Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Ethnopharmacol ; 272: 113943, 2021 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-33617967

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Xuesaitong (XST) is a traditional Chinese medicine injection with neuroprotective properties and has been extensively used to treat stroke for many years. The main component of XST is Panax notoginseng saponins (PNS), which is the main extract of the Chinese herbal medicine Panax notoginseng. AIM OF THE STUDY: In this study, we investigated whether XST provided long-term neuroprotection by inhibiting neurite outgrowth inhibitor-A (Nogo-A) and the ROCKII pathway in experimental rats after middle cerebral artery occlusion (MCAO) and in SH-SY5Y cells exposed to oxygen-glucose deprivation/reperfusion (OGD/R). MATERIALS AND METHODS: Rats with permanent MCAO were administered XST, Y27632, XST plus Y27632, and nimodipine for 14 and 28 days. Successful MCAO onset was confirmed by 2,3,5-triphenyl tetrazolium chloride (TTC) staining. Neurological deficit score (NDS) was used to assess neurological impairment. Hematoxylin-eosin (HE) staining and immunohistochemical (IHC) analysis of synaptophysin (SYN) and postsynaptic density protein-95 (PSD-95) were performed to evaluate cerebral ischemic injury and the neuroprotective capability of XST. Nogo-A levels and the ROCKII pathway were detected by IHC analysis, western blotting, and quantitative real-time polymerase chain reaction (qRT-PCR) to explore the protective mechanism of XST. OGD/R model was established in SH-SY5Y cells. Cell counting kit 8 (CCK8) was applied to detect the optimum OGD time and XST concentration. The expression levels Nogo-A and ROCKII pathway were determined using western blotting. RESULTS: Our results showed that XST reduced neurological dysfunction and pathological damage, promoted weight gain and synaptic regeneration, reduced Nogo-A mRNA and protein levels, and inhibited the ROCKII pathway in MCAO rats. CCK8 assay displayed that the optimal OGD time and optimal XST concentration were 7 h and 20 µg/mL respectively in SH-SY5Y cells. XST could evidently inhibit OGD/R-induced Nogo-A protein expression and ROCKII pathway activation in SH-SY5Y cells. CONCLUSIONS: The present study suggested that XST exerted long-term neuroprotective effects that assisted in stroke recovery, possibly through inhibition of the ROCKII pathway.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Saponinas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Medicamentos Herbarios Chinos/uso terapéutico , Humanos , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Panax notoginseng/química , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico , Saponinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Sinaptofisina/metabolismo , Factores de Tiempo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
2.
Drug Des Devel Ther ; 14: 2775-2787, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32764877

RESUMEN

PURPOSE: The aim of this study was to evaluate the neuroprotective effect of tanshinone IIA (TSA) on focal cerebral ischemia in rats and to investigate whether it was associated with Nogo-A/NgR1/RhoA/Rho-associated protein kinase 2 (ROCKII)/myosin light chain (MLC) signaling. METHODS: In this study, focal cerebral ischemia animal model was used. Neurological deficit scores and infarction volume were investigated to evaluate the neuroprotection of TSA. Hematoxylin-eosin staining, Nissl staining, and immunofluorescence staining were conducted to detect ischemic changes in brain tissue and changes in neurofilament protein 200 (NF200) and growth-associated protein-43 (GAP-43) expression, respectively. Western blotting and qRT-PCR analyses were used to detect the expression levels of NF200, GAP-43 and Nogo-A/NgR1/RhoA/ROCKII/MLC pathway-related signaling molecules. RESULTS: TSA treatment can improve the survival rate of rats, reduce the neurological score and infarct volume, and reduce neuron damage. In addition, TSA also increased axon length and enhanced expression of NF200 and GAP-43. Importantly, TSA significantly attenuated the expression of Nogo-A, NgR1, RhoA, ROCKII, and p-MLC, and thus inhibiting the activation of this signaling pathway. CONCLUSION: TSA promoted axonal regeneration by inhibiting the Nogo-A/NgR1/RhoA/ROCKII/MLC signaling pathway, thereby exerting neuroprotective effects in cerebral ischemia rats, which provided support for the clinical application of TSA in stroke treatment.


Asunto(s)
Abietanos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Axones/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Abietanos/química , Abietanos/aislamiento & purificación , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/aislamiento & purificación , Axones/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Humanos , Estructura Molecular , Cadenas Ligeras de Miosina/antagonistas & inhibidores , Cadenas Ligeras de Miosina/metabolismo , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/metabolismo , Receptor Nogo 1/antagonistas & inhibidores , Receptor Nogo 1/metabolismo , Ratas , Ratas Sprague-Dawley , Salvia miltiorrhiza/química , Transducción de Señal/efectos de los fármacos , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho
3.
Neurotherapeutics ; 17(3): 1153-1159, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32378027

RESUMEN

Antibody-based therapeutics targeting CNS antigens emerge as promising treatments in neurology. However, access to the CNS is limited by the blood-brain barrier. We examined the effects of a neurite growth-enhancing anti-Nogo A antibody therapy following 3 routes of administration-intrathecal (i.t.), intravenous (i.v.), and subcutaneous (s.c.)-after large photothrombotic strokes in adult rats. Intrathecal treatment of full-length IgG anti-Nogo A antibodies enhanced recovery of the grasping function, but intravenous or subcutaneous administration had no detectable effect in spite of large amounts of antibodies in the peripheral circulation. Thus, in contrast to intravenous and subcutaneous delivery, intrathecal administration is an effective and reliable way to target CNS antigens. Our data reveal that antibody delivery to the CNS is far from trivial. While intrathecal application is feasible and guarantees defined antibody doses in the effective range for a biological function, the identification and establishment of easier routes of administration remains an important task to facilitate antibody-based future therapies of CNS disorders.


Asunto(s)
Anticuerpos/administración & dosificación , Fármacos del Sistema Nervioso Central/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Proteínas Nogo/antagonistas & inhibidores , Accidente Cerebrovascular/tratamiento farmacológico , Administración Intravenosa , Animales , Anticuerpos/metabolismo , Fármacos del Sistema Nervioso Central/metabolismo , Femenino , Inyecciones Espinales , Inyecciones Subcutáneas , Proteínas Nogo/metabolismo , Ratas , Ratas Long-Evans , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/metabolismo , Resultado del Tratamiento
4.
Cell Death Dis ; 11(2): 101, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029703

RESUMEN

N-Methyl-D-aspartate (NMDA)-induced neuronal cell death is involved in a large spectrum of diseases affecting the brain and the retina such as Alzheimer's disease and diabetic retinopathy. Associated neurological impairments may result from the inhibition of neuronal plasticity by Nogo-A. The objective of the current study was to determine the contribution of Nogo-A to NMDA excitotoxicity in the mouse retina. We observed that Nogo-A is upregulated in the mouse vitreous during NMDA-induced inflammation. Intraocular injection of a function-blocking antibody specific to Nogo-A (11C7) was carried out 2 days after NMDA-induced injury. This treatment significantly enhanced visual function recovery in injured animals. Strikingly, the expression of potent pro-inflammatory molecules was downregulated by 11C7, among which TNFα was the most durably decreased cytokine in microglia/macrophages. Additional analyses suggest that TNFα downregulation may stem from cofilin inactivation in microglia/macrophages. 11C7 also limited gliosis presumably via P.Stat3 downregulation. Diabetic retinopathy was associated with increased levels of Nogo-A in the eyes of donors. In summary, our results reveal that Nogo-A-targeting antibody can stimulate visual recovery after retinal injury and that Nogo-A is a potent modulator of excitotoxicity-induced neuroinflammation. These data may be used to design treatments against inflammatory eye diseases.


Asunto(s)
Células Amacrinas/efectos de los fármacos , Antiinflamatorios/farmacología , Anticuerpos Neutralizantes/farmacología , Proteínas Nogo/antagonistas & inhibidores , Células Ganglionares de la Retina/efectos de los fármacos , Retinitis/prevención & control , Visión Ocular/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Células Amacrinas/metabolismo , Células Amacrinas/patología , Animales , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , N-Metilaspartato , Plasticidad Neuronal/efectos de los fármacos , Proteínas Nogo/metabolismo , Fosforilación , Recuperación de la Función , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Retinitis/inducido químicamente , Retinitis/metabolismo , Retinitis/fisiopatología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
5.
Neural Dev ; 15(1): 1, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31918754

RESUMEN

As humans, we cannot regenerate axons within the central nervous system (CNS), therefore, making any damage to it permanent. This leads to the loss of sensory and motor function below the site of injury and can be crippling to a person's health. Spontaneous recovery can occur from plastic changes, but it is minimal. The absence of regeneration is due to the inhibitory environment of the CNS as well as the inherent inability of CNS axons to form growth cones. Amongst many factors, one of the major inhibitory signals of the CNS environment is the myelin-associated Nogo pathway. Nogo-A, Nogo-B and Nogo-C (Nogo), stimulate the Nogo receptor, inhibiting neurite outgrowth by causing growth cones to collapse through activation of Rho Kinase (ROCK). Antibodies can be used to target this signalling pathway by binding to Nogo and thus promote the outgrowth of neuronal axons in the CNS. This use of anti-Nogo antibodies has been shown to upregulate CNS regeneration as well as drastically improve sensory and motor function in both rats and primates when coupled with adequate training. Here, we evaluate whether the experimental success of anti-Nogo at improving CNS regeneration can be carried over into the clinical setting to treat spinal cord injuries (SCI) and their symptoms successfully. Furthermore, we also discuss potential methods to improve the current treatment and any developmental obstacles.


Asunto(s)
Inmunoterapia/métodos , Regeneración Nerviosa , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/inmunología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/fisiopatología , Animales , Anticuerpos/administración & dosificación , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/fisiología , Humanos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
6.
CNS Neurosci Ther ; 26(2): 260-269, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31418518

RESUMEN

In spinal cord injured adult mammals, neutralizing the neurite growth inhibitor Nogo-A with antibodies promotes axonal regeneration and functional recovery, although axonal regeneration is limited in length. Neurotrophic factors such as BDNF stimulate neurite outgrowth and protect axotomized neurons. Can the effects obtained by neutralizing Nogo-A, inducing an environment favorable for axonal sprouting, be strengthened by adding BDNF? A unilateral incomplete hemicord lesion at C7 level interrupted the main corticospinal component in three groups of adult macaque monkeys: control monkeys (n = 6), anti-Nogo-A antibody-treated monkeys (n = 7), and anti-Nogo-A antibody and BDNF-treated monkeys (n = 5). The functional recovery of manual dexterity was significantly different between the 3 groups of monkeys, the lowest in the control group. Whereas the anti-Nogo-A antibody-treated animals returned to manual dexterity performances close to prelesion ones, irrespective of lesion size, both the control and the anti-Nogo-A/BDNF animals presented a limited functional recovery. In the control group, the limited spontaneous functional recovery depended on lesion size, a dependence absent in the combined treatment group (anti-Nogo-A antibody and BDNF). The functional recovery in the latter group was significantly lower than in anti-Nogo-A antibody-treated monkeys, although the lesion was larger in three out of the five monkeys in the combined treatment group.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Trastornos del Movimiento/tratamiento farmacológico , Proteínas Nogo/antagonistas & inhibidores , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Axones , Médula Cervical/lesiones , Mano , Macaca fascicularis , Masculino , Destreza Motora , Trastornos del Movimiento/etiología , Regeneración Nerviosa , Desempeño Psicomotor/efectos de los fármacos , Recuperación de la Función , Traumatismos de la Médula Espinal/complicaciones
7.
Mol Med Rep ; 21(1): 77-88, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31746353

RESUMEN

Brain injury after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) is the main cause of neurological dysfunction and death in cardiac arrest. To assess the effect of Nogo­A antibody on brain function in rats following CPR and to explore the underlying mechanisms, CA/CPR (ventricular fibrillation) rats were divided into the CPR+Nogo­A, CPR+saline and sham groups. Hippocampal caspase­3 levels were detected by RT­PCR and immunoblotting. Next, Nogo­A, glucose regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), cysteinyl aspartate specific proteinase­12 (casapse­12), Bcl­2 and Bax protein levels in the hippocampus were detected by immunoblotting. Coronal brain sections were analyzed by TUNEL assay to detect apoptosis at 72 h, while Nissl staining and electron microscopy were performed to detect Nissl bodies and microstructure at 24 h, respectively. Finally, rats were assessed for neurologic deficits at various times. Nissl staining revealed morphological improvement after Nogo­A antibody treatment. Sub­organelle structure was preserved as assessed by electron microscopy in model animals post­antibody treatment; neurological function was improved as well (P<0.05), while the apoptosis index was decreased (26.2±9.85 vs. 46.6±12.95%; P<0.05). Hippocampal caspase­3 mRNA and protein, Nogo­A protein levels were significantly decreased after antibody treatment (P<0.05). Hippocampal Nogo­A expression was positively correlated with caspase­3 (Pearson's correlation; r=0.790, P=0.000). Hippocampal GRP78 and Bcl­2 protein levels were higher after antibody treatment than these levels noted in the model animals (P<0.05), while CHOP, caspase­12 and Bax levels were reduced (P<0.05). Nogo­A antibody ameliorates neurological function after restoration of spontaneous circulation (ROSC), possibly by suppressing apoptosis induced by endoplasmic reticulum stress.


Asunto(s)
Anticuerpos/farmacología , Apoptosis , Reanimación Cardiopulmonar , Paro Cardíaco/metabolismo , Hipocampo/metabolismo , Proteínas Nogo/antagonistas & inhibidores , Recuperación de la Función , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Paro Cardíaco/patología , Paro Cardíaco/terapia , Hipocampo/patología , Masculino , Proteínas Nogo/biosíntesis , Ratas , Ratas Wistar
8.
Proc Natl Acad Sci U S A ; 116(28): 14270-14279, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31235580

RESUMEN

Stroke is a major cause of serious disability due to the brain's limited capacity to regenerate damaged tissue and neuronal circuits. After ischemic injury, a multiphasic degenerative and inflammatory response is coupled with severely restricted vascular and neuronal repair, resulting in permanent functional deficits. Although clinical evidence indicates that revascularization of the ischemic brain regions is crucial for functional recovery, no therapeutics that promote angiogenesis after cerebral stroke are currently available. Besides vascular growth factors, guidance molecules have been identified to regulate aspects of angiogenesis in the central nervous system (CNS) and may provide targets for therapeutic angiogenesis. In this study, we demonstrate that genetic deletion of the neurite outgrowth inhibitor Nogo-A or one of its corresponding receptors, S1PR2, improves vascular sprouting and repair and reduces neurological deficits after cerebral ischemia in mice. These findings were reproduced in a therapeutic approach using intrathecal anti-Nogo-A antibodies; such a therapy is currently in clinical testing for spinal cord injury. These results provide a basis for a therapeutic blockage of inhibitory guidance molecules to improve vascular and neural repair after ischemic CNS injuries.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Isquemia Encefálica/tratamiento farmacológico , Proteínas Nogo/genética , Receptores de Esfingosina-1-Fosfato/genética , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Neuronas/efectos de los fármacos , Neuronas/patología , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/inmunología , Tractos Piramidales/efectos de los fármacos , Tractos Piramidales/patología , Recuperación de la Función/genética , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Receptores de Esfingosina-1-Fosfato/inmunología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/patología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología
9.
J Neurosci ; 39(21): 4066-4076, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-30902870

RESUMEN

Loss of bladder control is common after spinal cord injury (SCI) and no causal therapies are available. Here we investigated whether function-blocking antibodies against the nerve-fiber growth inhibitory protein Nogo-A applied to rats with severe SCI could prevent development of neurogenic lower urinary tract dysfunction. Bladder function of rats with SCI was repeatedly assessed by urodynamic examination in fully awake animals. Four weeks after SCI, detrusor sphincter dyssynergia had developed in all untreated or control antibody-infused animals. In contrast, 2 weeks of intrathecal anti-Nogo-A antibody treatment led to significantly reduced aberrant maximum detrusor pressure during voiding and a reduction of the abnormal EMG high-frequency activity in the external urethral sphincter. Anatomically, we found higher densities of fibers originating from the pontine micturition center in the lumbosacral gray matter in the anti-Nogo-A antibody-treated animals, as well as a reduced number of inhibitory interneurons in lamina X. These results suggest that anti-Nogo-A therapy could also have positive effects on bladder function clinically.SIGNIFICANCE STATEMENT After spinal cord injury, loss of bladder control is common. Detrusor sphincter dyssynergia is a potentially life-threatening consequence. Currently, only symptomatic treatment options are available. First causal treatment options are urgently needed in humans. In this work, we show that function-blocking antibodies against the nerve-fiber growth inhibitory protein Nogo-A applied to rats with severe spinal cord injury could prevent development of neurogenic lower urinary tract dysfunction, in particular detrusor sphincter dyssynergia. Anti-Nogo-A therapy has entered phase II clinical trial in humans and might therefore soon be the first causal treatment option for neurogenic lower urinary tract dysfunction.


Asunto(s)
Anticuerpos/farmacología , Proteínas Nogo/antagonistas & inhibidores , Traumatismos de la Médula Espinal/complicaciones , Vejiga Urinaria Neurogénica/etiología , Animales , Femenino , Ratas , Ratas Endogámicas Lew
10.
Mol Oncol ; 12(12): 2042-2054, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30019429

RESUMEN

Tumor angiogenesis is one of the hallmarks of cancer as well as an attractive target for cancer therapy. Characterization of novel pathways that act in parallel with the VEGF/VEGFR axis to promote tumor angiogenesis may provide insights into novel anti-angiogenic therapeutic targets. We found that the expression level of Nogo-B is positively correlated with tumor vessel density in hepatocellular carcinoma (HCC). While Nogo-B depletion inhibited tumor angiogenesis, Nogo-B overexpression promoted tumor angiogenesis in a tumor xenograft subcutaneous model of the human HCC cell line. Mechanically, Nogo-B regulates tumor angiogenesis based on its association with integrin αv ß3 and activation of focal adhesion kinase. Moreover, Nogo-B antibody successfully abolished the function of Nogo-B in tumor angiogenesis in vitro and in vivo. Collectively, our results strongly suggest that Nogo-B is an important tumor angiogenic factor and blocking Nogo-B selectively inhibits tumor angiogenesis.


Asunto(s)
Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/patología , Neovascularización Patológica/patología , Proteínas Nogo/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Adhesión Celular , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones Endogámicos C57BL , Ratones Desnudos , Terapia Molecular Dirigida , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Proteínas Nogo/análisis , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/genética
11.
Glia ; 66(10): 2079-2093, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30051920

RESUMEN

Nogo-A is a potent glial-derived inhibitor of axon growth in the injured CNS and acts as a negative regulator of developmental angiogenesis by inhibiting vascular endothelial cell migration. However, its function in pathological angiogenesis has never been studied after ischemic injury in the CNS. Using the mouse model of oxygen-induced retinopathy (OIR) which yields defined zones of retinal ischemia, our goal was to investigate the role of Nogo-A in vascular regeneration. We demonstrate a marked upregulation of the Nogo-A receptor sphingosine 1-phosphate receptor 2 in blood vessels following OIR, while Nogo-A is abundantly expressed in surrounding glial cells. Acute inhibition of Nogo-A with function-blocking antibody 11C7 significantly improved vascular regeneration and consequently prevented pathological pre-retinal angiogenesis. Ultimately, inhibition of Nogo-A led to restoration of retinal function as determined by electrophysiological response of retinal cells to light stimulation. Our data suggest that anti-Nogo-A antibody may protect neuronal cells from ischemic damage by accelerating blood vessel repair in the CNS. Targeting Nogo-A by immunotherapy may improve CNS perfusion after vascular injuries.


Asunto(s)
Isquemia/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas Nogo/metabolismo , Regeneración/fisiología , Enfermedades de la Retina/metabolismo , Vasos Retinianos/metabolismo , Inductores de la Angiogénesis/farmacología , Animales , Modelos Animales de Enfermedad , Isquemia/tratamiento farmacológico , Isquemia/patología , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuroglía/patología , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Regeneración/efectos de los fármacos , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/patología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/patología , Receptores de Esfingosina-1-Fosfato , Visión Ocular/efectos de los fármacos , Visión Ocular/fisiología
12.
J Cereb Blood Flow Metab ; 38(8): 1327-1338, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28952904

RESUMEN

Many preclinical treatment strategies for stroke have failed when tested in human trials. Although the reasons for these translation failures are multifactorial, one potential concern is the statistical analysis of the preclinical data. One way to rigorously evaluate new therapies is to use an intention-to-treat analysis in preclinical studies. Therefore, in this study, we set out to evaluate the treatment efficacy of a potential clinically relevant therapeutic agent for stroke, i.e., anti-Nogo-A immunotherapy, using an intention-to-treat analysis. Adult rats were trained on the skilled forelimb reaching task and subsequently underwent an ischemic stroke. Nine weeks later, the rats either received intracerebroventricular anti-Nogo-A antibody, control antibody, or no treatment. Skilled reaching performance was assessed by a non-linear model using both an intention-to-treat and per-protocol analysis. Following testing, dendritic complexity was evaluated in the contralesional and perilesional sensorimotor cortex. Both intention-to-treat and per-protocol analysis showed that anti-Nogo-A immunotherapy resulted in statistically significant improved recovery on the skilled forelimb reaching task, although treatment effect was less (though statistically significant) in the intention-to-treat group. Improved functional performance was not shown to be associated with dendritic changes. In conclusion, this study provides evidence for the importance of using intention-to-treat paradigms in testing preclinical therapeutic strategies.


Asunto(s)
Anticuerpos/uso terapéutico , Inmunoterapia , Proteínas Nogo/antagonistas & inhibidores , Accidente Cerebrovascular/terapia , Animales , Dendritas/efectos de los fármacos , Dendritas/patología , Evaluación Preclínica de Medicamentos , Inmunoterapia/métodos , Análisis de Intención de Tratar , Masculino , Corteza Motora/efectos de los fármacos , Corteza Motora/patología , Corteza Motora/fisiopatología , Ratas , Ratas Long-Evans , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Resultado del Tratamiento
13.
Sci Rep ; 7(1): 9431, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28842591

RESUMEN

Blocking axonal growth inhibitor NogoA has been of great interest for promoting axonal recovery from neurological diseases. The present study investigates the therapeutic effects of blocking NogoA, inducing functional recovery and promoting white matter repair in an experimental animal model of stroke. Adult male rats were subjected to white matter injury by subcortical ischemic stroke. Twenty-four hours after surgery, 250 ug of anti-NogoA or anti-IgG-1 were administered through the tail vein. The quantity of NogoA protein was determined by immunohistochemistry in the brain and peripheral organs. In addition, functional status, lesion size, fiber tract integrity, axonal sprouting and white matter repair markers were analyzed. Moreover, an in vitro study was performed in order to strengthen the results obtained in vivo. A lower quantity of NogoA protein was found in the brain and peripheral organs of the animals that received anti-NogoA treatment. The animals receiving anti-NogoA treatment showed significantly better results in terms of functional recovery, fiber tract integrity, axonal sprouting and white matter repair markers compared with the control group at 28 days. White matter integrity was in part restored by antibody-mediated inhibition of NogoA administration in those animals that were subjected to an axonal injury by subcortical stroke. This white matter restoration triggered functional recovery.


Asunto(s)
Axones/metabolismo , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/metabolismo , Accidente Cerebrovascular/metabolismo , Sustancia Blanca/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Biomarcadores , Técnica del Anticuerpo Fluorescente , Vaina de Mielina/metabolismo , Células PC12 , Ratas , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología , Sustancia Blanca/patología
15.
Chem Commun (Camb) ; 53(53): 7234-7237, 2017 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-28352901

RESUMEN

Chemical genetics has arisen as a powerful approach for identifying novel anti-cancer agents. However, a major bottleneck of this approach is identifying the targets of lead compounds that arise from screens. Here, we coupled the synthesis and screening of fragment-based cysteine-reactive covalent ligands with activity-based protein profiling (ABPP) chemoproteomic approaches to identify compounds that impair colorectal cancer pathogenicity and map the druggable hotspots targeted by these hits. Through this coupled approach, we discovered a cysteine-reactive acrylamide DKM 3-30 that significantly impaired colorectal cancer cell pathogenicity through targeting C1101 on reticulon 4 (RTN4). While little is known about the role of RTN4 in colorectal cancer, this protein has been established as a critical mediator of endoplasmic reticulum tubular network formation. We show here that covalent modification of C1101 on RTN4 by DKM 3-30 or genetic knockdown of RTN4 impairs endoplasmic reticulum and nuclear envelope morphology as well as colorectal cancer pathogenicity. We thus put forth RTN4 as a potential novel colorectal cancer therapeutic target and reveal a unique druggable hotspot within RTN4 that can be targeted by covalent ligands to impair colorectal cancer pathogenicity. Our results underscore the utility of coupling the screening of fragment-based covalent ligands with isoTOP-ABPP platforms for mining the proteome for novel druggable nodes that can be targeted for cancer therapy.


Asunto(s)
Acrilamida/farmacología , Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Cisteína/química , Retículo Endoplásmico/efectos de los fármacos , Proteínas Nogo/antagonistas & inhibidores , Proteómica , Acrilamida/química , Antineoplásicos/química , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Retículo Endoplásmico/metabolismo , Humanos , Ligandos , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Membrana Nuclear/efectos de los fármacos , Membrana Nuclear/metabolismo
16.
Cell Physiol Biochem ; 41(1): 274-285, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28214833

RESUMEN

BACKGROUND/AIMS: Nogo-B, a member of the reticulon family of proteins, is mainly located in the endoplasmic reticulum (ER). Here, we investigate the function and mechanism of Nogo-B in the regulation of TLR4-associated immune responses in the macrophage cell line of RAW264.7. METHODS: Nogo-B was up- and down-regulated through the use of appropriate adenoviral vectors or siRNA, and the effects of Nogo-B on macrophages under liposaccharide (LPS) stimulation were evaluated via western blotting, immunofluorescence, enzyme-linked immunosorbent assay (ELISA), flow cytometric analysis, and transwell assay. RESULTS: Our data indicates that the protein of Nogo-B was down-regulated in a time- and dose-dependent manner following LPS administration in the macrophage. Nogo-B overexpression increased the production of inflammatory cytokines (MCP-1, TNF-α, IL-1ß, and TGF-ß), enhanced macrophage migration activities, activated major histocompatibility complex II (MHC II), and elevated the expression of macrophage scavenger receptor 1(MSR1), all of which suggest that Nogo-B is necessary for immune responses and plays an important role in regulating macrophage recruitment. Mechanistically, Nogo-B may enhance TLR4 expression in macrophage surfaces, activate mitogen-activated protein kinase (MAPK) pathways, and initiate inflammatory responses. CONCLUSION: These findings illustrate the key regulatory functions of Nogo-B in facilitating LPS-mediated immune responses through promoting the phosphorylation of MAP kinase.


Asunto(s)
Lipopolisacáridos/toxicidad , Proteínas Nogo/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Citocinas/análisis , Citocinas/genética , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Microscopía Fluorescente , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/genética , Células RAW 264.7 , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Depuradores de Clase A/metabolismo , Receptor Toll-Like 4/metabolismo
17.
Mol Neurobiol ; 54(10): 8404-8418, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27933584

RESUMEN

Myelin-associated inhibitors, such as NogoA, myelin-associated glycoprotein (MAG), and oligodendrocyte myelin glycoprotein (OMgp), play a pivotal role in the lack of neuroregeneration in multiple sclerosis, an inflammatory demyelinating disease of the central nervous system (CNS). Matrine (MAT), a monomer that is used in traditional Chinese medicine as an anti-inflammatory agent, has shown beneficial effects in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. However, the underlying mechanisms of MAT-induced EAE amelioration are not fully understood. In the present study, we show that MAT treatment suppressed ongoing EAE, and this effect correlated with an increased expression of growth-associated protein 43, an established marker for axonal regeneration. MAT treatment significantly reduced the levels of NogoA, its receptor complex NgR/p75NTR/LINGO-1, and their downstream RhoA/ROCK signaling pathway in the CNS. In contrast, intracellular cyclic AMP (cAMP) levels and its protein kinase (protein kinase A (PKA)), which can promote axonal regrowth by inactivating the RhoA, were upregulated. Importantly, adding MAT in primary astrocytes in vitro largely induced cAMP/PKA expression, and blockade of cAMP significantly diminished MAT-induced expression of PKA and production of BDNF, a potent neurotrophic factor for neuroregeneration. Taken together, our findings demonstrate that the beneficial effects of MAT on EAE can be attributed not only to its capacity for immunomodulation, but also to its directly promoting regeneration of the injured CNS.


Asunto(s)
Alcaloides/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Inhibición Neural/fisiología , Proteínas Nogo/metabolismo , Quinolizinas/uso terapéutico , Transducción de Señal/fisiología , Alcaloides/farmacología , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Células Cultivadas , Femenino , Cobayas , Ratones , Inhibición Neural/efectos de los fármacos , Proteínas Nogo/antagonistas & inhibidores , Quinolizinas/farmacología , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Matrinas
18.
Cereb Cortex ; 27(5): 2779-2792, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27166169

RESUMEN

Nogo-A restricts long-term potentiation (LTP) at the Schaffer collateral-CA1 pathway in the adult hippocampus via 2 extracellular domains: Nogo-A-Δ20 and Nogo-66. Nogo-66 signals via Nogo Receptor 1 (NgR1) to regulate synaptic function. Whether the NgR1 coreceptors Lingo1 and p75NTR are involved in the signaling in this context is still not known. Moreover, the intracellular cascade mediating the activity of Nogo-66 in restricting LTP is unexplored. We combine electrophysiology and biochemistry in acute hippocampal slices and demonstrate that a loss of function for Lingo1 results in a significant increase in LTP levels at the Schaffer collateral-CA1 pathway, and that Lingo1 is the NgR1 coreceptor mediating the role of Nogo-66 in restricting LTP. Our data show that p75NTR is not involved in mediating the Nogo-66 effect on LTP. Moreover, loss of function for p75NTR and NgR1 equally attenuate LTD, suggesting that p75NTR might mediate the NgR1-dependent regulation of LTD, independently of Nogo-66. Finally, our results indicate that Nogo-66 signaling limits LTP via the ROCK2-Cofilin pathway to control the dynamics of the actin cytoskeleton. The present results elucidate the signaling pathway activated by Nogo-66 to control LTP and contribute to the understanding of how Nogo-A stabilizes the neural circuits to limit activity-dependent plasticity events in the mature hippocampus.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Plasticidad Neuronal/fisiología , Proteínas Nogo/metabolismo , Transducción de Señal/fisiología , Quinasas Asociadas a rho/metabolismo , Factores Despolimerizantes de la Actina/genética , Amidas/farmacología , Animales , Biofisica , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Femenino , Hipocampo , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/efectos de los fármacos , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/química , Técnicas de Placa-Clamp , Péptidos/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Piridinas/farmacología , Receptor de Factor de Crecimiento Nervioso/deficiencia , Receptor de Factor de Crecimiento Nervioso/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética
19.
Sci Rep ; 6: 35969, 2016 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-27786289

RESUMEN

Reticulons (RTNs) are a large family of membrane associated proteins with various functions. NOGO-A/RTN4A has a well-known function in limiting neurite outgrowth and restricting the plasticity of the mammalian central nervous system. On the other hand, Reticulon 4 proteins were shown to be involved in forming and maintaining endoplasmic reticulum (ER) tubules. Using comparative transcriptome analysis and qPCR, we show here that NOGO-B/RTN4B and NOGO-A/RTN4A are simultaneously expressed in cultured epithelial, fibroblast and neuronal cells. Electron tomography combined with immunolabelling reveal that both isoforms localize preferably to curved membranes on ER tubules and sheet edges. Morphological analysis of cells with manipulated levels of NOGO-B/RTN4B revealed that it is required for maintenance of normal ER shape; over-expression changes the sheet/tubule balance strongly towards tubules and causes the deformation of the cell shape while depletion of the protein induces formation of large peripheral ER sheets.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas Nogo/genética , Animales , Línea Celular , Forma de la Célula , Células Cultivadas , Retículo Endoplásmico/ultraestructura , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Perfilación de la Expresión Génica , Humanos , Ratones , Microscopía Inmunoelectrónica , Células 3T3 NIH , Neuronas/metabolismo , Neuronas/ultraestructura , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/metabolismo , Isoformas de Proteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA