Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Nanomedicine ; 19: 4377-4409, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38774029

RESUMEN

Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.


Asunto(s)
Accidente Cerebrovascular Isquémico , Nanopartículas , Neovascularización Fisiológica , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Animales , Nanopartículas/química , Neovascularización Fisiológica/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Liposomas/química , Sistemas de Liberación de Medicamentos/métodos , Sistema de Administración de Fármacos con Nanopartículas/química , Angiogénesis
2.
Int Immunopharmacol ; 126: 111183, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37984250

RESUMEN

Once an ischemic stroke occurs, reactive oxygen species (ROS) and oxidative stress degrade the tight connections between cerebral endothelial cells resulting in their damage. The expression of antioxidant genes may be enhanced, and ROS formation may be reduced following Nrf2 activation, which is associated with protection against ischemic stroke. Overexpression of spermine oxidase (Smox) in the neocortex led to increased H2O2 production. However, how Smox impacts the regulation of the blood-brain barrier (BBB) through antioxidants has not been examined yet. We conducted experiments both in the cell level and in the transient middle cerebral artery occlusion (tMCAO) model to evaluate the effect of Smox siRNA lentivirus (si-Smox) knockdown on BBB protection against ischemic stroke. Mice treated with si-Smox showed remarkably decreased BBB breakdown and reduced endothelial inflammation following stroke. The treatment with si-Smox significantly elevated the Bcl-2 to Bax ratio and decreased the production of cleaved caspase-3 in the tMCAO model. Further investigation revealed that the neuroprotective effect was the result of the antioxidant properties of si-Smox, which reduced oxidative stress and enhanced CD31+ cells in the peri-infarct cortical areas. Of significance, si-Smox activated Nrf2 in both bEnd.3 cells and tMCAO animals, and blocking Nrf2 with brusatol diminished the protective effects of si-Smox. The study findings suggest that si-Smox exerts neuroprotective effects and promotes angiogenesis by activating the Nrf2 pathway, thus decreasing oxidative stress and apoptosis caused by tMCAO. As a result, si-Smox may hold potential as a therapeutic candidate for preserving BBB integrity while treating ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Fármacos Neuroprotectores , Accidente Cerebrovascular , Animales , Ratones , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Antioxidantes/metabolismo , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Peróxido de Hidrógeno/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/genética , Accidente Cerebrovascular Isquémico/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo
3.
Mol Neurobiol ; 60(7): 3741-3757, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36940077

RESUMEN

Excessive activation of aldose reductase (AR) in the brain is a risk factor for aggravating cerebral ischemia injury. Epalrestat is the only AR inhibitor with proven safety and efficacy, which is used in the clinical treatment of diabetic neuropathy. However, the molecular mechanisms underlying the neuroprotection of epalrestat remain unknown in the ischemic brain. Recent studies have found that blood-brain barrier (BBB) damage was mainly caused by increased apoptosis and autophagy of brain microvascular endothelial cells (BMVECs) and decreased expression of tight junction proteins. Thus, we hypothesized that the protective effect of epalrestat is mainly related to regulating the survival of BMVECs and tight junction protein levels after cerebral ischemia. To test this hypothesis, a mouse model of cerebral ischemia was established by permanent middle cerebral artery ligation (pMCAL), and the mice were treated with epalrestat or saline as a control. Epalrestat reduced the ischemic volume, enhanced BBB function, and improved the neurobehavior after cerebral ischemia. In vitro studies revealed that epalrestat increased the expression of tight junction proteins, and reduced the levels of cleaved-caspase3 and LC3 proteins in mouse BMVECs (bEnd.3 cells) exposed to oxygen-glucose deprivation (OGD). In addition, bicalutamide (an AKT inhibitor) and rapamycin (an mTOR inhibitor) increased the epalrestat-induced reduction in apoptosis and autophagy related protein levels in bEnd.3 cells with OGD treatment. Our findings suggest that epalrestat improves BBB function, which may be accomplished by reducing AR activation, promoting tight junction proteins expression, and upregulating AKT/mTOR signaling pathway to inhibit apoptosis and autophagy in BMVECs.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Ratones , Animales , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Aldehído Reductasa/metabolismo , Aldehído Reductasa/farmacología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Infarto Cerebral/metabolismo , Lesiones Encefálicas/metabolismo , Glucosa/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
4.
Int Immunopharmacol ; 97: 107811, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34091117

RESUMEN

Multiple sclerosis (MS) is a neurodegenerative and demyelinating autoimmune disease mediated by autoreactive T cells that affects the central nervous system (CNS). Electroacupuncture (EA) has emerged as an alternative or supplemental treatment for MS, but the mechanism by which EA may alleviate MS symptoms is unresolved. Here, we examined the effects of EA at the Zusanli (ST36) acupoint on mice with experimental autoimmune encephalomyelitis (EAE), the predominant animal model of MS. The effects of EA on EAE emergence, inflammatory cell levels, proinflammatory cytokines, and spinal cord pathology were examined. EA treatment attenuated the EAE clinical score and associated spinal cord demyelination, while reducing the presence of proinflammatory cytokines in mononuclear cells (MNCs), downregulating microRNA (miR)-155, and upregulating the opioid peptide precursor proopiomelanocortin (POMC) in the CNS. Experiments in which cultured neurons were transfected with a miR-155 mimic or a miR-155 inhibitor further showed that the direct modulation of miR-155 levels could regulate POMC levels in neurons. In conclusion, the alleviation of EAE by EA is characterized by reduced proportions of Th1/Th17 cells and increased proportions of Th2 cells, POMC upregulation, and miR-155 downregulation, while miR-155 itself can suppress POMC expression. These results, support the hypothesis that the effects of EA on EAE may involve the downregulation of miR-155.


Asunto(s)
Electroacupuntura , Encefalomielitis Autoinmune Experimental/terapia , MicroARNs/metabolismo , Esclerosis Múltiple/terapia , Animales , Regulación hacia Abajo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Humanos , Ratones , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Esclerosis Múltiple/inmunología , Proopiomelanocortina/genética , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología , Regulación hacia Arriba/inmunología
5.
Neurochem Int ; 141: 104860, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33022312

RESUMEN

Cerebral ischemia-reperfusion injury induces a secondary immune inflammatory reaction that exacerbates brain injury and clinical prognosis. Dendritic cells (DCs) and microglia are both important regulators of neuroinflammation. Studies have confirmed that a large number of cells express the DC surface marker CD11c in the ischemic area, and some of these cells also express microglial markers. However, the specific mechanism of transformation between microglia and DCs and their roles in the process of cerebral ischemia-reperfusion injury are still not clear. In this study, we established a mouse model and flow cytometry was used to detect the expression of mature DC surface molecules in activated microglia. IFN-γ knockout mice were used to determine the regulatory effect of IFN-γ on microglial transformation. We found that CD11c+ cells were derived from microglia after ischemia-reperfusion injury, and this group of cells highly expressed MHC-II molecules and other costimulatory molecules, such as CD80 and CD86, which were regulated by IFN-γ and its downstream signaling molecules ERK/c-myc. In summary, our results showed in cerebral ischemia-reperfusion injury, IFN-γ regulates the transformation of microglia to DC-like cells. Microglial-derived DC-like cells possess the ability to present antigens and activate naïve T cells which is regulated by the ERK/c-myc signaling pathway.


Asunto(s)
Dendritas/efectos de los fármacos , Interferón gamma/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/efectos de los fármacos , Daño por Reperfusión/patología , Animales , Antígenos CD11/metabolismo , Dendritas/patología , Genes MHC Clase II , Interferón gamma/fisiología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Cultivo Primario de Células , Receptores de Interferón/biosíntesis , Linfocitos T
6.
FASEB J ; 34(10): 13762-13775, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32808351

RESUMEN

Cerebral ischemia causes damage to the structure and function of the blood-brain barrier (BBB) and alleviating BBB destruction will be of great significance for the treatment and prognosis of ischemic stroke. Recently, microRNAs have been shown to play a critical role in BBB integrity. However, the potential mechanism by which microRNA-182 (miR-182) affects the BBB in ischemic stroke remains unclear. We demonstrated for the first time that cerebral ischemia leads to a significant progressive increase in miR-182 after pMCAO, and bEnd.3 cells are the primary target cells of miR-182. In miR-182 KD transgenic mice, infarct volume, and BBB permeability were attenuated, and tight junction (TJ) proteins increased. Inhibition of miR-182 with an antagomir reduced OGD-induced apoptosis of bEnd.3 cells and the loss of ZO-1 and Occludin. To further explore the mechanism by which miR-182 regulates BBB integrity, we detected the apoptotic proteins Bcl-2/Bax and demonstrated that mTOR and FOXO1 were the targets of miR-182. Inhibition of mTOR/FOXO1 by rapamycin/AS1842856 decreased the ratio of Bcl-2/Bax and exacerbated TJ protein loss. Taken together, inhibition of miR-182 protects BBB integrity by reducing endothelial cell apoptosis through the mTOR/FOXO1 pathway. Thus, miR-182 may be a potential target for the treatment of BBB disruption during cerebral ischemia.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Animales , Apoptosis , Línea Celular , Células Cultivadas , Regulación hacia Abajo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Infarto de la Arteria Cerebral Media/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo
7.
Int Immunopharmacol ; 81: 106176, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32044667

RESUMEN

A high salt diet (HSD) is among the most important risk factors for many diseases. One mechanism by which HSD aggravates cerebral ischemic injury is independent of blood pressure changes. The direct role of HSD in inflammation after cerebral ischemia is unclear. In this research, after twenty-one days of being fed a high salt diet, permanent focal ischemia was induced in mice via operation. At 12 h and 1, 3 and 5 days postischemia, the effects of HSD on the lesion volume, microglia polarization, aldose reductase (AR) expression, and inflammatory processes were analyzed. We report that in mice, surplus dietary salt promotes inflammation and increases the activation of classical lipopolysaccharide (LPS)-induced microglia/macrophages (M1). This effect depends on the expression of the AR protein in activated microglia after permanent middle cerebral artery ligation (pMCAL) in HSD mice. The administration of either the AR inhibitor Epalrestat or a p38-neutralizing antibody blocked the polarization of microglia and alleviated stroke injury. In conclusion, HSD promotes polarization in pro-inflammatory M1 microglia by upregulating the expression of the AR protein via p38/MAPK, thereby exacerbating the development of ischemia stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Macrófagos/fisiología , Microglía/fisiología , Sales (Química)/administración & dosificación , Antagonistas de Receptores Androgénicos/administración & dosificación , Animales , Isquemia Encefálica/patología , Diferenciación Celular , Citocinas/metabolismo , Ingestión de Alimentos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Androgénicos/metabolismo , Rodanina/administración & dosificación , Rodanina/análogos & derivados , Sales (Química)/efectos adversos , Transducción de Señal , Células TH1/inmunología , Tiazolidinas/administración & dosificación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Int Immunopharmacol ; 75: 105822, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31437793

RESUMEN

Metformin, the most widely used medicine for type 2 diabetes, displays anti-inflammatory functions via activating AMP-activated protein kinase (AMPK). Circulating autoantibodies and disequilibrium of helper T cells and regulatory T cells are pathological hallmarks of myasthenia gravis (MG). Rectify the imbalance of different T cell populations has become an important therapeutic strategy to treat MG. In this study, we assessed the effect of metformin on the development of autoimmunity using an experimental autoimmune myasthenia gravis (EAMG) rat model. We first provided evidence that oral administration of metformin attenuated the onset of EAMG. This effect was accompanied by a substantial decrease of circulating auto-antibody levels with no effect on blood glucose level. While metformin treatment in vitro showed little effect on inducible Treg, metformin strongly inhibited Th17 cell differentiation through the increase of reactive oxygen species and AMPK. Furthermore, an attenuation of antigen-induced IgG2b antibody production by two different doses of metformin was also observed in the AChR-specific recall response. In conclusion, the above results indicate that metformin may have therapeutic value for the clinical treatment of MG.


Asunto(s)
Antiinflamatorios/uso terapéutico , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/inmunología , Animales , Antiinflamatorios/farmacología , Anticuerpos/sangre , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Glucemia/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Hipoglucemiantes/farmacología , Metformina/farmacología , Ratas Endogámicas Lew , Especies Reactivas de Oxígeno/inmunología , Receptores Colinérgicos/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología
9.
Neurosci Bull ; 35(3): 507-518, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30796753

RESUMEN

Myasthenia gravis (MG) is a prototypical antibody-mediated neurological autoimmune disease with the involvement of humoral immune responses in its pathogenesis. T follicular helper (Tfh) cells have been implicated in many autoimmune diseases. However, whether and how Tfh cells are involved in MG remain unclear. Here, we established and studied a widely-used and approved animal model of human MG, the rat model with acetylcholine receptor alpha (AChRα) subunit (R-AChR97-116)-induced experimental autoimmune myasthenia gravis (EAMG). This model presented mild body-weight loss 10 days after the first immunization (representing the early stage of disease) and more obvious clinical manifestations and body-weight loss 7 days after the second immunization (representing the late stage of disease). AChR-specific pre-Tfh cells and mature Tfh cells were detected in these two stages, respectively. In co-cultures of Tfh cells and B cells, the number of IgG2b-secreting B cells and the level of anti-AChR antibodies in the supernatant were higher in the cultures containing EAMG-derived Tfh cells. In immunohistochemistry and immunofluorescence assays, a substantial number of CD4+/Bcl-6+ T cells and a greater number of larger germinal centers were observed in lymph node tissues resected from EAMG rats. Based on these results, we hypothesize that an AChR-specific Tfh cell-mediated humoral immune response contributes to the development of EAMG.


Asunto(s)
Inmunidad Humoral , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Colinérgicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Femenino , Ganglios Linfáticos/inmunología , Subunidades de Proteína/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Ratas Endogámicas Lew , Receptor Cross-Talk
10.
Mol Cell Neurosci ; 83: 65-73, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28684360

RESUMEN

Regulator of G protein signaling 5 (RGS5) acts as a GTPase-activating protein (GAP) for the Gαi subunit and negatively regulates G protein-coupled receptor signaling. However, its presence and function in postmitotic differentiated primary neurons remains largely uncharacterized. During neural development, sonic hedgehog (Shh) signaling is involved in cell signaling pathways via Gαi activity. In particular, Shh signaling is essential for embryonic neural tube patterning, which has been implicated in neuronal polarization involving neurite outgrowth. Here, we examined whether RGS5 regulates Shh signaling in neurons. RGS5 transcripts were found to be expressed in cortical neurons and their expression gradually declined in a time-dependent manner in culture system. When an adenovirus expressing RGS5 was introduced into an in vitro cell culture model of cortical neurons, RGS5 overexpression significantly reduced neurite outgrowth and FM4-64 uptake, while cAMP-PKA signaling was also affected. These findings suggest that RGS5 inhibits Shh function during neurite outgrowth and the presynaptic terminals of primary cortical neurons mature via modulation of cAMP.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neuronas/metabolismo , Proteínas RGS/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Corteza Cerebral/citología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones , Ratones Endogámicos C57BL , Proyección Neuronal , Neuronas/citología , Proteínas RGS/genética
11.
Atherosclerosis ; 251: 282-290, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27439214

RESUMEN

BACKGROUND AND AIMS: Atherosclerosis is a chronic inflammatory vascular disease related to macrophages uptake of low-density lipoprotein and their subsequent transformation into foam cells. M1 (inflammatory)/M2 (anti-inflammatory) balance was suggested to impact disease progression. In this study, we investigated whether the immunity related GTPase (Irgm1) regulates macrophage polarization during atherosclerosis development. METHODS: We used apolipoprotein E (ApoE) knockout and Irgm1 haplodeficient mice and induced atherosclerosis with high-cholesterol diet for the indicated months. Atherosclerotic arteries were collected from patients undergoing vascular surgery, to determine the lesional expression of Irgm1 and distribution of M1/M2 populations. RESULTS: Our results showed that IRGM/Irgm1 expression was increased in atherosclerotic artery samples (1.7-fold, p=0.0045) compared with non-atherosclerotic arteries, which was consistent with findings in the murine experimental atherosclerosis model (1.9-fold, p=0.0002). IRGM/Irgm1 expression was mostly found in lesional M1 macrophages. Haplodeficiency of Irgm1 in ApoE(-/-) mice resulted in reduced infiltrating M1 macrophages in atheroma (94%, p=0.0002) and delayed development of atherosclerotic plaques. In vitro experiments also confirmed that Irgm1 haplodeficiency reduced iNOS expression of polarized M1 macrophages (81%, p=0.0034), with negligible impact on the M2 phenotype. Moreover, we found that Irgm1 haplodeficiency in mice significantly reduced expression level of M1 function-related transcription factors, interferon regulatory factor (Irf) 5 and Irf8, but not Irf4, an M2-related transcription factor. CONCLUSIONS: This study shows that Irgm1/IRGM participates in the polarization of M1 macrophage and promotes development of atheroma in murine experimental atherosclerosis.


Asunto(s)
Aterosclerosis/sangre , Aterosclerosis/genética , Proteínas de Unión al GTP/fisiología , Macrófagos/citología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Polaridad Celular , Colesterol/metabolismo , Femenino , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/genética , Humanos , Factor 1 Regulador del Interferón/genética , Factores Reguladores del Interferón/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Persona de Mediana Edad , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo
12.
Sci Rep ; 5: 16548, 2015 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-26549644

RESUMEN

High salt diet (HSD) is one of the most important risk factors that contribute to many vascular diseases including ischemic stroke. One proposed mechanism underlying the disruption of blood-brain barrier (BBB) mediated by HSD is indirectly through enhancing blood pressure. The direct role of HSD on BBB integrity is unclear. Our purpose is to determine whether and how HSD might be involved in BBB breakdown during ischemia. To test that, we induced model of cerebral ischemia by permanent middle cerebral artery ligation (pMCAL) in either normal diet or HSD fed mice. We observed that HSD significantly enhanced ischemic brain damage which was associated with enhanced BBB disruption, increased leukocytes infiltration and loss of tight junction (TJ) proteins expression without apparently altering blood pressure. Our in vitro experiment also revealed that sodium chloride (NaCl) treatment down-regulated TJ protein expression by endothelial cells and substantially increased BBB permeability during starvation. Inhibition of p38/MAPK/SGK1 pathway eliminated the effect of NaCl on BBB permeability in vitro. In addition, we noticed a positive correlation between urinary sodium levels and ischemic lesion size in stroke patients. Together, our study demonstrates a hypertension-independent role of HSD during ischemia and provides rationale for post cerebral ischemic attack management.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Cloruro de Sodio Dietético/administración & dosificación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adulto , Anciano , Animales , Barrera Hematoencefálica/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Persona de Mediana Edad , Ocludina/genética , Ocludina/metabolismo , Permeabilidad , Sodio/orina , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
13.
Inflammation ; 38(6): 2067-75, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26063186

RESUMEN

We used samples from rheumatoid arthritis (RA) patients to examine whether Anti-citrullinated protein antibodies (ACPAs) alter macrophage subset distribution and promote RA development. Macrophage subset distributions and interferon regulatory factor 4 (IRF4) and IRF5 expressions were analyzed. ACPAs were purified by affinity column. After RA and osteoarthritis (OA) patients' macrophages were cocultured with ACPAs, macrophage subsets and IRF4 and IRF5 expressions were measured. Small interfering RNAs (siRNAs) were transfected into ACPA-activated cells to suppress IRF4 or IRF5. Fluorescence-activated cell sorting (FACS), Western blot, and immunohistochemistry were performed. Macrophage subset disequilibrium occurred in RA patient synovial fluids. IRF4 and IRF5 were all expressed in the synovial fluid and synovium. ACPAs (40 IU/ml) could induce macrophages to polarize to M1 subsets, and the percentage of increased M1/M2 ratio of RA patients was higher than that of the OA patients. ACPAs also induce IRF4 and IRF5 protein expressions. IRF5 siRNA transfection impaired ACPA activity significantly. We demonstrated that macrophage subset disequilibrium occurred in RA patients. ACPAs induced IRF5 activity and led to M1 macrophage polarization.


Asunto(s)
Artritis Reumatoide/inmunología , Autoanticuerpos/inmunología , Citrulina/inmunología , Macrófagos/inmunología , Péptidos/inmunología , Artritis Reumatoide/sangre , Artritis Reumatoide/genética , Autoanticuerpos/sangre , Estudios de Casos y Controles , Células Cultivadas , Femenino , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Factores Reguladores del Interferón/metabolismo , Macrófagos/metabolismo , Masculino , Fenotipo , Interferencia de ARN , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Transfección
14.
J Neuroinflammation ; 11: 79, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24742325

RESUMEN

BACKGROUND: Stroke is accompanied by a distinguished inflammatory reaction that is initiated by the infiltration of immunocytes, expression of cytokines, and other inflammatory mediators. As natural killer cells (NK cells) are a type of cytotoxic lymphocyte critical to the innate immune system, we investigated the mechanism of NK cells-induced brain injuries after cerebral ischemia and the chemotactic effect of IP-10 simultaneously. METHODS: NK cells infiltration, interferon-gamma (IFN-γ) and IP-10 expression were detected by immunohistochemistry, immunofluorescence, PCR and flow cytometry in human and C57/BL6 wild type mouse ischemic brain tissues. The ischemia area was detected via 2,3,5-triphenyltetrazolium chloride staining. CXCR3 mean fluorescence intensity of isolated NK cells was measured by flow cytometry. The neuronal injury made by NK cells was examined via apoptosis experiment. The chemotactic of IP-10 was detected by migration and permeability assays. RESULTS: In human ischemic brain tissue, infiltrations of NK cells were observed and reached a peak at 2 to 5 days. In a permanent middle cerebral artery occlusion (pMCAO) model, infiltration of NK cells into the ischemic infarct region reached their highest levels 12 hours after ischemia. IFN-γ-positive NK cells and levels of the chemokine IP-10 were also detected within the ischemic region, from 6 hours up to 4 days after pMCAO was performed, and IFN-γ levels decreased after NK cells depletion in vivo. Co-culture experiments of neural cells with NK cells also showed that neural necrosis was induced via IFN-γ. In parallel experiments with IP-10, the presence of CXCR3 indicates that NK cells were affected by IP-10 via CXCR3, and the effect was dose-dependent. After IP-10 depletion in vivo, NK cells decreased. In migration assays and permeability experiments, disintegration of the blood-brain barrier (BBB) was observed following the addition of NK cells. Moreover, in the presence of IP-10 this injury was aggravated. CONCLUSIONS: All findings support the hypothesis that NK cells participate in cerebral ischemia and promote neural cells necrosis via IFN-γ. Moreover, IP-10 intensifies injury to the BBB by NK cells via CXCR3.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Quimiocina CXCL10/metabolismo , Células Asesinas Naturales/fisiología , Animales , Animales Recién Nacidos , Antígenos Ly/metabolismo , Apoptosis/fisiología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/citología , Movimiento Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media/patología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Receptores CXCR3/metabolismo
15.
EMBO J ; 32(18): 2477-90, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-23942232

RESUMEN

In rheumatoid arthritis (RA), macrophage is one of the major sources of inflammatory mediators. Macrophages produce inflammatory cytokines through toll-like receptor (TLR)-mediated signalling during RA. Herein, we studied macrophages from the synovial fluid of RA patients and observed a significant increase in activation of inositol-requiring enzyme 1α (IRE1α), a primary unfolded protein response (UPR) transducer. Myeloid-specific deletion of the IRE1α gene protected mice from inflammatory arthritis, and treatment with the IRE1α-specific inhibitor 4U8C attenuated joint inflammation in mice. IRE1α was required for optimal production of pro-inflammatory cytokines as evidenced by impaired TLR-induced cytokine production in IRE1α-null macrophages and neutrophils. Further analyses demonstrated that tumour necrosis factor (TNF) receptor-associated factor 6 (TRAF6) plays a key role in TLR-mediated IRE1α activation by catalysing IRE1α ubiquitination and blocking the recruitment of protein phosphatase 2A (PP2A), a phosphatase that inhibits IRE1α phosphorylation. In summary, we discovered a novel regulatory axis through TRAF6-mediated IRE1α ubiquitination in regulating TLR-induced IRE1α activation in pro-inflammatory cytokine production, and demonstrated that IRE1α is a potential therapeutic target for inflammatory arthritis.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Citocinas/metabolismo , Endorribonucleasas/metabolismo , Activación Enzimática/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Toll-Like/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Western Blotting , Línea Celular , Sistemas de Liberación de Medicamentos , Endorribonucleasas/antagonistas & inhibidores , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Inmunoprecipitación , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Reacción en Cadena en Tiempo Real de la Polimerasa , Líquido Sinovial/citología , Factor 6 Asociado a Receptor de TNF/farmacología
16.
Immunology ; 138(4): 333-45, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23181351

RESUMEN

Experimental allergic encephalomyelitis (EAE) can be induced in animal models by injecting the MOG35-55 peptide subcutaneously. Dendritic cells (DCs) that are located at the immunization site phagocytose the MOG35-55 peptide. These DCs mature and migrate into the nearest draining lymph nodes (dLNs), then present antigen, resulting in the activation of naive T cells. T helper type 1 (Th1) and Th17 cells are the primary cells involved in EAE progression. All-trans-retinoic acid (AT-RA) has been shown to have beneficial effects on EAE progression; however, whether AT-RA influences DC maturation or mediates other functions is unclear. In the present study, we showed that AT-RA led to the down-regulation of MHC class II, CD80 (B7-1) and CD86 (B7-2) expressed on the surface of DCs that were isolated from dLNs or spleen 3 days post-immunization in an EAE model. Changes to DC function influenced Th1/Th17 subset polarization. Furthermore, the number of CD44(+) monocytes (which might trigger EAE progression) was also significantly decreased in dLNs, spleen, subarachnoid space and the spinal cord parenchyma after AT-RA treatment. These findings are the first to demonstrate that AT-RA impairs the antigen-presenting capacity of DCs, leading to down-regulation of pathogenic Th1 and Th17 inflammatory cell responses and reducing EAE severity.


Asunto(s)
Antioxidantes/uso terapéutico , Células Dendríticas/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Monocitos/efectos de los fármacos , Tretinoina/uso terapéutico , Animales , Presentación de Antígeno/efectos de los fármacos , Antígenos CD/genética , Antígenos CD/inmunología , Antioxidantes/farmacología , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunización , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/patología , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/inmunología , Médula Espinal/patología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/patología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/patología , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...