Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38798506

RESUMEN

Monocytes are circulating macrophage precursors and are generated from bone marrow hematopoietic stem cells. In the adults, monocytes continuously replenish cerebral border-associated macrophages under a physiological condition. Monocytes also rapidly infiltrate into the brain in the settings of pathological conditions. The mechanisms of recruiting monocyte-derived macrophages into the brain under pathological conditions have been extensively studied. However, it remains unclear how monocytes enter the brain for renewal of border-associated macrophages under the physiological condition. Using both in vitro and in vivo approaches, this study reveals that the combination of two hematopoietic growth factors, stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF), complementarily and synergistically enhances adhesion of monocytes to cerebral endothelial cells in a dose dependent manner. Cysteine-cysteine chemokine receptor 5 (CCR5) in brain endothelial cells, but not cell adhesion molecules mediating neuroinflammation-related infiltration of monocyte-derived macrophages, modulates the SCF+G-CSF-enhanced monocyte-endothelial cell adhesion. Blocking CCR5 or genetically deleting CCR5 reduces monocyte-endothelial cell adhesion induced by SCF+G-CSF. SCF+G-CSF-enhanced recruitment of bone marrow-derived monocytes/macrophages in cerebral perivascular space is also reduced in adult CCR5 knockout mice. This study demonstrates the contribution of SCF and G-CSF in regulating the entry of monocytes into the adult brain to replenish perivascular macrophages.

2.
Mol Neurobiol ; 2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38135853

RESUMEN

Subarachnoid hemorrhage (SAH) is a devastating disease associated with a high mortality and morbidity. Exosomes have been considered as a potential therapeutic target for SAH. However, the effect of exosomes in SAH remains to be elucidated. In this study, we focused on investigating the effect of plasma exosomal lncRNA TM7SF3-AU1 in white matter injury after SAH. The SAH model was established by means of endovascular perforation. Exosomes were extracted from rat plasma samples. The expression of RNAs in the exosomes was detected by the transcriptomic microarray. Differentially expressed circRNA, lncRNA, and mRNA were obtained. The ceRNA network showed that the lncRNA TM7SF3-AU1 and miR-702-3p were closely associated with SARM1. Knocking down TM7SF3-AU1 promoted the expression of miR-702-3p and suppressed the expression of SARM1, and knocking down TM7SF3-AU1 also attenuated white matter injury after SAH. In addition, knocking down TM7SF3-AU1 improved the neurological deficits in locomotion, anxiety, learning, memory, and electrophysiological activity after SAH. Mechanistically, TM7SF3-AU1 was able to absorb miR-702-3p, which directly bind the SARM1 mRNA. Furthermore, the white matter injury attenuated by knockdown of TM7SF3-AU1 was partially reversed by the miR-702-3p antagomir in SAH rats. Taken together, this study showed that TM7SF3-AU1 acts as a sponge for miR-702-3p, reducing the inhibitory effect of miR-702-3p on SARM1, resulting in increased SARM1 expression and thus leading to white matter injury after SAH. Our study provides new insights into exosome-associated white matter injury. It also highlights TM7SF3-AU1 as a potential therapeutic target for white matter injury after SAH.

3.
Neural Regen Res ; 17(8): 1776-1784, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35017438

RESUMEN

As a highly evolutionary conserved long non-coding RNA, metastasis associated lung adenocarcinoma transcript 1 (MALAT1) was first demonstrated to be related to lung tumor metastasis by promoting angiogenesis. To investigate the role of MALAT1 in traumatic brain injury, we established mouse models of controlled cortical impact and cell models of oxygen-glucose deprivation to mimic traumatic brain injury in vitro and in vivo. The results revealed that MALAT1 silencing in vitro inhibited endothelial cell viability and tube formation but increased migration. In MALAT1-deficient mice, endothelial cell proliferation in the injured cortex, functional vessel density and cerebral blood flow were reduced. Bioinformatic analyses and RNA pull-down assays validated enhancer of zeste homolog 2 (EZH2) as a downstream factor of MALAT1 in endothelial cells. Jagged-1, the Notch homolog 1 (NOTCH1) agonist, reversed the MALAT1 deficiency-mediated impairment of angiogenesis. Taken together, our results suggest that MALAT1 controls the key processes of angiogenesis following traumatic brain injury in an EZH2/NOTCH1-dependent manner.

4.
Front Neurosci ; 15: 627110, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679307

RESUMEN

BACKGROUND AND TARGET: Following brain trauma, blood-brain barrier (BBB) disruption and inflammatory response are critical pathological steps contributing to secondary injury, leading to high mortality and morbidity. Both pathologies are closely associated with endothelial remodeling. In the present study, we concentrated on annexin A1 (ANXA1) as a novel regulator of endothelial function after traumatic brain injury. METHODS: After establishing controlled cortical impact (CCI) model in male mice, human recombinant ANXA1 (rANXA1) was administered intravenously, followed by assessments of BBB integrity, brain edema, inflammatory response, and neurological deficits. RESULT: Animals treated with rANXA1 (1 µg/kg) at 1 h after CCI exhibited optimal BBB protection including alleviated BBB disruption and brain edema, as well as endothelial junction proteins loss. The infiltrated neutrophils and inflammatory cytokines were suppressed by rANXA1, consistent with decreased adhesive and transmigrating molecules from isolated microvessels. Moreover, rANXA1 attenuated the neurological deficits induced by CCI. We further found that the Ras homolog gene family member A (RhoA) inhibition has similar effect as rANXA1 in ameliorating brain injuries after CCI, whereas rANXA1 suppressed CCI-induced RhoA activation. CONCLUSION: Our findings suggest that the endothelial remodeling by exogenous rANXA1 corrects BBB disruption and inflammatory response through RhoA inhibition, hence improving functional outcomes in CCI mice.

6.
Cell Death Dis ; 12(1): 9, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33414448

RESUMEN

The level of microRNA-9-5p (miRNA-9-5p) in brain tissues is significantly changed in the chronic phase after traumatic brain injury (TBI). However, the effect of miRNA-9-5p on brain function after TBI has not been elucidated. In this study, we used a controlled cortical impact (CCI) model to induce TBI in Sprague-Dawley rats. Brain microvascular endothelial cells (BMECs), astrocytes, and neurons were extracted from immature Sprague-Dawley rats and cocultured to reconstruct the neurovascular unit (NVU) in vitro. The results showed that downregulation of miRNA-9-5p in the chronic phase contributed to neurological function recovery by promoting astrocyte proliferation and increasing the release of astrocyte-derived neurotrophic factors around injured brain tissues after TBI. A dual-luciferase reporter assay validated that miRNA-9-5p was a post-transcriptional modulator of thrombospondin 2 (Thbs-2), and downregulation of miRNA-9-5p promoted Thbs-2 expression in astrocytes. Furthermore, we verified that Thbs-2 can promote Notch pathway activation by directly binding to Jagged and Notch. Through in vitro experiments, we found that the expression of synaptic proteins and the number of synaptic bodies were increased in neurons in the NVU, which was constructed using astrocytes pretreated with miRNA-9-5p inhibitor. Moreover, we also found that downregulation of miRNA-9-5p promoted Thbs-2 expression in astrocytes, which activated the Notch/cylindromatosis/transforming growth factor-ß-activated kinase 1 pathway in neurons and promoted the expression of synaptic proteins, including post-synaptic density protein 95 and synaptotagmin. Based on these results, miRNA-9-5p may be a new promising prognostic marker and treatment target for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/genética , MicroARNs/metabolismo , Sinapsis/metabolismo , Animales , Regulación hacia Abajo , Ratas , Ratas Sprague-Dawley
7.
Exp Neurol ; 337: 113596, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33417892

RESUMEN

Subarachnoid haemorrhage (SAH) is a devastating cerebrovascular disease which has a high morbidity and mortality. The phenotypic transformation of smooth muscle cells (SMCs) lead to neurovascular injury after SAH. However, the underlying mechanism remains unclear. In the present study, we aimed to investigate the potential role of ET-1/ETAR on the phenotypic transformation of SMCs after SAH. The models of SAH were established in vivo and vitro. We observed ET-1 secretion by endothelial cells was increased, and the phenotypic transformation of SMCs was aggravated after SAH. Knocking down ETAR inhibited the phenotypic transformation of SMCs, decreased the migration ability of SMCs in vitro. Moreover, Knocking down ETAR ameliorated cerebral ischaemia and alleviated dysfunction of neurological function in vivo. In addition, Exogenous ET-1 increased the migration ability of SMCs and aggravated the phenotypic transformation of SMCs in vitro, which were partly reversed by the antagonist of Erk1/2 - SCH772984. Taken together, our results demonstrated that endothelial ET-1 aggravated the phenotypic transformation of SMCs after SAH. Knocking down ETAR inhibited the phenotypic transformation of SMCs through ERK/KLF4 thus ameliorating neurovascular injury after SAH. We also revealed that ET-1/ETAR is a potential therapeutic target after SAH.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , Sistema de Señalización de MAP Quinasas/genética , Miocitos del Músculo Liso/patología , Receptores de Endotelina/genética , Hemorragia Subaracnoidea/genética , Animales , Animales Modificados Genéticamente , Movimiento Celular , Circulación Cerebrovascular , Células Endoteliales/metabolismo , Endotelina-1/genética , Técnicas de Silenciamiento del Gen , Humanos , Factor 4 Similar a Kruppel , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Fenotipo , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Receptor de Endotelina A , Hemorragia Subaracnoidea/patología
8.
Exp Neurol ; 334: 113462, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32916173

RESUMEN

Traumatic brain injury (TBI) has been regarded as one of the leading cause of injury-related death and disability. White matter injury after TBI is characterized by axon damage and demyelination, resulting in neural network impairment and neurological deficit. Brain-derived neurotrophic factor (BDNF) can promote white matter repair. The activation of peroxisome proliferator-activated receptor gamma (PPARγ) has been reported to promote microglia/macrophages towards anti-inflammatory state and therefore to promote axon regeneration. Bexarotene, an agonist of retinoid X receptor (RXR), can activate RXR/PPARγ heterodimers. The aim of the present study was to identify the effect of bexarotene on BDNF in microglia/macrophages and axon sprouting after TBI in mice. Bexarotene was administered intraperitoneally in C57BL/6 mice undergoing controlled cortical impact (CCI). PPARγ dependency was determined by intraperitoneal administration of a PPARγ antagonist T0070907. We found that bexarotene promoted axon regeneration indicated by increased growth associated protein 43 (GAP43) expression, myelin basic protein (MBP) expression, and biotinylated dextran amine (BDA)+ axon sprouting. Bexarotene also increased microglia/macrophages-specific brain derived neurotrophic factor (BDNF) expression after TBI. In addition, bexarotene reduced the number of pro-inflammatory microglia/macrophages while increased the number of anti-inflammatory microglia/macrophages after TBI. Moreover, bexaortene inhibited pro-inflammatory cytokine secretion. In addition, bexarotene treatment improved neurological scores and cognitive function of CCI-injured mice. These effects of bexarotene were partially abolished by T0070907. In conclusion, bexarotene promotes axon sprouting, increases microglia/macrophages-specific BDNF expression, and induces microglia/macrophages from a pro-inflammatory state towards an anti-inflammatory one after TBI at least partially in a PPARγ-dependent manner.


Asunto(s)
Axones/efectos de los fármacos , Bexaroteno/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Axones/metabolismo , Bexaroteno/farmacología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/genética , Expresión Génica , Macrófagos/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología
9.
Neuroscience ; 440: 160-174, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32502567

RESUMEN

MicroRNA-9-5p (miRNA-9-5p) is an important regulator of angiogenesis in many pathological states. However, the effect of miRNA-9-5p on angiogenesis after traumatic brain injury (TBI) has not been elucidated. In this study, a controlled cortical impact (CCI) model was used to induce TBI in Sprague-Dawley rats, and an oxygen glucose deprivation (OGD) model was used to mimic the pathological state in vitro. Brain microvascular endothelial cells (BMECs) were extracted from immature rats. The results showed that the level of miRNA-9-5p was significantly increased in the traumatic foci after TBI, and the upregulation of miRNA9-5p promoted the recovery of neurological function. Moreover, the upregulation of miRNA-9-5p with miRNA agomir significantly increased the density of the microvascular and neurons around the traumatic foci in rats after TBI. The results of the in vitro experiments confirmed that the upregulation of miRNA-9-5p with a miRNA mimic improved cellular viability and alleviated cellular apoptosis. Dual luciferase reporter assay validated that miRNA-9-5p was a posttranscriptional modulator of Ptch-1. Activation of the Hedgehog pathway by increasing the level of miRNA-9-5p promoted the migration and tube formation of BMECs in vitro. In addition, we found that the upregulation of miRNA-9-5p activated the Hedgehog pathway and increased the phosphorylation of AKT, which promoted the expression of cyclin D1, MMP-9 and VEGF in BMECs. All these results indicate that the upregulation of miRNA-9-5p promotes angiogenesis and improves neurological functional recovery after TBI, mainly by activating the Hedgehog pathway. MiRNA-9-5p may be a potential new therapeutic target for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , MicroARNs , Animales , Células Endoteliales/metabolismo , Proteínas Hedgehog/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
10.
Brain Res ; 1746: 147000, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32579949

RESUMEN

Traumatic brain injury (TBI) is a major cause of death and disability in young adults worldwide. TBI-induced long-term cognitive deficits represent a growing clinical problem. Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) are involved in neuroprotection and neuronal plasticity. However, the knowledge concerning reparative efficacy of SCF + G-CSF treatment in post-acute TBI recovery remains incomplete. This study aims to determine the efficacy of SCF + G-CSF on post-acute TBI recovery in young adult mice. The controlled cortical impact model of TBI was used for inducing a severe damage in the motor cortex of the right hemisphere in 8-week-old male C57BL mice. SCF + G-CSF treatment was initiated 3 weeks after induction of TBI. Severe TBI led to persistent motor functional deficits (Rota-Rod test) and impaired spatial learning function (water maze test). SCF + G-CSF treatment significantly improved the severe TBI-impaired spatial learning function 6 weeks after treatment. TBI also caused significant increases of Fluoro-Jade C positive degenerating neurons in bilateral frontal cortex, striatum and hippocampus, and significant reductions in MAP2+ apical dendrites and overgrowth of SMI312+ axons in peri-TBI cavity frontal cortex and in the ipsilateral hippocampal CA1 at 24 weeks post-TBI. SCF + G-CSF treatment significantly reduced TBI-induced neurodegeneration in the contralateral frontal cortex and hippocampal CA1, increased MAP2+ apical dendrites in the peri-TBI cavity frontal cortex, and prevented TBI-induced axonal overgrowth in both the peri-TBI cavity frontal cortex and ipsilateral hippocampal CA1.These findings reveal a novel pathology of axonal overgrowth after severe TBI and demonstrate a therapeutic potential of SCF + G-CSF in ameliorating severe TBI-induced long-term neuronal pathology, neurostructural network malformation, and impairments in spatial learning.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Encéfalo/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Degeneración Nerviosa/patología , Factor de Células Madre/farmacología , Animales , Encéfalo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Recuperación de la Función/efectos de los fármacos , Aprendizaje Espacial/efectos de los fármacos
11.
Transl Stroke Res ; 11(4): 720-733, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31898187

RESUMEN

The homeostasis of the neurovascular unit (NVU) is disrupted after traumatic brain injury (TBI), and therapeutic strategies targeting the NVU would likely improve neurological outcomes after TBI. Sonic Hedgehog (Shh), which is an endogenous activator of the Hedgehog pathway, promotes brain repair in various injuries. In this study, the controlled cortical impact (CCI) was used to establish a moderate TBI model in adult male Sprague-Dawley rats (250-300 g), and the NVU was reconstructed in vitro from the blood-brain barrier (BBB) and neurons to investigate the effects of exogenous Shh protein on TBI. The modified neurological severity scores (mNSS) and Morris water maze tests were used to evaluate the effect of Shh on neurological function after TBI. The effect of Shh on the NVU in vivo was evaluated by detecting the degrees of cerebral edema and neuronal apoptosis. The integrity and permeability of the BBB, the level of inflammatory factors, and the expression of apoptotic proteins were detected to explore the effect of exogenous Shh on the NVU in vitro. The results showed that exogenous Shh reduced cerebral edema and neuronal apoptosis and promoted neurological recovery after TBI in rats. In vitro experiments showed that Shh-induced activation of the Hedgehog pathway promoted stability of the NVU by reducing damage to the tight junction structure and inhibiting the release of inflammatory factors and neuron apoptosis. Based on these results, the Shh-induced activation of the Hedgehog pathway might be a new promising treatment for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Proteínas Hedgehog/administración & dosificación , Proteínas Hedgehog/metabolismo , Acoplamiento Neurovascular , Recuperación de la Función , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/prevención & control , Lesiones Traumáticas del Encéfalo/prevención & control , Células Cultivadas , Modelos Animales de Enfermedad , Encefalitis/prevención & control , Masculino , Acoplamiento Neurovascular/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
12.
J Neurochem ; 153(6): 710-726, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31951014

RESUMEN

The level of microRNA-9-5p (miRNA-9-5p) in brain tissues is significantly changed after traumatic brain injury (TBI). However, the effect of miRNA-9-5p for brain function in TBI has not been elucidated. In this study, a controlled cortical impact model was used to induce TBI in Sprague-Dawley rats, and an oxygen glucose deprivation model was used to mimic the pathological state in vitro. Brain microvascular endothelial cells (BMECs) and astrocytes were extracted from immature Sprague-Dawley rats and cocultured to reconstruct blood-brain barrier (BBB) in vitro. The results show that the level of miRNA-9-5p was significantly increased in brain tissues after TBI, and up-regulation of miRNA9-5p contributed to the recovery of neurological function. Up-regulation of miRNA-9-5p with miRNA agomir may significantly alleviate apoptosis, neuroinflammation, and BBB damage in rats after TBI. Moreover, a dual luciferase reporter assay confirmed that miRNA-9-5p is a post-transcriptional modulator of Ptch-1. In in vitro experiments, the results confirmed that up-regulation of miRNA-9-5p with miRNA mimic alleviates cellular apoptosis, inflammatory response, and BBB damage mainly by inhibiting Ptch-1. In addition, we found that the activation of Hedgehog pathway was accompanied by inhibition of NF-κB/MMP-9 pathway in the BMECs treated with miRNA-9-5p mimic. Taken together, these results indicate that up-regulation of miRNA-9-5p alleviates BBB damage and neuroinflammatory responses by activating the Hedgehog pathway and inhibiting NF-κB/MMP-9 pathway, which promotes the recovery of neurological function after TBI.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Mediadores de Inflamación/metabolismo , MicroARNs/biosíntesis , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Proteínas Hedgehog/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Masculino , MicroARNs/administración & dosificación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
13.
Cell Cycle ; 18(11): 1241-1253, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31081721

RESUMEN

Researches have been focusing on the role of Slit2 in angiogenesis, specifically in cell migration and vessel permeability. Nevertheless, the role of Slit2-N, the bioactive fragment of Slit2, in the proliferation of vascular endothelia in choroidal neovascularization and some related mechanisms have not been studied yet. Thus, our study aimed to explore the role of Slit2-N in proliferation of vascular endothelia and the related mechanisms in choroidal neovascularization. Fluorescein isothiocyanate perfusion and HE staining were performed to evaluate volumes of choroidal neovascularization lesions. The effect of Slit2-N on VEGF165-induced cell proliferation and some related mechanisms were detected by CCK8 assay, flow cytometry, siRNA transfection, and western blotting. We found that Slit2-N reduced volumes of laser-induced choroidal neovascularization networks in vivo. Results of the in vitro study showed Slit2-N reduced VEGF165-induced cell proliferation of both human umbilical vascular endothelial cells and human microvascular endothelial cells possibly via activation of AKT rather than that of ERK1/2. Additionally, Robo4, one of the receptors binding to Slit2-N, was involved in the inhibitory effect of Slit2-N. Generally, our findings revealed the inhibitory role of Slit2-N in proliferation of vascular endothelia and some related mechanisms, and presented some potential targets, molecules along Slit2-N-Robo4-AKT axis, to choroidal neovascularization therapy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Neovascularización Coroidal/patología , Endotelio Vascular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas del Tejido Nervioso/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Células Cultivadas , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/fisiología , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Masculino , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/fisiología , Proteínas del Tejido Nervioso/uso terapéutico , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Endogámicas BN , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/química
14.
Cell Transplant ; 28(9-10): 1161-1172, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31010302

RESUMEN

Vascular smooth muscle cells (VSMCs) play an important role after a subarachnoid hemorrhage (SAH). The changes in VSMCs following bexarotene treatment after SAH are unknown. In the present study, neurological impairment, decreased cerebral cortical blood flow and transformation of cerebral VSMCs from a contractile to a synthetic phenotype were observed after SAH. Bexarotene reduced neurological impairment, improved cerebral cortical blood flow, inhibited VSMC phenotypic transformation and suppressed the expression of 5-lipoxygenase-activating protein (FLAP) and leukotriene B4 (LTB4), which was partly reversed by GW9662, an inhibitor of peroxisome proliferator-activated receptor gamma (PPARγ). Mechanistically, sh-PPARγ-mediated phenotypic transformation of VSMCs was partially suppressed by MK886, an antagonist of FLAP. Therefore, we conclude that bexarotene reduced neurological impairment, improved cerebral cortical blood flow and inhibited the VSMC phenotypic transformation after SAH, which was achieved by activating PPARγ-mediated inhibition of FLAP/LTB4 in VSMCs.


Asunto(s)
Proteínas Activadoras de la 5-Lipooxigenasa/metabolismo , Bexaroteno/farmacología , Corteza Cerebral/metabolismo , Leucotrieno B4/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , PPAR gamma/metabolismo , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/patología , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , PPAR gamma/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/patología
15.
Neuroscience ; 408: 105-114, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30910642

RESUMEN

Lipoxin A4 (LXA4) has been reported to reduce inflammation in experimental subarachnoid hemorrhage (SAH), but the mechanism remains unclear. In this study, we investigated the role of LXA4 in inflammation-mediated cerebrovascular endothelial dysfunction and the potential mechanism after SAH. SAH was induced by endovascular perforation in male Sprague-Dawley rats, and recombinant LXA4 was injected intracerebroventricularly 1.5 h after the operation. The expression changes in the markers of endothelial dysfunction (endothelial microparticles and nitric oxide) were analyzed by flow cytometry or Nitric Oxide (NO) assay kit. Microflow in the cerebral cortex was assayed by laser speckle contrast imaging. Neutrophil infiltration was observed by a marker of leukocyte activity (myeloperoxidase, MPO) that colocalized with a specific marker of endothelial cells (von Willebrand factor, VWF). The expression of LXA4 and its downstream molecules, formyl peptide receptor 2 (FPR2), extracellular signal-regulated kinase (ERK1/2), nuclear factor-κB (NF-κB), matrix metalloproteinase-9 (MMP9), and the levels of pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6), leukocyte adhesion molecule (intercellular adhesion molecule-1, ICAM-1) and MPO were measured either by Western blot or enzyme-linked immunosorbent assay (ELISA). SAH resulted in endothelial dysfunction and a reduction in microflow in the cerebral cortex. The expression of LXA4 was decreased, and the expression of pro-inflammatory factors (NF-κB, MMP9, ICAM-1, MPO) and cytokines (TNF-α, IL-1ß, IL-6) was increased after SAH. The administration of LXA4 significantly ameliorated endothelial dysfunction, recovered microflow, and suppressed the inflammation and infiltration of neutrophils in SAH rats. The underlying mechanism of this outcome may involve the LXA4/FPR2/ERK1/2 pathway. LXA4 might be a promising candidate for acute SAH treatment.


Asunto(s)
Edema Encefálico/metabolismo , Corteza Cerebral/metabolismo , Endotelio Vascular/metabolismo , Lipoxinas/metabolismo , Hemorragia Subaracnoidea/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Edema Encefálico/tratamiento farmacológico , Corteza Cerebral/efectos de los fármacos , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipoxinas/farmacología , Lipoxinas/uso terapéutico , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/tratamiento farmacológico
16.
Front Immunol ; 9: 1713, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30090104

RESUMEN

Uveitis is characterized as a common cause of blindness worldwide. Aryl hydrocarbon receptor (AhR), a ligand-activated nuclear receptor, has been implicated to play a role in human uveitis, although the exact mechanisms remain poorly understood. The purpose of this study was to enhance our knowledge concerning the role of AhR during intraocular inflammation. We immunized wild-type and AhR-knockout C57BL/6J mice with IRBP651-670 to induce experimental autoimmune uveitis (EAU). Disease severity was evaluated with both clinical and histopathological grading. Blood-retinal barrier (BRB) integrity was tested by Evans blue and tight junction proteins qualifications. Apoptosis was measured using TdT-mediated dUTP nick end labeling staining. Macrophage/microglia activation and polarization were studied by immunofluorescence and Western blot. Following EAU induction, AhR-/- mice had more severe clinical and histopathological manifestations of uveitis than AhR+/+ mice. Increased vascular permeability and apoptotic cells were observed in AhR-/- EAU mice when compared with AhR+/+ EAU mice. In addition, AhR-/- EAU mice showed evidence of a significantly increased macrophage/microglia cells and a stronger polarization from the M2 to the M1 phenotype as compared to AhR+/+ EAU mice. The levels of pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1ß were increased in AhR-/- EAU mice, which was associated with the activation of NF-κB and signal transducers and activators of transcription (STAT) pathways. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an agonist of AhR, caused a significant decrease in the clinical and histopathological manifestations, preserved BRB integrity, reduced apoptotic cells, inhibited macrophage/microglia activation, and shifted their polarization from M1 toward M2. Moreover, decreased expression of pro-inflammatory cytokines including TNF-α, IL-6, and IL-1ß and inhibition of NF-κB and STAT pathways were found in EAU mice following TCDD treatment. In conclusion, AhR activation with TCDD exhibits an immunomodulatory effect by reducing BRB breakdown, inhibiting retinal cell apoptosis, and reducing pro-inflammatory cytokine expression during EAU. The underlying mechanism may involve the modulation of macrophages/microglia polarization and the downregulation of NF-κB and STAT pathways.

17.
Brain Res ; 1700: 118-125, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29990491

RESUMEN

The role of the cerebrovascular network during the acute and chronic phases after traumatic brain injury (TBI) is poorly defined and emerging evidence suggests that cerebral perfusion is altered. The purpose of this study is to explore how the cortical blood flow is pathologically altered following TBI using a newly developed technique, laser speckle imaging. The controlled cortical impact (CCI) model was established in mice. Then, cerebral blood flow was monitored in vivo laser speckle imaging and vessel painting was labeled by Lectin in the peri-contusional cortex. Lastly, mice were assessed for lesion size and neurological functions. Our results indicated that: 1) In the acute phase of TBI, cerebral blood flow and microvessel counts decreased significantly (P < 0.05) 2) In the chronic phase of TBI, cerebral blood flow and microvessel counts recovered gradually (P < 0.05) 3) Cortical lesion volume reduced significantly in the chronic phase of TBI (P < 0.05) 4) Spontaneous neurocognitive recovery occurred following CCI in mice (P < 0.05). In the acute phase of TBI, there is a reduction in cerebral perfusion at the lesion site. However, this reduction recovers in the chronic phase of TBI ultimately, followed by an improvement of ameliorated neurobehavioral functions and a decrease in the lesion size. The novel approach for cerebral blood flow monitoring by laser speckle imaging can be extended from bench to bedside and provide potential therapeutic strategies for TBI patients.


Asunto(s)
Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Corteza Cerebral/diagnóstico por imagen , Circulación Cerebrovascular , Microvasos/diagnóstico por imagen , Animales , Lesiones Traumáticas del Encéfalo/fisiopatología , Corteza Cerebral/fisiopatología , Circulación Cerebrovascular/fisiología , Cognición/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neuroimagen Funcional , Rayos Láser , Masculino , Aprendizaje por Laberinto/fisiología , Ratones Endogámicos C57BL , Microvasos/fisiopatología , Destreza Motora/fisiología , Distribución Aleatoria , Recuperación de la Función/fisiología
18.
Int J Med Sci ; 15(10): 1072-1082, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30013449

RESUMEN

Background: As a major antioxidant in serum, uric acid (UA) was once considered only as the leading cause of gout; however, recent studies have validated its neuroprotective role in ischemic stroke. Because the potential protective effects of UA in traumatic brain injury (TBI) remain largely unknown, this study investigated the role of UA in TBI in both clinical patients and experimental animals. Methods: In TBI patients, serum UA concentrations were measured within 3 days after injury. Clinical outcomes at discharge were classified according to the Glasgow Outcome Scale: good outcome (4-5) and poor outcome (1-3). Risk factors for good outcome were identified via backward logistic regression analysis. For the animal study, a controlled cortical impact (CCI) injury model was established in mice. These mice were given UA at different doses intraperitoneally, and subsequent UA concentrations in mouse serum and brain tissue were determined. Neurological function, oxidative stress, inflammatory response, neuronal maintenance, cerebral blood flow, and lesion size were also assessed. Results: The serum UA level was significantly lower in TBI patients who had a good outcome (P<0.01), and low serum UA was an independent predictor of good outcome after TBI (P<0.01; odds ratio, 0.023; 95% confidence interval, 0.006-0.082). Consistently, decreased levels of serum UA were observed in both TBI patients and CCI animals (P<0.05), whereas the UA concentration was increased in CCI brain tissue (P<0.05). Administration of UA further increased the UA level in brain tissue as compared to that in control animals (P<0.05). Among the different doses administered, 16 mg/kg UA improved sensorimotor functional recovery, spatial learning, and memory in CCI mice (P<0.05). Moreover, oxidative stress and the inflammatory response were inhibited by UA treatment (P<0.05). UA treatment also improved neuronal maintenance and cortical blood flow (P<0.05) but not lesion size (P>0.05). Conclusions: UA acted to attenuate neuronal loss, cerebral perfusion impairment and neurological deficits in TBI mice through suppression of neuronal and vascular oxidative stress. Following TBI, active antioxidant defense in the brain may result in consumption of UA in the serum, and thus, a decreased serum UA level could be predictive of good clinical recovery.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Ácido Úrico , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Ácido Úrico/sangre , Ácido Úrico/orina
19.
Neurobiol Dis ; 117: 114-124, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29886067

RESUMEN

Traumatic brain injury (TBI) causes a high rate of mortality and disability worldwide, and there exists almost none effective drugs to protect against TBI. Neurotoxicity occurring after TBI can be derived from microglia and astrocytes, and causes neuronal death and synapse loss. Bexarotene has been demonstrated to protect neurons in CNS diseases. In the present study, we aimed to investigate the potential role of bexarotene in protecting against neurotoxicity after TBI, as well as the underlying mechanism. The controlled cortical impact (CCI) model was established on adult C57BL/6 mice, followed by intraperitoneal administration of bexarotene for 14 consecutive days. We found that bexarotene improved sensorimotor function and cognitive recovery in CCI mice. In addition, bexarotene decreased neuronal death and synapse loss, as well as inhibited apoptotic cascade. Moreover, bexarotene treatment reduced M1 microglia polarization, microglia-derived pro-inflammatory cytokines, and the number of A1 astrocytes after CCI. These effects of bexarotene were partially abolished by T0070907, an antagonist of peroxisome proliferator-activated receptor gamma (PPARγ). Additionally, bexarotene enhanced nuclear translocation and transcriptional activity of PPARγ. These findings show that bexarotene inhibits neurotoxicity in mice after TBI, at least in part through a PPARγ-dependent mechanism.


Asunto(s)
Bexaroteno/uso terapéutico , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/prevención & control , Fármacos Neuroprotectores/uso terapéutico , PPAR gamma/metabolismo , Animales , Benzamidas/toxicidad , Bexaroteno/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Piridinas/toxicidad
20.
Brain Behav Immun ; 65: 183-194, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28483659

RESUMEN

OBJECTIVE: Bexarotene treatments exert neuroprotective effects on mice following traumatic brain injury (TBI). The present study aims to investigate the potential roles of the long noncoding RNA Neat1 in the neuroprotective effects of bexarotene. MATERIALS AND METHODS: Adult male C57BL/6J mice (n=80) and newborn mice (within 24h after birth) (n=20) were used to generate a "controlled cortical impact" (CCI) model and harvest primary cortex neurons, respectively. The HT22 cell line and the BV2 cell line were cultured under "normal" or "oxygen/glucose-deprived" (OGD) conditions. The relationship between RXR-α and the Neat1 promoter was clarified using ChIP-qPCR and dual-luciferase reporter gene assays. The mRNA alterations induced by Neat1 knockdown were measured using next-generation RNA sequencing. Proteins were captured by Neat1, pulled down and subjected to mass spectrometry. The neurological severity score, rotarod test and water maze test were employed to measure the animals' motor and cognitive functions. RESULTS: Bexarotene prominently up-regulated the Neat1 level in an RXR-α-dependent manner. Neat1 knockdown induced significant changes in mRNA expression, and the altered mRNAs were involved in many biological processes, including synapse formation and axon guidance. In primary neurons, Neat1 knockdown inhibited and Neat1 over-expression prompted axon elongation. Multiple proteins, including Pidd1, were captured by Neat1. Neat1 inhibited cell apoptosis and restricted inflammation by capturing Pidd1. The in vitro anti-apoptotic and anti-inflammatory effects of Neat1 were further confirmed in C57BL/6 mice, which resulted in better motor and cognitive function after TBI. CONCLUSION: Bexarotene up-regulates the lncRNA Neat1, which inhibits apoptosis and inflammation, thereby resulting in better functional recovery in mice after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , ARN Largo no Codificante/metabolismo , ARN Largo no Codificante/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Bexaroteno , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , ARN Largo no Codificante/genética , Tetrahidronaftalenos/farmacología , Tetrahidronaftalenos/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...