miR-10 targeted to regulate NgR. Overexpression of NgR restored the regulation effects of miR-10 on MC proliferation and inflammatory cytokine secretion. In summary, miR-10 can promote MC proliferation and inhibit the inflammatory cytokine secretion via targeting the NgR gene to down-regulate its expression.


Asunto(s)
Citocinas/metabolismo , Regulación de la Expresión Génica/genética , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Receptor Nogo 1/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Proliferación Celular/genética , Células Cultivadas , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Glicoproteínas de Membrana/genética , Receptor Nogo 1/genética , Ratas , Receptores Inmunológicos/genética
17.
Curr Biol ; 28(12): 1914-1923.e5, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29887305

RESUMEN

Degrading vision by one eye during a developmental critical period yields enduring deficits in both eye dominance and visual acuity. A predominant model is that "reactivating" ocular dominance (OD) plasticity after the critical period is required to improve acuity in amblyopic adults. However, here we demonstrate that plasticity of eye dominance and acuity are independent and restricted by the nogo-66 receptor (ngr1) in distinct neuronal populations. Ngr1 mutant mice display greater excitatory synaptic input onto both inhibitory and excitatory neurons with restoration of normal vision. Deleting ngr1 in excitatory cortical neurons permits recovery of eye dominance but not acuity. Reciprocally, deleting ngr1 in thalamus is insufficient to rectify eye dominance but yields improvement of acuity to normal. Abolishing ngr1 expression in adult mice also promotes recovery of acuity. Together, these findings challenge the notion that mechanisms for OD plasticity contribute to the alterations in circuitry that restore acuity in amblyopia.


Asunto(s)
Ambliopía/fisiopatología , Predominio Ocular/fisiología , Neuronas/metabolismo , Agudeza Visual/fisiología , Ambliopía/genética , Animales , Predominio Ocular/genética , Femenino , Masculino , Ratones , Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Agudeza Visual/genética
18.
PLoS One ; 13(5): e0196565, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29768445

RESUMEN

A variety of conditions ranging from glaucoma to blunt force trauma lead to optic nerve atrophy. Identifying signaling pathways for stimulating axon growth in the optic nerve may lead to treatments for these pathologies. Inhibiting signaling by the nogo-66 receptor 1 (NgR1) promotes the re-extension of axons following a crush injury to the optic nerve, and while NgR1 mRNA and protein expression are observed in the retinal ganglion cell (RGC) layer and inner nuclear layer, which retinal cell types express NgR1 remains unknown. Here we determine the expression pattern of NgR1 in the mouse retina by co-labeling neurons with characterized markers of specific retinal neurons together with antibodies specific for NgR1 or Green Fluorescent Protein expressed under control of the ngr1 promoter. We demonstrate that more than 99% of RGCs express NgR1. Thus, inhibiting NgR1 function may ubiquitously promote the regeneration of axons by RGCs. These results provide additional support for the therapeutic potential of NgR1 signaling in reversing optic nerve atrophy.


Asunto(s)
Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Axones/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Receptor Nogo 1/deficiencia , Nervio Óptico/metabolismo , Nervio Óptico/fisiología , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Ganglionares de la Retina/patología , Transducción de Señal
19.
Am J Rhinol Allergy ; 32(4): 208-216, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29754498

RESUMEN

Background Dysfunctional innervation might contribute to the pathogenesis of chronic rhinosinusitis with nasal polyps (CRSwNP), but the state of the axonal outgrowth signaling in CRSwNP is unknown. The purpose of this study was to explore the axonal outgrowth pathway-related protein expression in CRSwNP. Methods Institutional review board approved study in which tissue proteomes were compared between control and CRSwNP patients (n = 10/group) using an aptamer-based proteomic array and confirmed by whole transcriptomic analysis. Results Compared with controls, proteins associated with axonal guidance signaling pathway such as beta-nerve growth factor, semaphorin 3A, Ras-related C3 botulinum toxin substrate 1, Bcl-2, protein kinase C delta type, and Fyn were significantly decreased in patients with CRSwNP (fold change [FC] = -1.17, P = .002; FC = -1.09, P < .001; FC = -1.33, P < .001; FC = -1.31, P < .001; FC = -1.31, P = .004; and FC = -1.20, P = 0.012, respectively). In contrast, reticulon-4 receptor, an inhibitory factor, was significantly increased in patients with CRSwNP (FC = 1.25, P < .001). Furthermore, neuronal growth-associated proteins such as ciliary neurotrophic factor receptor subunit alpha, neuronal growth regulator 1, neuronal cell adhesion molecule, neural cell adhesion molecule L1, platelet-derived growth factor subunit A, and netrin-4 were all significantly decreased in patients with CRSwNP (FC = -1.25, P < .001; FC = -1.27, P = .002; FC = -1.65, P = .013; FC = -4.20, P < .001; FC = -1.28, P < .001; and FC = -2.31, P < .001, respectively). In contrast, tissue eosinophil count ( P < .001) and allergic inflammation factors such as IgE, periostin, and galectin-10 were all significantly increased in patients with CRSwNP (FC = 12.28, P < .001; FC = 3.95, P < .001; and FC = 2.44, P < .001, respectively). Furthermore, the log FC of the studied proteins expression significantly and positively correlated with log FC of their mRNA expression ( P < .001, r = .88). Conclusions Axonal guidance signaling and neural growth factors pathways proteins are significantly suppressed in eosinophilic CRSwNP.


Asunto(s)
Orientación del Axón/genética , Eosinófilos/fisiología , Pólipos Nasales/fisiopatología , Nariz/fisiología , Rinitis/fisiopatología , Sinusitis/fisiopatología , Adulto , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica , Humanos , Inmunoglobulina E/sangre , Inflamación/genética , Masculino , Persona de Mediana Edad , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Proteoma , Transducción de Señal , Adulto Joven
20.
Sci Signal ; 11(524)2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29615517

RESUMEN

Axonal growth after traumatic spinal cord injury is limited by endogenous inhibitors, selective blockade of which promotes partial neurological recovery. The partial repair phenotypes suggest that compensatory pathways limit improvement. Gene expression profiles of mice deficient in Ngr1, which encodes a receptor for myelin-associated inhibitors of axonal regeneration such as Nogo, revealed that trauma increased the mRNA expression of ORL1, which encodes the receptor for the opioid-related peptide nociceptin. Endogenous and overexpressed ORL1 coimmunoprecipitated with immature NgR1 protein, and ORL1 enhanced the O-linked glycosylation and surface expression of NgR1 in HEK293T and Neuro2A cells and primary neurons. ORL1 overexpression inhibited cortical neuron axon regeneration independently of NgR1. Furthermore, regeneration was inhibited by an ORL1 agonist and enhanced by the ORL1 antagonist J113397 through a ROCK-dependent mechanism. Mice treated with J113397 after dorsal hemisection of the mid-thoracic spinal cord recovered greater locomotor function and exhibited lumbar raphespinal axon sprouting. These effects were further enhanced by combined Ngr1 deletion and ORL1 inhibition. Thus, ORL1 limits neural repair directly and indirectly by enhancing NgR1 maturation, and ORL1 antagonists enhance recovery from traumatic CNS injuries in wild-type and Ngr1 null mice.


Asunto(s)
Axones/fisiología , Regeneración Nerviosa/fisiología , Receptor Nogo 1/metabolismo , Receptores Opioides/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Axones/metabolismo , Células COS , Línea Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/genética , Neuronas/citología , Neuronas/metabolismo , Neuronas/fisiología , Receptor Nogo 1/genética , Péptidos Opioides/farmacología , Receptores Opioides/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/fisiopatología , Receptor de Nociceptina , Nociceptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA