Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Stroke ; 55(8): 2151-2162, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38946544

RESUMEN

BACKGROUND: GPR65 (G protein-coupled receptor 65) can sense extracellular acidic environment to regulate pathophysiological processes. Pretreatment with the GPR65 agonist BTB09089 has been proven to produce neuroprotection in acute ischemic stroke. However, whether delayed BTB09089 treatment and neuronal GPR65 activation promote neurorestoration remains unknown. METHODS: Ischemic stroke was induced in wild-type (WT) or GPR65 knockout (GPR65-/-) mice by photothrombotic ischemia. Male mice were injected intraperitoneally with BTB09089 every other day at days 3, 7, or 14 poststroke. AAV-Syn-GPR65 (adenoassociated virus-synapsin-GPR65) was utilized to overexpress GPR65 in the peri-infarct cortical neurons of GPR65-/- and WT mice. Motor function was monitored by grid-walk and cylinder tests. The neurorestorative effects of BTB09089 were observed by immunohistochemistry, Golgi-Cox staining, and Western blotting. RESULTS: BTB09089 significantly promoted motor outcomes in WT but not in GPR65-/- mice, even when BTB09089 was delayed for 3 to 7 days. BTB09089 inhibited the activation of microglia and glial scar progression in WT but not in GPR65-/- mice. Meanwhile, BTB09089 reduced the decrease in neuronal density in WT mice, but this benefit was abolished in GPR65-/- mice and reemerged by overexpressing GPR65 in peri-infarct cortical neurons. Furthermore, BTB09089 increased the GAP43 (growth-associated protein-43) and synaptophysin puncta density, dendritic spine density, dendritic branch length, and dendritic complexity by overexpressing GPR65 in the peri-infarct cortical neurons of GPR65-/- mice, which was accompanied by increased levels of p-CREB (phosphorylated cAMP-responsive element-binding protein). In addition, the therapeutic window of BTB09089 was extended to day 14 by overexpressing GPR65 in the peri-infarct cortical neurons of WT mice. CONCLUSIONS: Our findings indicated that delayed BTB09089 treatment improved neurological functional recovery and brain tissue repair poststroke through activating neuronal GRP65. GPR65 overexpression may be a potential strategy to expand the therapeutic time window of GPR65 agonists for neurorehabilitation after ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Ratones Noqueados , Neuronas , Receptores Acoplados a Proteínas G , Animales , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/agonistas , Ratones , Accidente Cerebrovascular Isquémico/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Rehabilitación de Accidente Cerebrovascular , Fármacos Neuroprotectores/farmacología , Ratones Endogámicos C57BL
2.
J Cardiovasc Pharmacol ; 83(5): 474-481, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38113918

RESUMEN

ABSTRACT: Studies have examined the therapeutic effect of levosimendan on cardiovascular diseases such as heart failure, perioperative cardiac surgery, and septic shock, but the specific mechanism in mice remains largely unknown. This study aimed to investigate the relaxation mechanism of levosimendan in the thoracic aorta smooth muscle of mice. Levosimendan-induced relaxation of isolated thoracic aortic rings that were precontracted with norepinephrine or KCl was recorded in an endothelium-independent manner. Vasodilatation by levosimendan was not associated with the production of the endothelial relaxation factors nitric oxide and prostaglandins. The voltage-dependent K + channel (K V ) blocker (4-aminopyridine) and selective K Ca blocker (tetraethylammonium) had no effect on thoracic aortas treated with levosimendan, indicating that K V and K Ca channels may not be involved in the levosimendan-induced relaxation mechanism. Although the inwardly rectifying K + channel (K ir ) blocker (barium chloride) and the K ATP channel blocker (glibenclamide) significantly inhibited levosimendan-induced vasodilation in the isolated thoracic aorta, barium chloride had a much stronger inhibitory effect on levosimendan-induced vasodilation than glibenclamide, suggesting that levosimendan-induced vasodilation may be mediated by K ir channels. The vasodilation effect and expression of K ir 2.1 induced by levosimendan were further enhanced by the PKC inhibitor staurosporine. Extracellular calcium influx was inhibited by levosimendan without affecting intracellular Ca 2+ levels in the isolated thoracic aorta. These results suggest that K ir channels play a more important role than K ATP channels in regulating vascular tone in larger arteries and that the activity of the K ir channel is enhanced by the PKC pathway.


Asunto(s)
Aorta Torácica , Músculo Liso Vascular , Proteína Quinasa C , Simendán , Vasodilatación , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Simendán/farmacología , Masculino , Vasodilatación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Ratones , Canales de Potasio de Rectificación Interna/metabolismo , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Vasodilatadores/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Bloqueadores de los Canales de Potasio/farmacología
3.
Exp Neurol ; 380: 114892, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39047809

RESUMEN

T-cell death-associated gene 8 (TDAG8), a G-protein-coupled receptor sensing physiological or weak acids, regulates inflammatory responses. However, its role in traumatic brain injury (TBI) remains unknown. Our recent study showed that delayed CO2 postconditioning (DCPC) has neuroreparative effects after TBI. We hypothesized that activating astrocytic TDAG8 is a key mechanism for DCPC. WT and TDAG8-/- mice received DCPC daily by transiently inhaling 10% CO2 after controlled cortical impact (CCI). HBAAV2/9-GFAP-m-TDAG8-3xflag-EGFP was used to overexpress TDAG8 in astrocytes. The beam walking test, mNSS, immunofluorescence and Golgi-Cox staining were used to evaluate motor function, glial activation and dendritic plasticity. DCPC significantly improved motor function; increased total dendritic length, neuronal complexity and spine density; inhibited overactivation of astrocytes and microglia; and promoted the expression of astrocytic brain-derived neurotrophic factor in WT but not TDAG8-/- mice. Overexpressing TDAG8 in astrocytes surrounding the lesion in TDAG8-/- mice restored the beneficial effects of DCPC. Although the effects of DCPC on Days 14-28 were much weaker than those of DCPC on Days 3-28 in WT mice, these effects were further enhanced by overexpressing astrocytic TDAG8. Astrocytic TDAG8 is a key target of DCPC for TBI rehabilitation. Its overexpression is a strategy that broadens the therapeutic window and enhances the effects of DCPC.

4.
Neuromolecular Med ; 25(4): 632-643, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37843792

RESUMEN

Orexin and its receptors are closely related to the pathogenesis of Alzheimer's disease (AD). Although the expression of orexin system genes under physiological condition has circadian rhythm, the diurnal characteristics of orexin system genes, and its potential role in the pathogenesis in AD are unknown. In the present study, we hope to elucidate the diurnal characteristics of orexin system genes at the early stage of AD, and to investigate its potential role in the development of AD neuropathology. We firstly detected the mRNA levels of orexin system genes, AD risk genes and core clock genes (CCGs) in hypothalamus and hippocampus in 6-month-old male 3xTg-AD mice and C57BL/6J (wild type, WT) control mice, then analyzed diurnal expression profiles of all genes using JTK_CYCLE algorithm, and did the correlation analysis between expression of orexin system genes and AD risk genes or CCGs. In addition, the expression of ß-amyloid protein (Aß) and phosphorylated tau (p-tau) protein were measured. The results showed that the diurnal mRNA expression profiles of PPO, OX1R, OX2R, Bace2, Bmal1, Per1, Per2 and Cry1 in the hypothalamus, and gene expression of OX1R, OX2R, Bace1, Bmal1, Per1 and Cry2 in the hippocampus in 3xTg-AD mice were different from that in WT mice. Furthermore, there is positive correlation between orexin system genes and AD risk genes or CCGs in the brain in 3xTg-AD mice. In addition, the expression of Aß and p-tau in hippocampus in 3xTg-AD mice were significantly increased, and the expression of p-tau is higher in night than in day. These results indicate that the abnormal expression profiles of orexin system genes and its interaction with AD risk genes or CCGs might exert important role in the pathogenesis of AD, which will increase the expression of Aß and p-tau, and accelerate the development of AD.


Asunto(s)
Enfermedad de Alzheimer , Orexinas , Animales , Masculino , Ratones , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Factores de Transcripción ARNTL/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Transgénicos , Orexinas/genética , ARN Mensajero/genética , Proteínas tau/genética , Proteínas tau/metabolismo
5.
Transl Stroke Res ; 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36853417

RESUMEN

Acidic postconditioning by transient CO2 inhalation applied within minutes after reperfusion has neuroprotective effects in the acute phase of stroke. However, the effects of delayed chronic acidic postconditioning (DCAPC) initiated during the subacute phase of stroke or other acute brain injuries are unknown. Mice received daily DCAPC by inhaling 5%/10%/20% CO2 for various durations (three cycles of 10- or 20-min CO2 inhalation/10-min break) at days 3-7, 7-21, or 3-21 after photothrombotic stroke. Grid-walk, cylinder, and gait tests were used to assess motor function. DCAPC with all CO2 concentrations significantly promoted motor functional recovery, even when DCAPC was delayed for 3-7 days. DCAPC enhanced the puncta density of GAP-43 (a marker of axon growth and regeneration) and synaptophysin (a marker of synaptogenesis) and reduced the amoeboid microglia number, glial scar thickness and mRNA expression of CD16 and CD32 (markers of proinflammatory M1 microglia) compared with those of the stroke group. Cerebral blood flow (CBF) increased in response to DCAPC. Furthermore, the mRNA expression of TDAG8 (a proton-activated G-protein-coupled receptor) was increased during the subacute phase of stroke, while DCAPC effects were blocked by systemic knockout of TDAG8, except for those on CBF. DCAPC reproduced the benefits by re-expressing TDAG8 in the peri-infarct cortex of TDAG8-/- mice infected with HBAAV2/9-CMV-TDAG8-3flag-ZsGreen. Taken together, we first showed that DCAPC promoted functional recovery and brain tissue repair after stroke with a wide therapeutic time window of at least 7 days after stroke. Brain-derived TDAG8 is a direct target of DCAPC that induces neuroreparative effects.

6.
CNS Neurosci Ther ; 29(11): 3378-3390, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37208955

RESUMEN

AIMS: Few treatments are available in the subacute phase of traumatic brain injury (TBI) except rehabilitation training. We previously reported that transient CO2 inhalation applied within minutes after reperfusion has neuroprotective effects against cerebral ischemia/reperfusion injury. In this study, it was hypothesized that delayed CO2 postconditioning (DCPC) starting at the subacute phase may promote neurological recovery of TBI. METHODS: Using a cryogenic TBI (cTBI) model, mice received DCPC daily by inhaling 5%/10%/20% CO2 for various time-courses (one/two/three cycles of 10-min inhalation/10-min break) at Days 3-7, 3-14 or 7-18 after cTBI. Beam walking and gait tests were used to assess the effect of DCPC. Lesion size, expression of GAP-43 and synaptophysin, amoeboid microglia number and glia scar area were detected. Transcriptome and recombinant interferon regulatory factor 7 (Irf7) adeno-associated virus were applied to investigate the molecular mechanisms. RESULTS: DCPC significantly promoted recovery of motor function in a concentration and time-course dependent manner with a wide therapeutic time window of at least 7 days after cTBI. The beneficial effects of DCPC were blocked by intracerebroventricular injection of NaHCO3 . DCPC also increased puncta density of GAP-43 and synaptophysin, and reduced amoeboid microglia number and glial scar formation in the cortex surrounding the lesion. Transcriptome analysis showed many inflammation-related genes and pathways were altered by DCPC, and Irf7 was a hub gene, while overexpression of IRF7 blocked the motor function improvement of DCPC. CONCLUSIONS: We first showed that DCPC promoted functional recovery and brain tissue repair, which opens a new therapeutic time window of postconditioning for TBI. Inhibition of IRF7 is a key molecular mechanism for the beneficial effects of DCPC, and IRF7 may be a potential therapeutic target for rehabilitation after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Dióxido de Carbono , Factor 7 Regulador del Interferón , Animales , Ratones , Lesiones Traumáticas del Encéfalo/metabolismo , Dióxido de Carbono/metabolismo , Dióxido de Carbono/uso terapéutico , Modelos Animales de Enfermedad , Proteína GAP-43/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/uso terapéutico , Sinaptofisina/metabolismo , Sinaptofisina/uso terapéutico
7.
Neurochem Int ; 148: 105080, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34048845

RESUMEN

Astrocytes play a pivotal role in maintaining the central nervous system (CNS) homeostasis and function. In response to CNS injuries and diseases, reactive astrocytes are triggered. By purifying and genetically profiling reactive astrocytes, it has been now found that astrocytes can be activated into two polarization states: the neurotoxic or pro-inflammatory phenotype (A1) and the neuroprotective or anti-inflammatory phenotype (A2). Although the simple dichotomy of the A1/A2 phenotypes does not reflect the wide range of astrocytic phenotypes, it facilitates our understanding of the reactive state of astrocytes in various CNS disorders. This article reviews the recent evidences regarding A1/A2 astrocytes, including (a) the specific markers and morphological characteristics, (b) the effects of A1/A2 astrocytes on the neurovascular unit, and (c) the molecular mechanisms involved in the phenotypic switch of astrocytes. Although many questions remain, a deeper understanding of different phenotypic astrocytes will eventually help us to explore effective strategies for neurological disorders by targeting astrocytes.


Asunto(s)
Astrocitos/patología , Enfermedades del Sistema Nervioso Central/patología , Sistema Nervioso Central/lesiones , Sistema Nervioso Central/patología , Animales , Humanos , Enfermedades Neuroinflamatorias/patología
8.
J Biomater Sci Polym Ed ; 31(14): 1793-1805, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32510282

RESUMEN

Most chemotherapeutic drugs commonly suffer from low aqueous solubility that can potentially limit drugs absorption. Drug nanomerization is an advanced approach to overcoming their poor water-solubility. In this study, class I hydrophobin recombinant HGFI (rHGFI)-based curcumin (Cur) nanoparticles (rHGFI-Cur) were prepared by freeze-drying method. The rHGFI-Cur nanocomposites were characterized by contact angle, transmission electron microscopy, fluorescence microscopy and dynamic light scattering. The results showed that rHGFI could lead to the wettability conversion and stability improved of Cur in water. X-ray photoelectron spectroscopy and Fourier transform infrared suggested that rHGFI could non-covalently bind to Cur to render them hydrophilic through hydrophobic forces. Additionally, drug release and cytotoxicity assays illustrated that rHGFI-Cur nanoparticles could facilitate Cur release and exhibited higher cytotoxicity than free Cur for human esophageal cancer cells TE-1. Thus, it suggested that rHGFI has a great potential application for hydrophobic drug delivery without toxicity.[Formula: see text].


Asunto(s)
Curcumina , Nanopartículas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Solubilidad , Agua
9.
Brain Res Bull ; 164: 146-156, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32858129

RESUMEN

Accumulating evidence suggests that chronic metformin posttreatment offers potent neuroreparative effects against acute brain injury. However, in previous studies, metformin was not initially administered beyond 24 h postinjury, and the effects of delayed metformin treatment in traumatic brain injury (TBI) and other types of acute brain injury and the related mechanisms are unclear. To test this, male C57BL/6 mice received once daily metformin treatment (20, 50 or 100 mg/kg/d, i.p.) at day 1-14, day 1-2, day 1-10, day 3-10, day 5-12 or day 5-28 after cryogenic TBI (cTBI). The results showed that 100 mg/kg/d metformin administered at day 1-14 postinjury significantly promoted motor functional recovery in the beam walking and gait tests and reduced the infarct volume. Metformin (100 mg/kg/d) administered at day 1-10 or day 3-10 but not day 1-2 or day 5-12 after cTBI significantly improved motor functional outcomes at day 7 and 14, and reduced the infarct volume at day 14. Interestingly, the therapeutic time window was further expanded when the duration of metformin treatment starting at day 5 postinjury was extended to 2 weeks. Furthermore, compared with cTBI, the administration of metformin at day 3-10 or day 5-28 after cTBI significantly elevated the expression of phosphorylated adenosine monophosphate-activated protein kinase (AMPK) and growth associated protein 43 (an axonal regeneration marker) and the number of vascular branch points and decreased the area of glial scar and the number of amoeboid microglia in the peri-infarct area at day 14 or 28 postinjury. The above beneficial effects of metformin were blocked by the intracerebroventricular injection of the AMPK inhibitor compound C (40 µg/mouse/d). Our data provide the first evidence that metformin has a wide therapeutic time window for at least 5 days after cTBI, during which it can improve functional recovery by promoting tissue repair and inhibiting glial scar formation and microglial activation in a central AMPK-dependent manner.


Asunto(s)
Adenilato Quinasa/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Destreza Motora/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Animales , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hipoglucemiantes/farmacología , Masculino , Metformina/farmacología , Ratones , Fármacos Neuroprotectores/farmacología , Fosforilación/efectos de los fármacos
10.
Arch Virol ; 154(7): 1093-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19526193

RESUMEN

SARS-CoV infection of human results in antigen-specific cellular and humoral immune responses. However, it is critical to determine whether SARS-CoV-specific memory T cells can persist for long periods of time. In this study, we analyzed the cellular immune response from 21 SARS-recovered individuals who had been diagnosed with SARS in 2003 by using ELISA, CBA, ELISpot and multiparameter flow cytometry assays. Our results demonstrated that low levels of specific memory T cell responses to SARS-CoV S, M, E and N peptides were detected in a proportion of SARS-recovered patients, and IFN-gamma was the predominant cytokine produced by T cells after stimulation with peptides. Cytometry analysis indicated that the majority of memory CD8(+) T cells produced IFN-gamma, whereas memory CD4(+) T cells produced IFN-gamma, IL-2 or TNF-alpha. These results might provide valuable information on the cellular immune response in recovered SARS-CoV patients for the rational design of vaccines against SARS-CoV infection.


Asunto(s)
Memoria Inmunológica , Síndrome Respiratorio Agudo Grave/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Linfocitos T/inmunología , Adulto , Formación de Anticuerpos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diseño de Fármacos , Femenino , Citometría de Flujo , Estudios de Seguimiento , Humanos , Inmunidad Celular , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Masculino , Persona de Mediana Edad , Valores de Referencia , Linfocitos T/virología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/biosíntesis , Adulto Joven
11.
Clin Exp Pharmacol Physiol ; 36(8): 816-21, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19215234

RESUMEN

1. The present study examined the role of the hippocampal histaminergic system in the regulation of spatial memory deficit in rats using the radial arm maze task after scopolamine injection into the bilateral dorsal (DH) or ventral (VH) hippocampus. 2. Bilateral injection of scopolamine (5 microg/site) into both the DH and VH impaired spatial memory in the retrieval memory process. Injection of histamine (50 or 100 ng/site) in the DH and intraperitoneal injection of histidine (100 mg/kg) markedly improved working memory and reference memory deficits induced by scopolamine injection into the DH. The histamine H(1) receptor antagonist pyrilamine (1 microg/site) abolished the ameliorative effects of histidine on working memory deficits, whereas both pyrilamine and the H(2) receptor antagonist cimetidine (0.5 microg/site) abolished the effect of histidine on reference memory. 3. Local injection of histamine (25 or 50 ng/site) into the VH and systemic injection of histidine (50 or 100 mg/kg) markedly improved working memory deficits induced by scopolamine injection into the VH, but did not improve the deficits in reference memory. Injection of both pyrilamine (0.2, 0.5 and 1 microg/site) and cimetidine (0.1 and 0.5 microg/site) into the VH reversed the effects of histidine. 4. The results of the present study indicate that histamine has different actions on cholinergic-related memory in the the DH and VH. Histamine in the DH ameliorates spatial working memory deficits by acting on histamine H(1) receptors and reference memory deficits through both H(1) and H(2) receptors. However, histamine in the VH ameliorates working memory deficits via an action on both H(1) and H(2) receptors.


Asunto(s)
Hipocampo/efectos de los fármacos , Histamina/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/prevención & control , Memoria a Corto Plazo/efectos de los fármacos , Escopolamina/efectos adversos , Animales , Cimetidina/farmacología , Relación Dosis-Respuesta a Droga , Histamina/administración & dosificación , Histamina/metabolismo , Histamina/fisiología , Antagonistas de los Receptores Histamínicos H1/farmacología , Antagonistas de los Receptores H2 de la Histamina/farmacología , Histidina/administración & dosificación , Histidina/farmacología , Masculino , Trastornos de la Memoria/inducido químicamente , Pirilamina/farmacología , Ratas , Ratas Sprague-Dawley , Conducta Espacial/efectos de los fármacos
12.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 38(6): 644-8, 2009 11.
Artículo en Zh | MEDLINE | ID: mdl-20014493

RESUMEN

Vascular endothelial growth factor (VEGF or VEGF-A) is a hypoxia induced angiogenic growth factor that is potent in neurotrophy,neuroprotection, anti-apoptosis and cell proliferation. Recent reports suggest that VEGF is related to many central nervous system diseases, such as cerebral ischemic disease, Alzheimer's disease and Parkinson's disease. Further study of the relationship between VEGF and central nervous system diseases,and investigation of VEGF related drugs will shed light on a new way for treatment of central nervous system diseases.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Isquemia Encefálica/metabolismo , Enfermedad de Parkinson/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Humanos , Fármacos Neuroprotectores/farmacología , Factor A de Crecimiento Endotelial Vascular/clasificación , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 38(6): 579-83, 2009 11.
Artículo en Zh | MEDLINE | ID: mdl-20014482

RESUMEN

OBJECTIVE: To investigate the effect of endogenous histamine on ischemic preconditioning induced cerebral ischemic tolerance in rats. METHODS: Wild-type (WT) mice and histidine decarboxylase knock-out (HDC-KO) mice were preconditioned by bilateral carotid artery occlusion (BCCAO) for 6, 10,or 14 min and reperfused for 48 h,then subjected to permanent BCCAO and the survival time of WT and HDC-KO mice subjected to permanent BCCAO was observed. Histamine levels in the hypothalamus, hippocampus, striatum and cortex at 0.5 h,5 h or 48 h after 10 min BCCAO were determined with high-performance liquid chromatography. RESULT: Ten minutes ischemic preconditioning significantly prolonged the survival time of WT mice subjected to permanent BCCAO. However,in HDC-KO mice, the ischemic tolerance was not induced with 10 min preconditioning. The histamine levels at 0.5 h or 48 h increased after 10 min preconditioning, but not at 5 h. CONCLUSION: Endogenous histamine in brain may be an essential mediator in ischemic preconditioning induced cerebral ischemic tolerance.


Asunto(s)
Isquemia Encefálica/terapia , Histamina/metabolismo , Precondicionamiento Isquémico , Daño por Reperfusión/prevención & control , Animales , Isquemia Encefálica/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria
14.
Int J Biol Macromol ; 121: 1118-1125, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30340010

RESUMEN

In this study, we fabricated a series of novel sodium alginate/ZnO hydrogel beads to optimize the release profile of curcumin (Cur) and to avoid the burst release associated with pure hydrogels, which were used to mitigate the weaknesses of Cur, such as rapid physiological clearance and sensitivity to ultraviolet (UV) light and alkaline solutions. The results show that the composite hydrogel beads exhibit good pH sensitivity and controlled-release capacity, which could prolong the residence time of Cur in the gastrointestinal tract. After exposure to UV irradiation for 6 h, the 1,1-Diphenyl-2-picrylhydrazyl (DPPH) scavenging capacity of Cur-loaded hydrogel beads was decreased by only 13.70%, whereas that of pure Cur decreased by 62.04% under the same conditions; therefore, the encapsulated Cur showed a higher antioxidant activity. The composite hydrogel beads protected the Cur from light degradation and can therefore prolong the antioxidant activity of Cur. These results are beneficial for the design of delivery systems to entrap and control the release of unstable drugs.


Asunto(s)
Alginatos/química , Antioxidantes/química , Curcumina/química , Portadores de Fármacos/química , Liberación de Fármacos , Hidrogeles/química , Óxido de Zinc/química , Compuestos de Bifenilo/química , Curcumina/metabolismo , Tracto Gastrointestinal/metabolismo , Concentración de Iones de Hidrógeno , Microesferas , Picratos/química
15.
Food Chem ; 283: 397-403, 2019 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-30722890

RESUMEN

To deal with serious environmental pollution resulting from plastic packaging materials, biodegradable films using chitosan (CS) are gaining considerable increase gradually. However, chitosan films lack important properties to meet the preserved demands. This study aimed to develop new bio-based films incorporated with carboxymethyl chitosan-ZnO (CMCS-ZnO) nanoparticles and sodium alginate (SA) to overcome the weakness of CS films. CMCS-ZnO nanoparticles were successfully synthesized in the matrix of CMCS through direct precipitation method, which showed an average diameter of 100 nm. Multilayer films with CS film as the outer layer and SA film as the inner layer were prepared by solution casting method. The addition of CMCS-ZnO nanoparticles led to enhanced tensile strength, and to better water vapor resistance. The as-prepared films exhibited distinctive antibacterial activity against S. aureus and E. coli. The results suggested that the as-prepared film is expected to be a promising material for food packaging.


Asunto(s)
Antibacterianos/química , Quitosano/análogos & derivados , Nanopartículas del Metal/química , Nanocompuestos/química , Óxido de Zinc/química , Alginatos/química , Antibacterianos/farmacología , Quitosano/química , Escherichia coli/efectos de los fármacos , Embalaje de Alimentos , Nanocompuestos/toxicidad , Tamaño de la Partícula , Solubilidad , Staphylococcus aureus/efectos de los fármacos , Resistencia a la Tracción , Agua/química
16.
Int J Biol Macromol ; 141: 1191-1198, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31518622

RESUMEN

To control release of drugs sensitive to gastrointestinal (GI) environmental effects or irritating to stomach, such as diclofenac sodium (DS), sodium alginate (SA) hydrogel beads are gaining considerable attention gradually. However, due to high swelling ratio, the sustained release performance of SA hydrogel is still far from satisfactory. The objective of this research was to develop new drug delivery device based on SA and ZnO nanoparticles (ZnO NPs). ZnO NPs were prepared by direct precipitation method, and carboxymethyl chitosan (CMCS) acted as stabilizing agent to dominate the preparation of ZnO NPs. The incorporation of CMCS-ZnO NPs resulted in slower and sustained release of DS in vitro. In vivo pharmacokinetics studies showed the bioavailability of DS was better after oral administration of DS-loaded SA/CMCS-ZnO hydrogel beads. These results suggested that SA/CMCS-ZnO hydrogel beads will be a prospective material for loading drugs sensitive to GI environmental effects or irritating to stomach.


Asunto(s)
Alginatos/química , Quitosano/análogos & derivados , Diclofenaco/química , Liberación de Fármacos , Hidrogeles/química , Microesferas , Óxido de Zinc/química , Células 3T3 , Animales , Quitosano/química , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/toxicidad , Ratones , Ratas , Distribución Tisular
17.
Immunol Invest ; 37(1): 79-96, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18214801

RESUMEN

Natural killer cells (NK) from umbilical cord blood (CB) play an important role in allogeneic stem cell transplantation and defending infections of newborn. Based on the surface expression of CD56 and CD16 or inhibitory and activatory receptors, NK cells could be subdivided into various subsets with distinct functions. To investigate the biological characterization of NK subsets, the phenotypes and intracellular proteins in freshly isolated CB NK subsets were analyzed at the single cell level by flow cytometry in current study. The production of IFN-gamma and cytotoxicity against K562 target cells were also evaluated after stimulation with IL-12. The results showed that NK cells from CB could be divided into four subsets on the basis of CD56 and CD16 expression. Interestingly, CB NK cells expressed CD45RA but not CD45RO molecules that is similar to the naïve T cells. Moreover, CD27, a memory T cell marker, highly expressed on CD56(hi)CD16- NK cells. The killing-associated molecules, NKG2A, NKG2D, CD95 and the intracellular granzyme B and perforin were heterogeneously expressed among the 4 subsets. Addition of IL-12 into cultures resulted in the induction of IFN-gamma expression by CD56(hi)CD16- and CD56(lo)CD16- subsets and the enhancement of NK cytolytic activity. Taken together, this study elucidated the heterogeneity in phenotypes and biological functions of CB NK cells.


Asunto(s)
Sangre Fetal/citología , Inmunofenotipificación , Células Asesinas Naturales/clasificación , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/citología , Moléculas de Adhesión Celular/metabolismo , Femenino , Humanos , Memoria Inmunológica , Recién Nacido , Interleucina-12/metabolismo , Interleucina-2/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Embarazo
18.
Neurosci Lett ; 678: 68-75, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29727731

RESUMEN

Glial scar impedes axon regeneration and functional recovery following traumatic brain injury (TBI). Although it has been shown that rapamycin (a specific inhibitor of mammalian target of rapamycin) can reduce astrocyte reactivation in the early stage of TBI, its effect on glial scar formation has not been characterized in TBI and other acute brain injury models. To test this, ICR mice received daily administration of rapamycin (0.5 or 1.5 mg/kg, i.p.) beginning at 1 h after cryogenic TBI (cTBI). The results showed that at 3 d post-injury, 1.5 mg/kg rapamycin increased cTBI-induced motor functional deficits and infarct size, and attenuated astrocyte reactivation in the ipsilateral cortex, while 0.5 mg/kg rapamycin did not worsen brain damage and only slightly attenuated astrocyte reactivation. Furthermore, at 7 and 14 d after cTBI, 0.5 mg/kg rapamycin group showed a better motor functional performance than cTBI group. At 14 d post-injury, 0.5 mg/kg rapamycin significantly reduced the area and thickness of glial scar and chondroitin sulfate proteoglycan expression, accompanied by decreased expression of p-S6 and enhanced expression of growth associated protein 43 (an axon regeneration marker) in the region of glial scar. Our data suggest that long-term treatment with rapamycin can inhibit glial scar formation after cTBI, which may be involved in the mechanisms of increased axon regeneration and improved neurological functional recovery, and low-dose rapamycin may be more beneficial for such a therapy.


Asunto(s)
Astrocitos/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/complicaciones , Encéfalo/efectos de los fármacos , Cicatriz/metabolismo , Sirolimus/administración & dosificación , Animales , Astrocitos/metabolismo , Axones/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Cicatriz/etiología , Frío , Masculino , Ratones Endogámicos ICR , Regeneración Nerviosa/efectos de los fármacos , Recuperación de la Función , Prueba de Desempeño de Rotación con Aceleración Constante , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
19.
Neurochem Int ; 107: 43-56, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28087295

RESUMEN

Ischemic postconditioning (PostC) is defined as a series of rapid intermittent interruptions of blood flow at the phase of reperfusion, which produces neuroprotection against cerebral ischemia/reperfusion injury via mobilizing the brain's own endogenous adaptive mechanisms. Now the concept of conventional ischemic PostC has been extended to limb remote ischemic PostC and chemical PostC with hypoxia, volatile anesthetic, CO2, etc. According to the different temporal profile of PostC, it is divided into rapid and delayed PostC. Rapid PostC is applied within a few seconds to minutes after reperfusion, while delayed PostC is applied at a few hours to days after reperfusion. Although the neuroprotective mechanisms of PostC are not completely elucidated, a series of mechanisms have been found to connect with PostC in the central nervous system, such as regulating synaptic signaling, attenuating oxidative stress and inflammation, maintaining mitochondrial integrity, inhibiting endoplasmic reticulum stress, regulating autophagy, activating PI3K/Akt pathway, inhibiting apoptosis, protecting neurovascular unit, etc. Based on these multiple protective mechanisms, PostC has high expectations to translate to the clinic, but a few issues should be resolved such as the time window, risks, efficiency, the impact of age, gender, hypertension, hyperlipidemia and t-PA, and clinical maneuverability. Even so, PostC could soon be at the bedside if the clinical trials are carefully planned.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Poscondicionamiento Isquémico/métodos , Fármacos Neuroprotectores/uso terapéutico , Animales , Humanos , Neuroprotección/efectos de los fármacos , Neuroprotección/fisiología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
20.
J Cereb Blood Flow Metab ; 34(2): 275-83, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24192637

RESUMEN

Acidosis is one of the key components in cerebral ischemic postconditioning that has emerged recently as an endogenous strategy for neuroprotection. We set out to test whether acidosis treatment at reperfusion can protect against cerebral ischemia/reperfusion injury. Adult male C57BL/6 J mice were subjected to 60-minute middle cerebral arterial occlusion followed by 24-hour reperfusion. Acidosis treatment by inhaling 10%, 20%, or 30% CO2 for 5 or 10 minutes at 5, 50, or 100 minutes after reperfusion was applied. Our results showed that inhaling 20% CO2 for 5 minutes at 5 minutes after reperfusion-induced optimal neuroprotection, as revealed by reduced infarct volume. Attenuating brain acidosis with NaHCO3 significantly compromised the acidosis or ischemic postconditioning-induced neuroprotection. Consistently, both acidosis-treated primary cultured cortical neurons and acute corticostriatal slices were more resistant to oxygen-glucose deprivation/reperfusion insult. In addition, acidosis inhibited ischemia/reperfusion-induced apoptosis, caspase-3 expression, cytochrome c release to cytoplasm, and mitochondrial permeability transition pore (mPTP) opening. The neuroprotection of acidosis was inhibited by the mPTP opener atractyloside both in vivo and in vitro. Taken together, these findings indicate that transient mild acidosis treatment at reperfusion protects against cerebral ischemia/reperfusion injury. This neuroprotection is likely achieved, at least partly, by inhibiting mPTP opening and mitochondria-dependent apoptosis.


Asunto(s)
Acidosis , Dióxido de Carbono/farmacología , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión , Bicarbonato de Sodio/sangre , Accidente Cerebrovascular , Acidosis/sangre , Acidosis/inducido químicamente , Acidosis/tratamiento farmacológico , Animales , Isquemia Encefálica/sangre , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/fisiopatología , Masculino , Ratones , Daño por Reperfusión/sangre , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/fisiopatología , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA