Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Cell ; 184(24): 5869-5885.e25, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34758294

RESUMEN

RTN4-binding proteins were widely studied as "NoGo" receptors, but their physiological interactors and roles remain elusive. Similarly, BAI adhesion-GPCRs were associated with numerous activities, but their ligands and functions remain unclear. Using unbiased approaches, we observed an unexpected convergence: RTN4 receptors are high-affinity ligands for BAI adhesion-GPCRs. A single thrombospondin type 1-repeat (TSR) domain of BAIs binds to the leucine-rich repeat domain of all three RTN4-receptor isoforms with nanomolar affinity. In the 1.65 Å crystal structure of the BAI1/RTN4-receptor complex, C-mannosylation of tryptophan and O-fucosylation of threonine in the BAI TSR-domains creates a RTN4-receptor/BAI interface shaped by unusual glycoconjugates that enables high-affinity interactions. In human neurons, RTN4 receptors regulate dendritic arborization, axonal elongation, and synapse formation by differential binding to glial versus neuronal BAIs, thereby controlling neural network activity. Thus, BAI binding to RTN4/NoGo receptors represents a receptor-ligand axis that, enabled by rare post-translational modifications, controls development of synaptic circuits.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Encéfalo/metabolismo , Neurogénesis , Neuronas/metabolismo , Proteínas Nogo/metabolismo , Receptores Nogo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adipoquinas/metabolismo , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular Neuronal/metabolismo , Complemento C1q/metabolismo , Dendritas/metabolismo , Glicosilación , Células HEK293 , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ligandos , Ratones Endogámicos C57BL , Red Nerviosa/metabolismo , Polisacáridos/metabolismo , Unión Proteica , Dominios Proteicos , Eliminación de Secuencia , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
2.
Development ; 151(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38300806

RESUMEN

Defective tissue fusion during mammalian embryogenesis results in congenital anomalies, such as exencephaly, spina bifida and cleft lip and/or palate. The highly conserved transcription factor grainyhead-like 2 (Grhl2) is a crucial regulator of tissue fusion, with mouse models lacking GRHL2 function presenting with a fully penetrant open cranial neural tube, facial and abdominal clefting (abdominoschisis), and an open posterior neuropore. Here, we show that GRHL2 interacts with the soluble morphogen protein and bone morphogenetic protein (BMP) inhibitor noggin (NOG) to impact tissue fusion during development. The maxillary prominence epithelium in embryos lacking Grhl2 shows substantial morphological abnormalities and significant upregulation of NOG expression, together with aberrantly distributed pSMAD5-positive cells within the neural crest cell-derived maxillary prominence mesenchyme, indicative of disrupted BMP signalling. Reducing this elevated NOG expression (by generating Grhl2-/-;Nog+/- embryos) results in delayed embryonic lethality, partial tissue fusion rescue, and restoration of tissue form within the craniofacial epithelia. These data suggest that aberrant epithelial maintenance, partially regulated by noggin-mediated regulation of BMP-SMAD pathways, may underpin tissue fusion defects in Grhl2-/- mice.


Asunto(s)
Labio Leporino , Fisura del Paladar , Defectos del Tubo Neural , Animales , Ratones , Proteínas Morfogenéticas Óseas/metabolismo , Mamíferos/metabolismo , Tubo Neural/metabolismo , Receptores Nogo/metabolismo
3.
Curr Opin Neurol ; 36(6): 516-522, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37865850

RESUMEN

PURPOSE OF REVIEW: After traumatic spinal cord injury (SCI), neurological deficits persist due to the disconnection of surviving neurons. While repair of connectivity may restore function, no medical therapy exists today.This review traces the development of the neural repair-based therapeutic AXER-204 from animal studies to the recent clinical trial for chronic cervical SCI. RECENT FINDINGS: Molecular studies reveal a Nogo-66 Receptor 1 (NgR1, RTN4R) pathway inhibiting axon regeneration, sprouting, and plasticity in the adult mammalian central nervous system (CNS). Rodent and nonhuman primate studies demonstrate that the soluble receptor decoy NgR(310)ecto-Fc or AXER-204 promotes neural repair and functional recovery in transection and contusion SCI. Recently, this biological agent completed a first-in-human and randomized clinical trial for chronic cervical SCI. The intervention was safe and well tolerated. Across all participants, upper extremity strength did not improve with treatment. However, posthoc and biomarker analyses suggest that AXER-204 may benefit treatment-naïve patients with incomplete SCI in the chronic stage. SUMMARY: NgR1 signaling restricts neurological recovery in animal studies of CNS injury. The recent clinical trial of AXER-204 provides encouraging signals supporting future focused trials of this neural repair therapeutic. Further, AXER-204 studies provide a roadmap for the development of additional and synergistic therapies for chronic SCI.


Asunto(s)
Axones , Traumatismos de la Médula Espinal , Animales , Humanos , Axones/metabolismo , Receptores Nogo/metabolismo , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Proteínas de la Mielina/uso terapéutico , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/terapia , Receptor Nogo 1/metabolismo , Recuperación de la Función , Médula Espinal , Mamíferos/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Proc Natl Acad Sci U S A ; 117(21): 11744-11752, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32404418

RESUMEN

Auditory experience drives neural circuit refinement during windows of heightened brain plasticity, but little is known about the genetic regulation of this developmental process. The primary auditory cortex (A1) of mice exhibits a critical period for thalamocortical connectivity between postnatal days P12 and P15, during which tone exposure alters the tonotopic topography of A1. We hypothesized that a coordinated, multicellular transcriptional program governs this window for patterning of the auditory cortex. To generate a robust multicellular map of gene expression, we performed droplet-based, single-nucleus RNA sequencing (snRNA-seq) of A1 across three developmental time points (P10, P15, and P20) spanning the tonotopic critical period. We also tone-reared mice (7 kHz pips) during the 3-d critical period and collected A1 at P15 and P20. We identified and profiled both neuronal (glutamatergic and GABAergic) and nonneuronal (oligodendrocytes, microglia, astrocytes, and endothelial) cell types. By comparing normal- and tone-reared mice, we found hundreds of genes across cell types showing altered expression as a result of sensory manipulation during the critical period. Functional voltage-sensitive dye imaging confirmed GABA circuit function determines critical period onset, while Nogo receptor signaling is required for its closure. We further uncovered previously unknown effects of developmental tone exposure on trajectories of gene expression in interneurons, as well as candidate genes that might execute tonotopic plasticity. Our single-nucleus transcriptomic resource of developing auditory cortex is thus a powerful discovery platform with which to identify mediators of tonotopic plasticity.


Asunto(s)
Corteza Auditiva , Núcleo Celular/metabolismo , ARN , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Animales , Corteza Auditiva/crecimiento & desarrollo , Corteza Auditiva/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Ratones , Receptores Nogo/genética , Receptores Nogo/metabolismo , ARN/análisis , ARN/genética , ARN/metabolismo , Análisis de Secuencia de ARN/métodos
5.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638567

RESUMEN

Dopaminergic treatment in combination with rehabilitative training enhances long-term recovery after stroke. However, the underlying mechanisms on structural plasticity are unknown. Here, we show an increased dopaminergic innervation of the ischemic territory during the first week after stroke induced in Wistar rats subjected to transient occlusion of the middle cerebral artery (tMCAO) for 120 min. This response was also found in rats subjected to permanent focal ischemia induced by photothrombosis (PT) and mice subjected to PT or tMCAO. Dopaminergic branches were detected in the infarct core of mice and rats in both stroke models. In addition, the Nogo A pathway was significantly downregulated in rats treated with levodopa (LD) compared to vehicle-treated animals subjected to tMCAO. Specifically, the number of Nogo A positive oligodendrocytes as well as the levels of Nogo A and the Nogo A receptor were significantly downregulated in the peri-infarct area of LD-treated animals, while the number of Oligodendrocyte transcription factor 2 positive cells increased in this region after treatment. In addition, we observed lower protein levels of Growth Associated Protein 43 in the peri-infarct area compared to sham-operated animals without treatment effect. The results provide the first evidence of the plasticity-promoting actions of dopaminergic treatment following stroke.


Asunto(s)
Dopaminérgicos/farmacología , Dopaminérgicos/uso terapéutico , Levodopa/farmacología , Levodopa/uso terapéutico , Plasticidad Neuronal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Regulación hacia Abajo/efectos de los fármacos , Proteína GAP-43/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Ratones , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Receptores Nogo/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Ratas Wistar , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Trombosis/complicaciones
6.
Neuropediatrics ; 50(6): 387-390, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31370080

RESUMEN

BACKGROUND: Leukoencephalopathy associated with dysmorphic features may be attributed to chromosomal abnormalities such as 17p13.3 microdeletion syndrome. CASE: A 19-year-old female patient was referred to our hospital for diagnostic evaluation of her leukoencephalopathy. She demonstrated moderate intellectual disability, minor dysmorphic features, and short stature. Serial brain magnetic resonance images obtained within a 16-year interval revealed prolonged T2 signals in the deep cerebral white matter with enlarged Virchow-Robin spaces. A nonsymptomatic atlas anomaly was also noted. Using microarray-based comparative genomic hybridization, we identified a 2.2-Mb terminal deletion at 17p13.3, encompassing YWHAE, CRK, and RTN4RL1 but not PAFAH1B1. CONCLUSION: Except for atlas anomaly, the patient's clinical and imaging findings were compatible with the diagnosis of 17p13.3 microdeletion syndrome. The white matter abnormality was static and nonprogressive. The association between the atlas abnormality and this deletion remains elusive. We note the importance of exploring submicroscopic chromosomal imbalance when patients show prominent but static white matter abnormalities with discrepantly mild and stable neurological signs.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 17/genética , Leucoencefalopatías/genética , Proteínas 14-3-3/genética , Estatura , Atlas Cervical/anomalías , Atlas Cervical/diagnóstico por imagen , Femenino , Humanos , Discapacidad Intelectual/etiología , Discapacidad Intelectual/genética , Leucoencefalopatías/diagnóstico por imagen , Imagen por Resonancia Magnética , Receptores Nogo/genética , Proteínas Proto-Oncogénicas c-crk/genética , Sustancia Blanca/diagnóstico por imagen , Adulto Joven
7.
Zhonghua Yan Ke Za Zhi ; 55(1): 37-45, 2019 Jan 11.
Artículo en Zh | MEDLINE | ID: mdl-30641674

RESUMEN

Objective: To investigate the postnatal development of perineuronal net (PNN), Nogo receptor (Nogo R) in visual development and the effect of fluoxetine(Flx) on remodeling it in the visual cortex of adult rats. Methods: Experimental study. (1) Wistar rats were divided into postnatal weeks (PW)1,PW3,PW5,PW7,PW9 group (8 rats in each group) according to the age of PW. The changes of PNN and Nogo R were observed in the visual cortex of each group. (2) The adult rats (10 weeks after birth) were randomized into Flx 0W, Flx 2W, Flx 4W, Flx 6W and Flx 8W group (8 rats in each group) according to Flx administrational weeks. The influence of Flx on the expression of PNN and Nogo receptor in the visual cortex was detected by immunofluorescence and western blots. (3) The adult rats were randomized into Cont (negative control), Flx, binocular form deprivation(BFD,positive control) and BFD+Flx group (8 rats in each group). Flx group accepted oral administration at the dosage of 0.2 mg/ml once per day for 4 weeks. The eyelids were binocularly sutured for 2 weeks to form the BFD group, and the combination of Flx administration and BFD was performed in the BFD+Flx group.No intervention was conducted in the control group (Cont group). Immunofluorescence was used to observe the expression pattern of PNN staining by biotinylated wisteria floribunda lectin (WFA). The expression of Nogo R in the visual cortex was detected by immunofluorescence and Western blots. The expression of PNN and Nogo R were examined in each group. And t test, analysis of variance and rank sum test were employed for inter-group comparison based on the homogeneity of variance. Bonferroni method was used for multiple comparison and simple linear regression analysis was used for the trend. Results: (1) The expression of PNN (standardized b=0.97, P=0.005) and Nogo R (standardized b=0.96, P=0.010) increased during the postnatal development and the Nogo R reached the matured level at PW7 (PW7 vs. PW9, 131.83±3.78 vs. 135.11±3.92, Z=1.93, P=0.062). (2) Flx significantly decreased PNN in the visual cortex of adult rats. The density of PNN-positive cells in the visual cortex of healthy adult rats fed with Flx for 4 weeks was (86.22±7.68)/mm(2), which was similar to that of 3 weeks old rats [(84.21±6.68)/mm(2), t=2.08, P=0.073]. The expression of PNN (standardized b=-0.88, P=0.040) and it's receptor Nogo R (standardized b=-0.90, P=0.007) decreased with prolongation of Flx use. (3) The expression of Nogo R (t=13.42,11.47, 18.13; P=0.012, 0.013, 0.001; Flx, BFD and BFD+Flx group vs. Cont group) and the density of PNN (t=10.09, 7.64, 13.01; P=0.007, 0.011, 0.001; Flx, BFD and BFD+Flx group vs. Cont group) could be modulated by Flx and BFD after the critical period. There are no differences in BFD and Flx group on Nogo R changes (t=2.41, P=0.153). The expression of Nogo R protein was also different among the 4 groups (H=5.69, P=0.041). The effect of Flx combined with BFD was better than the Flx or BFD alone (Flx vs. BFD+Flx, Z=4.22, P=0.005; BFD vs. BFD+Flx, Z=3.09, P=0.010). Conclusions: The expression of PNN and Nogo R increase during the postnatal development. Chronic Flx treatment decrease the expression of PNN and Nogo R after the critical period in the visual cortex of adult rats, that is the same as BFD. (Chin J Ophthalmol, 2019, 55:37-45).


Asunto(s)
Fluoxetina , Inhibidores Selectivos de la Recaptación de Serotonina , Corteza Visual , Animales , Matriz Extracelular , Fluoxetina/farmacología , Receptores Nogo , Distribución Aleatoria , Ratas , Ratas Wistar , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Corteza Visual/efectos de los fármacos
8.
J Neuroinflammation ; 15(1): 210, 2018 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-30029608

RESUMEN

BACKGROUND: Alzheimer's disease is characterized by progressive accumulation of ß-amyloid (Aß)-containing amyloid plaques, and microglia play a critical role in internalization and degradation of Aß. Our previous research confirmed that Nogo-66 binding to Nogo receptors (NgR) expressed on microglia inhibits cell adhesion and migration in vitro. METHODS: The adhesion and migration of microglia isolated from WT and APP/PS1 mice from different ages were measured by adhesion assays and transwells. After NEP1-40 (a competitive antagonist of Nogo/NgR pathway) was intracerebroventricularly administered via mini-osmotic pumps for 2 months in APP/PS1 transgenic mice, microglial recruitment toward Aß deposits and CD36 expression were determined. RESULTS: In this paper, we found that aging led to a reduction of microglia adhesion and migration to fAß1-42 in WT and APP/PS1 mice. The adhesion and migration of microglia to fAß1-42 were downregulated by the Nogo, which was mediated by NgR, and the increased inhibitory effects of the Nogo could be observed in aged mice. Moreover, Rho GTPases contributed to the effects of the Nogo on adhesion and migration of microglia to fAß1-42 by regulating cytoskeleton arrangement. Furthermore, blocking the Nogo/NgR pathway enhanced recruitment of microglia toward Aß deposits and expression of CD36 in APP/PS1 mice. CONCLUSION: Taken together, Nogo/NgR pathway could take part in Aß pathology in AD by modulating microglial adhesion and migration to Aß and the Nogo/NgR pathway might be an important target for treating AD.


Asunto(s)
Envejecimiento , Péptidos beta-Amiloides/farmacología , Adhesión Celular/efectos de los fármacos , Microglía/efectos de los fármacos , Proteínas Nogo/metabolismo , Receptores Nogo/metabolismo , Fragmentos de Péptidos/farmacología , Envejecimiento/efectos de los fármacos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/patología , Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de la Mielina/farmacología , Presenilina-1/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
9.
BMC Complement Altern Med ; 17(1): 346, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28668079

RESUMEN

BACKGROUND: Axon growth inhibitory factors NogoA/Nogo receptor (NgR) and its signaling pathways RhoA/Rho kinase (ROCK) play a critical role in the repair of nerve damage in multiple sclerosis (MS). Bu Shen Yi Sui Capsule (BSYSC) is an effective Chinese formula utilized to treat MS in clinical setting and noted for its potent neuroprotective effects. In this study, we focus on the effects of BSYSC on promoting nerve repair and the underlying mechanisms in mice with experimental autoimmune encephalomyelitis (EAE), an animal model of MS. METHODS: The EAE mouse model was induced by injecting subcutaneously with myelin oligodendrocyte glycoprotein (MOG) 35-55 supplemented with pertussis toxin. BSYSC was orally administrated at dose of 3.0 g/kg once a day for 40 days. The levels of protein gene product (PGP) 9.5, p-Tau, growth associated protein (GAP) -43, KI67 and Nestin in the brain or spinal cord on 20 and 40 day post-induction (dpi) were detected via immunofluorescence and Western blot analysis. Furthermore, NogoA/NgR and RhoA/ROCK signaling molecules were studied by qRT-PCR and Western blot analysis. RESULTS: Twenty or 40 days of treatment with BSYSC increased markedly PGP9.5 and GAP-43 levels, reduced p-Tau in the brain or spinal cord of mice with EAE. In addition, BSYSC elevated significantly the expression of KI67 and Nestin in the spinal cord 40 dpi. Further study showed that the activation of NogoA/NgR and RhoA/ROCK were suppressed by the presence of BSYSC. CONCLUSIONS: BSYSC could attenuate axonal injury and promote repair of axonal damage in EAE mice in part through the down-regulation of NogoA/NgR and RhoA/ROCK signaling pathways.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteínas Nogo/metabolismo , Receptores Nogo/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Nogo/genética , Receptores Nogo/genética , Transducción de Señal , Quinasas Asociadas a rho/genética , Proteína de Unión al GTP rhoA/genética
10.
J Neuroinflammation ; 13(1): 265, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27724971

RESUMEN

BACKGROUND: Nogo-A and its putative receptor NgR are considered to be among the inhibitors of axonal regeneration in the CNS. However, few studies so far have addressed the issue of local NgR complex multilateral localization within inflammation in an MS mouse model of autoimmune demyelination. METHODS: Chronic experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice. Analyses were performed on acute (days 18-22) and chronic (day 50) time points and compared to controls. The temporal and spatial expression of the Nogo receptor complex (NgR and coreceptors) was studied at the spinal cord using epifluorescent and confocal microscopy or real-time PCR. Data are expressed as cells/mm2, as mean % ± SEM, or as arbitrary units of integrated density. RESULTS: Animals developed a moderate to severe EAE without mortality, followed by a progressive, chronic clinical course. NgR complex spatial expression varied during the main time points of EAE. NgR with coreceptors LINGO-1 and TROY was increased in the spinal cord in the acute phase whereas LINGO-1 and p75 signal seemed to be dominant in the chronic phase, respectively. NgR was detected on gray matter NeuN+ neurons of the spinal cord, within the white matter inflammatory foci (14.2 ± 4.3 % NgR+ inflammatory cells), and found to be colocalized with GAP-43+ axonal growth cones while no ß-TubIII+, SMI-32+, or APP+ axons were found as NgR+. Among the NgR+ inflammatory cells, 75.6 ± 9.0 % were microglial/macrophages (lectin+), 49.6 ± 14.2 % expressed CD68 (phagocytic ED1+ cells), and no cells were Mac-3+. Of these macrophages/monocytes, only Arginase-1+/NgR+ but not iNOS+/NgR+ were present in lesions both in acute and chronic phases. CONCLUSIONS: Our data describe in detail the expression of the Nogo receptor complex within the autoimmune inflammatory foci and suggest a possible immune action for NgR apart from the established inhibitory one on axonal growth. Its expression by inflammatory macrophages/monocytes could signify a possible role of these cells on axonal guidance and clearance of the lesioned area during inflammatory demyelination.


Asunto(s)
Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica/inmunología , Receptores Nogo/metabolismo , Transducción de Señal/fisiología , Animales , Antígenos de Diferenciación/metabolismo , Arginasa/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Adyuvante de Freund/inmunología , Adyuvante de Freund/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Receptores Nogo/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Estadísticas no Paramétricas
12.
Am J Med Genet A ; 167A(12): 3174-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26383923

RESUMEN

Chromosome region 10q22.3-q23.3 contains several low copy repeats (LCRs) and is prone to recombination. Deletions with breakpoints within LCR3 and LCR4 have been described to be associated with intellectual disability and dysmorphic features, while the reciprocal duplications are rarely reported. We present an additional case with multiple congenital anomalies that include microcephaly, cardiac defect, and mild intellectual disability, in which a de novo interstitial 8.2-Mb duplication of 10q22.3-q23.3, including BMPR1A and NGR3, was identified by Illumina SNP array platform. Our study is consistent with the hypothesis that the BMPR1A is a plausible candidate gene for congenital heart disease (CHD) and should contribute to the diagnosis and treatment of these genomic diseases.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Cromosomas Humanos Par 10/genética , Duplicación de Gen , Cardiopatías Congénitas/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Receptores de Superficie Celular/genética , Adulto , Niño , Aberraciones Cromosómicas , Femenino , Proteínas Ligadas a GPI/genética , Cardiopatías Congénitas/patología , Humanos , Discapacidad Intelectual/patología , Masculino , Microcefalia/patología , Receptores Nogo , Pronóstico , Adulto Joven
13.
J Psychiatr Res ; 170: 207-216, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38157668

RESUMEN

Some immune molecules including neurite outgrowth inhibitor (Nogo) ligands and their receptor(Nogo receptor-1: NgR1)are expressed at the neuronal synaptic sites. Paired immunoglobulin-like receptor B (PirB) is another Nogo receptor that also binds to major histocompatibility complex I and ß-amyloid and suppresses dendritic immune cell functions and neuronal plasticity in the central nervous system. Augmenting structural and functional neural plasticity by manipulating the Nogo signaling pathway is a novel promising strategy for treating brain ischemia and degenerative processes such as Alzheimer's disease. In recent decades psychiatric research using experimental animals has focused on the attenuation of neural plasticity by stress loadings and on the enhanced resilience by psychopharmacological treatments. In the present study, we examined possible expressional alterations in Nogo signal-related proteins in the rat hippocampus after behavioral stress loadings and antidepressant treatments. To validate the effectiveness of the procedures, previously reported increase in brain-derived neurotrophic factor (BDNF) by ECS or ketamine administration and decrease of BDNF by stress loadings are also shown in the present study. Significant increases in hippocampal NgR1 and PirB expression were observed following chronic variable stress, and a significant increase in NgR1 expression was observed under a single prolonged stress paradigm. These results indicate a possible contribution of enhanced Nogo signaling to the attenuation of neural plasticity in response to stressful experiences. Additionally, the suppression of hippocampal NgR1 expression using electroconvulsive seizure treatment and administration of subanesthetic dose of ketamine supported the increased neural plasticity induced by the antidepressant treatments.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Ketamina , Ratas , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Antidepresivos/farmacología , Receptores Nogo/metabolismo
14.
Cell Signal ; 114: 110978, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37972801

RESUMEN

Spinal cord injury (SCI) is a devastating disorder and a leading cause of disability in adults worldwide. Multiple studies have reported the upregulation of programmed cell death 1 (PD-1) following SCI. However, the underlying mechanism of PD-1 deficiency in SCI is not well established. Therefore, we aimed to investigate the role and potential mechanism of PD-1 in SCI pathogenesis. PD-1 Knockout (KO) SCI mouse model was established, and PD-1 expression was evaluated in tissue samples by western blot assay. We then used a series of function gain-and-loss assays to determine the role of PD-1 in SCI pathogenesis. Moreover, mechanistic assays were performed to explore the association between PD-1, neuron-glia antigen-2 (NG2) glia cells, and miR-23b-5p and then investigated the involved signaling pathway. Results illustrated that PD-1 deficiency enhanced the inflammatory response, neuron loss, and functional impairment induced by SCI. We found that NG2 glia depletion aggravated inflammation, reduced neural survival, and suppressed locomotor recovery in murine SCI model. Further analysis indicated that NG2+ cells were increased in the spinal cord of SCI mice, and PD-1 deficiency increased the number of NG2+ cells by activating the Nogo receptor/ras homolog family member A/Rho kinase (NgR/RhoA/ROCK) signaling. Mechanistically, miR-23b-5p was identified as the negative regulator of PD-1 in NG2 glia. MiR-23b-5p deficiency reduced the expression of inflammatory cytokines, enhanced neural survival, and promoted locomotor recovery in SCI mice, which was counteracted by PD-1 deficiency. In conclusion, PD-1 deficiency exacerbates SCI in vivo by regulating reprogramming of NG2 glia and activating the NgR/RhoA/ROCK signaling.


Asunto(s)
MicroARNs , Receptor de Muerte Celular Programada 1 , Traumatismos de la Médula Espinal , Animales , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Neuroglía/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Transducción de Señal , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Receptores Nogo/metabolismo , Quinasas Asociadas a rho/metabolismo
15.
Biochem Pharmacol ; 210: 115461, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36828272

RESUMEN

Neurodegenerative diseases (NDs) such as Alzheimer's, Parkinson's, Multiple Sclerosis, Hereditary Spastic Paraplegia, and Amyotrophic Lateral Sclerosis have emerged as the most dreaded diseases due to a lack of precise diagnostic tools and efficient therapies. Despite the fact that the contributing factors of NDs are still unidentified, mounting evidence indicates the possibility that genetic and cellular changes may lead to the significant production of abnormally misfolded proteins. These misfolded proteins lead to damaging effects thereby causing neurodegeneration. The association between Neurite outgrowth factor (Nogo) with neurological diseases and other peripheral diseases is coming into play. Three isoforms of Nogo have been identified Nogo-A, Nogo-B and Nogo-C. Among these, Nogo-A is mainly responsible for neurological diseases as it is localized in the CNS (Central Nervous System), whereas Nogo-B and Nogo-C are responsible for other diseases such as colitis, lung, intestinal injury, etc. Nogo-A, a membrane protein, had first been described as a CNS-specific inhibitor of axonal regeneration. Several recent studies have revealed the role of Nogo-A proteins and their receptors in modulating neurite outgrowth, branching, and precursor migration during nervous system development. It may also modulate or affect the inhibition of growth during the developmental processes of the CNS. Information about the effects of other ligands of Nogo protein on the CNS are yet to be discovered however several pieces of evidence have suggested that it may also influence the neuronal maturation of CNS and targeting Nogo-A could prove to be beneficial in several neurodegenerative diseases.


Asunto(s)
Proteínas de la Mielina , Enfermedades Neurodegenerativas , Humanos , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Proteínas Nogo , Regeneración Nerviosa/fisiología , Factores de Crecimiento Nervioso , Receptores Nogo
16.
Sci Rep ; 13(1): 22898, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38129508

RESUMEN

Recovery after spinal cord injury (SCI) may be propagated by plasticity-enhancing treatments. The myelin-associated nerve outgrowth inhibitor Nogo-A (Reticulon 4, RTN4) pathway has been shown to restrict neuroaxonal plasticity in experimental SCI models. Early randomized controlled trials are underway to investigate the effect of Nogo-A/Nogo-Receptor (NgR1) pathway blockers. This systematic review and meta-analysis of therapeutic approaches blocking the Nogo-A pathway interrogated the efficacy of functional locomotor recovery after experimental SCI according to a pre-registered study protocol. A total of 51 manuscripts reporting 76 experiments in 1572 animals were identified for meta-analysis. Overall, a neurobehavioral improvement by 18.9% (95% CI 14.5-23.2) was observed. Subgroup analysis (40 experiments, N = 890) revealed SCI-modelling factors associated with outcome variability. Lack of reported randomization and smaller group sizes were associated with larger effect sizes. Delayed treatment start was associated with lower effect sizes. Trim and Fill assessment as well as Egger regression suggested the presence of publication bias. Factoring in theoretically missing studies resulted in a reduced effect size [8.8% (95% CI 2.6-14.9)]. The available data indicates that inhibition of the Nogo-A/NgR1pathway alters functional recovery after SCI in animal studies although substantial differences appear for the applied injury mechanisms and other study details. Mirroring other SCI interventions assessed earlier we identify similar factors associated with outcome heterogeneity.


Asunto(s)
Traumatismos de la Médula Espinal , Animales , Proteínas Nogo , Vaina de Mielina/metabolismo , Modelos Animales de Enfermedad , Receptores Nogo , Médula Espinal/metabolismo , Recuperación de la Función
17.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(7): 625-631, 2022 Jul.
Artículo en Zh | MEDLINE | ID: mdl-35786457

RESUMEN

Objective To investigate the effect of Fasudil on H2O2-induced apoptosis and synaptic plasticity in human neuroblastoma SY5Y cells and its mechanism. Methods The cells were divided into three groups: PBS control group, H2O2 model group (250 µmol/L H2O2 treatment) and Fasudil intervention group (250 µmol/L H2O2 combined with 15 µg/mL Fasudil treatment). MTT assay was applied to detect cell activity and TUNEL was performed to detect cell apoptosis respectively. Immunofluorescence cytochemical staining was used to determine the expression of neurite outgrowth inhibitor A (NogoA), Nogo receptor (NgR) and synaptophysin (Syn). Western blotting was then conducted to detect the expression of NogoA, NgR, p75 neurotrophin receptor (p75NTR), leucine-rich repeat Ig domain-containing Nogo-interacting protein 1 (LINGO-1), Syn and postsynaptic density protein-95 (PSD-95). Results Compared with the PBS group, the H2O2 group showed decreased cell viability and increased apoptosis rate while Fasudil treatment significantly increased the cell viability and reduced the apoptosis rate. Compared with the H2O2 model group, Fasudil intervention increased expressions of Syn and PSD-95. Compared with the PBS group, the expression of NogoA and its receptor complex NgR/p75NTR/LINGO-1 grew significantly in the H2O2 group, suggesting Fasudil treatment could inhibit the expression of NogoA and its receptor complex NgR/p75NTR/LINGO-1. Conclusion Fasudil may inhibit the activation of the NogoA/NgR signaling pathway, therefore reducing the apoptosis induced by H2O2 in SH-SY5Y cells and enhancing the plasticity of the synapses.


Asunto(s)
Neuroblastoma , Receptores Nogo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Apoptosis , Humanos , Peróxido de Hidrógeno/farmacología , Proyección Neuronal , Plasticidad Neuronal , Receptor Nogo 1 , Receptor de Factor de Crecimiento Nervioso , Transducción de Señal
18.
Anal Cell Pathol (Amst) ; 2022: 4588999, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36600931

RESUMEN

The effect of Shenfu injection on brain injury after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) along with the underlying mechanism of axonal regeneration was explored. CA/CPR model in rats was established for subsequent experiments. A total of 160 rats were randomly divided into sham group, model group, conventional western medicine (CWM) group, Shenfu group, and antagonist group (n = 32 per group). After 3 hours, 24 hours, 3 days, and 7 days of drug administration, the modified Neurological Severity Score tests were performed. The ultrastructure of the brain and hippocampus was observed by electron microscopy. Real-time quantitative polymerase chain reaction (PCR), western blotting, and immunohistochemistry were used to detect Nogo receptor (NgR) expression in the hippocampus and cerebral cortex, and Nogo-NgR expression in CA/CPR model. Neurological deficits in the model group were severe at 3 hours, 24 hours, 3 days, and 7 days after the recovery of natural circulation, whereas the neurological deficits in CWM, antagonist, and Shenfu group were relatively mild. The ultrastructure of neuronal cells in Shenfu group had relatively complete cell membranes and more vesicles than those in the model group. The results of PCR and western blotting showed lower messenger ribonucleic acid and protein expression of NgR in Shenfu group than the model group and CWM group. Immunohistochemical examination indicated a reduction of Nogo-NgR expression in Shenfu group and antagonist group. Our results suggested that Shenfu injection reduced brain injury by attenuating Nogo-NgR signaling pathway and promoting axonal regeneration.


Asunto(s)
Lesiones Encefálicas , Paro Cardíaco , Ratas , Animales , Receptores Nogo , Ratas Sprague-Dawley , Proteínas de la Mielina/análisis , Proteínas de la Mielina/metabolismo , Proteínas Nogo , Receptores de Superficie Celular/metabolismo , Receptor Nogo 1 , Proteínas Ligadas a GPI/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Paro Cardíaco/complicaciones , Paro Cardíaco/tratamiento farmacológico
19.
Cell Rep ; 41(4): 111505, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36288715

RESUMEN

Gene-based therapeutic strategies to lower ataxin-2 levels are emerging for the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia type 2 (SCA2). Additional strategies to lower levels of ataxin-2 could be beneficial. Here, we perform a genome-wide arrayed small interfering RNA (siRNA) screen in human cells and identify RTN4R, the gene encoding the RTN4/NoGo-Receptor, as a potent modifier of ataxin-2 levels. RTN4R knockdown, or treatment with a peptide inhibitor, is sufficient to lower ataxin-2 protein levels in mouse and human neurons in vitro, and Rtn4r knockout mice have reduced ataxin-2 levels in vivo. We provide evidence that ataxin-2 shares a role with the RTN4/NoGo-Receptor in limiting axonal regeneration. Reduction of either protein increases axonal regrowth following axotomy. These data define the RTN4/NoGo-Receptor as a novel therapeutic target for ALS and SCA2 and implicate the targeting of ataxin-2 as a potential treatment following nerve injury.


Asunto(s)
Esclerosis Amiotrófica Lateral , Ataxias Espinocerebelosas , Animales , Ratones , Humanos , Ataxina-2/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , ARN Interferente Pequeño , Receptores Nogo/metabolismo , Ataxias Espinocerebelosas/genética , Ratones Noqueados , Péptidos/metabolismo , Proteínas Nogo/genética , Proteínas Nogo/metabolismo
20.
J Neuroinflammation ; 8: 113, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21906273

RESUMEN

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


Asunto(s)
Adhesión Celular/fisiología , Células Dendríticas/metabolismo , Proteínas de la Mielina/metabolismo , Vaina de Mielina/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Diferenciación Celular , Citocinas/metabolismo , Células Dendríticas/citología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Subgrupos Linfocitarios , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Monocitos/fisiología , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteínas de la Mielina/genética , Vaina de Mielina/genética , Proteínas Nogo , Receptor Nogo 1 , Proteína NgR2 , Receptores Nogo , Isoformas de Proteínas/genética , Receptores de Superficie Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA