Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 527(1): 22-28, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32446371

RESUMEN

Acute spontaneous intracerebral hemorrhage (ICH) is a life-threatening disease. It is often accompanied by severe neurological sequelae largely caused by the loss of integrity of the neural circuits. However, these neurological sequelae have few strong medical interventions. Designer receptors exclusively activated by designer drugs (DREADDs) are important chemogenetic tools capable of precisely modulating the activity of neural circuits. They have been suggested to have therapeutic effects on multiple neurological diseases. Despite this, no empirical research has explored the effects of DREADDs on functional recovery after ICH. We aimed to explore whether the long-term excitation of glutamatergic neurons in primary motor cortex (M1) by DREADD could promote functional recovery after ICH. We used CaMKII-driven Gq/Gi-DREADDs to activate/inhibit M1 glutamatergic neurons for 21 consecutive days, and examined their effects on behavioral and cognitive deficits caused by ICH in a mouse model of ICH targeting striatum. Long-term chemogenetic activation of the M1 glutamatergic neurons increased the spatial memory and sensorimotor ability of mice suffering from ICH. It also attenuated the mitochondrial dysfunctions of striatal neurons by raising the ATP levels and mitochondrial membrane potential while decreasing the 8-OHdG levels. These results strongly suggest that selective stimulation of the M1 glutamatergic neurons contributes to functional recovery after ICH presumably through alleviation of mitochondrial dysfunctions.


Asunto(s)
Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/tratamiento farmacológico , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Neuronas/efectos de los fármacos , Animales , Células Cultivadas , Hemorragia Cerebral/fisiopatología , Disfunción Cognitiva/fisiopatología , Modelos Animales de Enfermedad , Ligandos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Neuronas/patología , Recuperación de la Función
2.
Int J Clin Exp Pathol ; 11(1): 427-437, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-31938128

RESUMEN

Multiple chromosome aberrations are responsible for tumorigenesis of esophagus squamous cell carcinoma (ESCC). To characterize genetic alterations by comparative genomic hybridization (CGH) and their relation to ESCC, We enrolled 54 members with ESCC from Kazakh's patients. We found that the deletions of 3p (P = 0.032), 17p (P = 0.004), 22q (P = 0.000) and gains of 5p (P = 0.000), 11q (P = 0.000) were significantly correlated with the location of tumors. Losses of 1p (P = 0.005), 3p (P = 0.006), 22q (P = 0.024) and gains of 3q (P = 0.043), 8q (P = 0.038), 18q (P = 0.046) were also found more frequently in patients with larger diameter disease. The loss of 19q (P = 0.005) and gains of l3q (P = 0.045), 18p (P = 0.018) were significantly correlated with pathologic grade. The gain of 7p (P = 0.009) and deletion of 19q (P = 0.018) were seen more frequently in patients with Grade III-IV tumors. Chromosome amplifications in ESCC at 1q (P = 0.008), 7p (P = 0.008), 8q (P = 0.018) and deletions at 3p (P = 0.021), 11q (P = 0.002), 17p (P = 0.012) were related to lymph node metastasis; the gains of 1q (P = 0.026) and 6q (P = 0.017) and the loss of 11q (P = 0.001) were significant in different isoforms of HPV infection. We identified some chromosomes in which the genes were related to the tumorgenesis of ESCC, which may be a theme for future investigation.

3.
Int J Mol Med ; 41(2): 659-668, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29207055

RESUMEN

Pulmonary fibrosis (PF) is a chronic lung disease. The transforming growth factor-ß1 (TGF-ß1)/Smad3 signaling pathway plays an important role in the pathogenesis of pulmonary fibrosis. Bone marrow-derived mesenchymal stem cells (BMSCs) have been shown to be a modulator of the molecular aspects of the fibrosis pathway. However, it is still unknown as to whether the conditioned medium from BMSCs (BMSCs-CM) inhibits the epithelial-mesenchymal transition (EMT) process. This study confirmed the hypothesis that BMSCs-CM exerts an anti-fibrotic effect on human type II alveolar epithelial cells (A549) by suppressing the phosphorylation of Smad3. We used the A549 cells in vitro to detect morphological evidence of EMT by phase-contrast microscopy. These cells were randomly divided into 4 groups as follows: the control group, the TGF-ß1 group, the SIS3 (specific inhibitor of Smad3) group and the BMSCs-CM group. The immunofluorescence method was used to determined the location of E-cadherin (E-calcium mucins; E-cad), α-smooth muscle actin (α-SMA) and p-Smad3. The expression levels of E-cad, CK8, α-SMA, vimentin, p-Smad3, Snail1, collagen I (COLI) and collagen III (COLIII) were detected by western blot analysis. Following exposure to TGF-ß1, the A549 cells displayed a spindle-shaped fibroblast-like morphology. In accordance with these morphological changes, the expression levels of E-cad and CK8 were downregulated, while the expression levels of α-SMA and vimentin were upregulated. Along with this process, the expression levels of p-Smad3, Snail1, COLI and COLIII were increased. However, the cells in the BMSCs-CM group and SIS3 group exhibited a decrease in the levels of α-SMA and vimentin (which had been upregulated by TGF-ß1), and an increase in the levels of E-cad and CK8 expression (which had been downregulated by TGF-ß1). On the whole, these results indicated that BMSCs-CM suppressed the EMT which might be associated with TGF-ß1/Smad3. This study provides the theoretical basis for the research of the mechanisms responsible for pulmonary disease.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Fibrosis Pulmonar/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/genética , Células A549 , Actinas , Células de la Médula Ósea/química , Células de la Médula Ósea/metabolismo , Cadherinas/genética , Medios de Cultivo Condicionados/química , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Isoquinolinas/farmacología , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/metabolismo , Fibrosis Pulmonar/metabolismo , Piridinas/farmacología , Pirroles/farmacología , Proteína smad3/antagonistas & inhibidores
4.
Exp Ther Med ; 11(6): 2577-2582, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27284351

RESUMEN

The aim of the present study was to evaluate the effects of bone marrow-derived mesenchymal stem cells (BMSCs) on the expression of the autophagy-associated proteins, microtubule-associated protein light chain 3 (LC-3) and autophagy-related gene Beclin-1 (Beclin-1), in alveolar macrophages (AMs) in a rat model of silicosis. Furthermore, the study investigated the molecular mechanisms underlying the effects of BMSC treatment. A population of 60 adult female Sprague-Dawley (SD) rats were allocated at random into three groups, namely the control, model and BMSC treatment groups (n=20 per group). BMSCs were isolated from five male SD rats (age, 6-8 weeks) and cultured in vitro. The silicosis model was established using a single 1.0-ml infusion of silicon dioxide suspension administered via non-exposed tracheal intubation. Rats in the BMSC treatment group received a 1.0-ml transplantation of BMSCs (1×106/ml). The rats were sacrificed on days 1, 7, 14 and 28 after modeling, and AMs were extracted from the rats using bronchoalveolar lavage. Third-generation BMSCs were identified using flow cytometry with fluorescein isothiocyanate staining, and the morphological characteristics of the AMs were observed using hematoxylin and eosin staining. The expression levels of LC-3 and Beclin-1 were determined using immunocytochemistry sand western blot analysis. The expression levels of LC-3 and Beclin-1 were found to be increased at all the time points in the model group. LC-3 and Beclin-1 levels began to increase at day 1, peaked at day 14 and decreased after day 28; however, the levels remained elevated compared with the basal expression levels. The AMs of the BMSC treatment group exhibited significantly alleviated pathological symptoms compared with the model group AMs, as indicated by significantly decreased expression levels of LC-3 and Beclin-1 at each time point. Therefore, the results indicated that autophagy was promoted in the AMs of the silicosis model rats. Furthermore, treatment with BMSCs was demonstrated to reduce the expression levels of LC-3 and Beclin-1, subsequently inhibiting autophagic activity and mitigating the damage associated with silicosis.

5.
Mol Med Rep ; 13(6): 5248-54, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27122047

RESUMEN

Traumatic brain injury (TBI) involves primary and secondary injury cascades that underlie delayed neuronal dysfunction and death, leading to long­term cognitive deficits, and effective therapeutic strategies targeting neuronal death remain elusive. The present study aimed to determine whether the administration of resveratrol (100 mg/kg) was able to significantly enhance functional recovery in a rat model of TBI and whether resveratrol treatment was able to upregulate synaptic protein expression and suppress post­TBI neuronal autophagy. The results demonstrated that daily treatment with resveratrol attenuated TBI­induced brain edema and improved spatial cognitive function and neurological impairment in rats. The expression of synaptic proteins was downregulated following TBI and this phenomenon was partly reversed by treatment with resveratrol. In addition, resveratrol was observed to significantly reduce the levels of the autophagic marker proteins, microtubule­associated protein light chain 3­II and Beclin1, in the hippocampus compared with the TBI group. Therefore, these results suggest that resveratrol may represent a novel therapeutic strategy for TBI, and that this protection may be associated with the upregulation of synaptophysin, postsynaptic density protein 95 and the suppression of neuronal autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/prevención & control , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Estilbenos/farmacología , Sinapsis/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Masculino , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Resveratrol , Sinapsis/patología
6.
Int J Mol Med ; 37(4): 921-30, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26936125

RESUMEN

Previous research has demonstrated that traumatic brain injury (TBI) activates autophagy and a neuroinflammatory cascade that contributes to substantial neuronal damage and behavioral impairment, and Toll-like receptor 4 (TLR4) is an important mediator of this cascade. In the present study, we investigated the hypothesis that resveratrol (RV), a natural polyphenolic compound with potent multifaceted properties, alleviates brain damage mediated by TLR4 following TBI. Adult male Sprague Dawley rats, subjected to controlled cortical impact (CCI) injury, were intraperitoneally injected with RV (100 mg/kg, daily for 3 days) after the onset of TBI. The results demonstrated that RV significantly reduced brain edema, motor deficit, neuronal loss and improved spatial cognitive function. Double immunolabeling demonstrated that RV decreased microtubule-associated protein 1 light chain 3 (LC3), TLR4­positive cells co-labeled with the hippocampal neurons, and RV also significantly reduced the number of TLR4­positive neuron­specific nuclear protein (NeuN) cells following TBI. Western blot analysis revealed that RV significantly reduced the protein expression of the autophagy marker proteins, LC3II and Beclin1, in the hippocampus compared with that in the TBI group. Furthermore, the levels of TLR4 and its known downstream signaling molecules, nuclear factor-κB (NF-κB), and the inflammatory cytokines, interleukin (IL)-1ß and tumor necrosis factor (TNF)-α were also decreased after RV treatment. Our results suggest that RV reduces neuronal autophagy and inflammatory reactions in a rat model of TBI. Thus, we suggest that the neuroprotective effect of RV is associated with the TLR4/NF-κB signaling pathway.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Encéfalo/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Fármacos Neuroprotectores/uso terapéutico , Estilbenos/uso terapéutico , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Autofagia/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Masculino , Memoria/efectos de los fármacos , Actividad Motora/efectos de los fármacos , FN-kappa B/análisis , FN-kappa B/inmunología , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/patología , Ratas Sprague-Dawley , Resveratrol , Receptor Toll-Like 4/análisis , Receptor Toll-Like 4/inmunología
7.
Oncol Rep ; 35(3): 1711-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26707131

RESUMEN

In previous studies, we demonstrated that rhein lysinate (RHL), the salt of rhein and lysine that is easily dissolved in water, inhibited the growth of tumor cells derived from breast and ovarian cancer, hepatocellular carcinoma, cervical cancer and lung carcinoma. Based on these observations, human glioma U87 cells and a xenograft model in BALB/c nude mice were used to examine the antitumor activity of RHL against human glioma. Notably, RHL statistically significantly suppressed the growth of human glioma U87 xenografts in BALB/c nude mice. In vitro, there was a significant reduction in cell proliferation after treatment with RHL in a dose- and time-dependent manner. The overall growth inhibition was correlated with the increase in reactive oxygen species (ROS) production and cell apoptosis. The apoptosis- and cell cycle-related proteins including BAX and Bim were increased, whereas Bcl-2 and cyclin D were decreased in the RHL-treated cells. The results demonstrated that RHL is highly effective against the growth of human glioma U87 xenografts in BALB/c nude mice. The potent antitumor activity of RHL may be mediated through downregulation of Bcl-2 and cyclin D expression and upregulation of BAX and Bim expression.


Asunto(s)
Antraquinonas/administración & dosificación , Proteínas Reguladoras de la Apoptosis/biosíntesis , Ciclina D/biosíntesis , Glioma/tratamiento farmacológico , Lisina/análogos & derivados , Proteínas de la Membrana/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Proteína X Asociada a bcl-2/biosíntesis , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/patología , Humanos , Lisina/administración & dosificación , Ratones , Especies Reactivas de Oxígeno , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Med Rep ; 12(2): 2149-54, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25873133

RESUMEN

The P2X7 inhibitor, brilliant blue G (BBG), has been reported as a neuroprotective drug against a variety of disorders, including neuropathic pain and brain ischemia. Currently, no studies have examined the potential for BBG to provide neuroprotection in animal models of TBI. The aim of the present study was to investigate the neuroprotective effect of BBG on TBI and to determine the underlying mechanisms. The rats were subjected to a diffuse cortical impact injury caused by a modified weight-drop device, and then divided randomly into three groups: the sham-operated, BBG treatment and vehicle groups. In the BBG treatment group, 50 mg/kg brilliant blue G (BBG; 100% pure), a highly specific and clinically useful P2X7 antagonist, was administered via the tail vein 15 min prior to or up to 8 h following TBI. The co-localization of NeuN and protein kinase Cγ (PKCγ) was followed with immunofluorescent staining. The expression of P2X7, PKCγ and inflammatory cytokines was identified by western blot analysis. Wet-dry weight method was used to evaluate brain edema, and motor function outcome was examined using the neurological severity score. The present study demonstrated that the administration of BBG attenuated TBI-induced cerebral edema and the associated motor deficits. Following trauma, BBG treatment significantly reduced the levels of PKCγ and interleukin-1ß in the cortex. The results provide in vivo evidence that BBG exerted neuroprotective effects by attenuating brain edema and improving neurological functions via reducing PKCγ and interleukin-1ß levels following TBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Encéfalo/patología , Fármacos Neuroprotectores/uso terapéutico , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Colorantes de Rosanilina/uso terapéutico , Animales , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Interleucina-1beta/análisis , Proteína Quinasa C/análisis , Ratas Sprague-Dawley , Receptores Purinérgicos P2X7/análisis
9.
Mol Med Rep ; 12(2): 2323-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25872478

RESUMEN

The antimalarial drug, chloroquine (CQ), has been reported as an autophagy inhibitor in a variety of disorders, including Alzheimer's disease and brain ischemia. To the best of our knowledge, no studies to date have examined the potential for CQ to provide neuroprotection in animal models of traumatic brain injury (TBI). The aim of this study was to investigate the neuroprotective actions of CQ in TBI and to determine the mechanisms underlying this effect. Rats were immediately subjected to a diffuse cortical impact injury caused by a modified weight-drop device and divided randomly into three groups: sham-operated, CQ treatment and vehicle. The CQ treatment group was administered CQ (intraperitoneally, 3 mg/kg body weight) immediately following the induction of injury. The co-localization of neuron-specific nuclear protein (NeuN) and microtubule-associated protein 1 light chain 3 (LC3), was followed by immunofluorescent staining. The expression of LC3 and inflammatory cytokines was identified by western blot analysis. Wet-dry weight method was utilized to evaluate TBI-induced brain edema. Motor function was evaluated using the Neurological Severity Score (NSS) scale and the Morris water maze was employed to assess spatial learning ability. This study demonstrated that the administration of CQ attenuates TBI-induced cerebral edema, and the associated motor and cognitive functional deficits that occur post-injury. Following the induction of cerebral trauma, CQ treatment significantly suppressed neuronal autophagy and reduced expression levels of the inflammatory cytokines, interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α), in the rat hippocampus. Our results have provided in vivo evidence that CQ may exert neuroprotective effects following TBI, in attenuating brain edema and improving neurological functioning, by reducing the damaging consequences of neuronal autophagy and cerebral inflammation.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Cloroquina/farmacología , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Autofagia/efectos de los fármacos , Biomarcadores/metabolismo , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Esquema de Medicación , Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Inyecciones Intraperitoneales , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Interleucina-1beta/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Índices de Gravedad del Trauma , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
10.
Mol Med Rep ; 11(6): 4419-23, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25626126

RESUMEN

Connexins, gap junction proteins, have short half­lives of only a few hours; therefore, degradation of these proteins can rapidly modulate their function. Autophagy is a type of degradation pathway that has been implicated in several diseases and was reported to be induced following traumatic brain injury (TBI). The aim of the present study was to investigate the involvement of neuronic autophagy in proteolysis of phosphorylated connexin 43 (p­Cx43) in hippocampal astrocytes following TBI in rats. Western blot analysis and immunofluorescence showed a TBI­induced increase in levels of astrocytic p­Cx43 following treatment with 3­methyladenine, an inhibitor of autophagy, in the hippocampus. Internalized gap junctions were observed in the neuronic cytoplasm using transmission electron microscopy. These results demonstrated that neuronic autophagy may regulate cellular levels of p­Cx43 in hippocampal astrocytes following TBI. This therefore indicated that the persistence of p­Cx43 accumulation was due to insufficient degradation capacity of constitutive autophagy.


Asunto(s)
Astrocitos/metabolismo , Autofagia , Lesiones Encefálicas/metabolismo , Conexina 43/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Animales , Lesiones Encefálicas/patología , Uniones Comunicantes/metabolismo , Uniones Comunicantes/ultraestructura , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Proteolisis , Ratas
11.
Mol Med Rep ; 9(1): 77-82, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24220542

RESUMEN

Gap junctions are conductive channels formed by membrane proteins termed connexins, which permit the intercellular exchange of metabolites, ions and small molecules. Previous data demonstrated that traumatic brain injury (TBI) activates autophagy and increases microtubule­associated protein 1 light chain 3 (LC3) immunostaining predominantly in neurons. Although previous studies have identified several extracellular factors that modulate LC3 expression, knowledge of the regulatory network controlling LC3 in health and disease remains incomplete. The aim of the present study was to assess whether gap junctions control the in vivo expression of LC3 in TBI. Using a modified weight­drop device, adult male Sprague­Dawley rats (weight, 350­375 g) were subjected to TBI. Phosphorylated gap junction protein levels and LC3­â…¡ levels were quantified using western blot analysis. The spatial distribution of immunoreactivity for phosphorylated connexin 43 (p­CX43) and LC3­â…¡ was analyzed by immunofluorescence. The results showed that p­CX43 expression in the hippocampus reached a maximum level 6 h following injury. In addition, the immunoreactivity of p­CX43 was localized in the astrocytes surrounding pyramidal neurons. The LC3­â…¡ protein content remained at high levels 24 h following injury. Double immunolabeling demonstrated that LC3­II dots colocalized with the hippocampus pyramidal neurons. Furthermore, inhibition of p­CX43 reduced TBI­induced autophagy, according to western blot analysis. As astrocytic gap junction coupling is affected in various forms of brain injury, the results suggest that point gap junctions/connexins are important regulators of autophagy in the hippocampal neurons following TBI.


Asunto(s)
Astrocitos/metabolismo , Autofagia , Conexina 43/metabolismo , Hipocampo/metabolismo , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Carbenoxolona/farmacología , Carbenoxolona/uso terapéutico , Antagonistas de los Receptores H2 de la Histamina/farmacología , Antagonistas de los Receptores H2 de la Histamina/uso terapéutico , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
12.
Mol Med Rep ; 8(3): 741-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23842733

RESUMEN

Silicosis is a well-known occupational disease, characterized by epithelial injury, fibroblast proliferation, expansion of the lung matrix and dyspnea. At present, no effective treatment methods for silicosis have been identified. The present study aimed to investigate the protective potential of exogenous bone marrow-derived mesenchymal stem cell (BMSC) transplantation on experimental silica-induced pulmonary fibrosis in rats and analyze the underlying paracrine mechanisms associated with its therapeutic effects. BMSCs were isolated, cultured and passaged from male Sprague-Dawley (SD) rat bone marrow. Third-generation BMSCs were identified by flow cytometry using FITC staining. Following the successful establishment of the silicosis model, exogenous BMSCs were infused into female adult SD rats via the tail vein. Lungs were evaluated using hematoxylin and eosin (H&E) staining. The expression of interleukin-1 receptor antagonist (IL­1RA), interleukin-1 (IL-1) and tumor necrosis factor α (TNF-α) protein was detected by immunohistochemistry and western blot analysis. Co-localization of sex determining region Y (SRY) and IL-1RA expression was determined by double-label immunofluorescence. The distribution of transplanted BMSCs was tracked by monitoring the expression of SRY in rats. Treatment with BMSCs was found to protect the lungs against injury and fibrosis by the suppression of upregulated IL-1 and TNF-α protein, via triggering IL-1RA secretion. This mechanism was hypothesized to be mediated by paracrine signaling. These results indicate that the release of IL­1RA from BMSCs via paracrine mechanisms significantly blocks the production and/or activity of IL-1 and TNF-α. The present study provides an experimental basis for cellular therapy in silicosis.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Silicosis/terapia , Animales , Células Cultivadas , Femenino , Inmunohistoquímica , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1/metabolismo , Pulmón/metabolismo , Pulmón/patología , Masculino , Comunicación Paracrina , Ratas , Ratas Sprague-Dawley , Proteína de la Región Y Determinante del Sexo/metabolismo , Silicosis/metabolismo , Silicosis/patología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Mol Med Rep ; 7(1): 327-31, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23128834

RESUMEN

Diffuse brain injury (DBI) is a leading cause of mortality and disability among young individuals and adults worldwide. In specific cases, DBI is associated with permanent spatial learning dysfunction and motor deficits due to primary and secondary brain damage. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) is a major complex that produces reactive oxygen species (ROS) during the ischemic period. The complex aggravates brain damage and cell death following ischemia/reperfusion injury; however, its role in DBI remains unclear. The present study aimed to investigate the hypothesis that levels of NOX2 (a catalytic subunit of NOX) protein expression and the activation of NOX are enhanced following DBI induction in rats and are involved in aggravating secondary brain damage. A rat model of DBI was created using a modified weight-drop device. Our results demonstrated that NOX2 protein expression and NOX activity were enhanced in the CA1 subfield of the hippocampus at 48 and 72 h following DBI induction. Treatment with apocynin (50 mg/kg body weight), a specific inhibitor of NOX, injected intraperitoneally 30 min prior to DBI significantly attenuated NOX2 protein expression and NOX activation. Moreover, treatment with apocynin reduced brain edema and improved spatial learning function assessed using the Morris water maze. These results reveal that treatment with apocynin may provide a new neuroprotective therapeutic strategy against DBI by diminishing the upregulation of NOX2 protein and NOX activity.


Asunto(s)
Acetofenonas/farmacología , Edema Encefálico/etiología , Lesiones Encefálicas/complicaciones , Discapacidades para el Aprendizaje/etiología , NADPH Oxidasas/antagonistas & inhibidores , Acetofenonas/administración & dosificación , Animales , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/enzimología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/enzimología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Memoria/efectos de los fármacos , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Ratas
14.
Chin Med J (Engl) ; 125(11): 2019-24, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22884071

RESUMEN

BACKGROUND: Activation of c-Jun NH(2)-terminal kinase (JNK) has been implicated in neuron apoptosis as well as autophagy in response to various stressors after traumatic brain injury (TBI). However, the underlying molecular pathway remains unclear. Our study assessed whether JNK-mediated p53 phosphorylation might be an important mechanism for enhancing neuron autophagy in response to TBI. METHODS: A total of 186 male Sprague-Dawley (SD) rats (300 - 350 g) were used in this study. By randomized block method rats were randomly divided into four groups: sham-operated (n = 46), TBI (n = 60), TBI + dimethyl sulfoxide (DMSO) (n = 40), and TBI + SP600125 (n = 40). JNK was treated with SP600125, a specific JNK inhibitor. JNK, p-P53, Beclin-1, damage-regulated autophagy modulator (DRAM) and p-bcl-2 were evaluated by Western blotting analysis. The cellular localization and expression of Beclin-1 and DRAM was observed by immunofluorescence and immunohistochemistry, and the expression of Beclin-1-Bcl-2/Bcl-xL complexes was evaluated by immunoprecipitation. Multiple-group comparisons were conducted using analysis of variance (ANOVA). P values of less than 0.05 were considered statistically significant. RESULTS: It was observed that the expression of JNK, p-P53, Beclin-1, DRAM and p-bcl-2 was increasing after TBI, and the expression of Beclin-1 and DRAM was mainly located in the cytoplasm of neurons. But these were significantly inhibited in SP600125 group compared with sham group and TBI + SP600125 group (P < 0.05). The expression of Beclin-1-Bcl-2/Bcl-xL complexes was reduced after TBI. CONCLUSION: JNK-mediated p53 phosphorylation might be an important mechanism for enhancing neuron autophagy in response to TBI.


Asunto(s)
Lesiones Encefálicas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Beclina-1 , Western Blotting , Técnica del Anticuerpo Fluorescente , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Microscopía Fluorescente , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína bcl-X/metabolismo
15.
Zhonghua Wai Ke Za Zhi ; 50(2): 166-70, 2012 Feb 01.
Artículo en Chino | MEDLINE | ID: mdl-22490359

RESUMEN

OBJECTIVE: To study the effect and potential mechanism of expression of c-jun N-terminal kinase (JNK) signal pathway on neuron autophagy after diffuse brain injury (DBI). METHODS: Male Sprague Dawley rats (n = 216) were randomly divided into four groups: DBI group (n = 54), SP600125 intervene group (n = 54), DMSO group (n = 54) and sham operation group (n = 54). DBI rat model was established according to the description of Marmarou DBI. At different time points (1, 6, 12, 24, 48 and 72 h) after operation, the histopathologic changes of neurons in cortex were observed by HE staining method; The expression of p-JNK, p-P53, DRAM and Beclin-1 were detected by Western blot and immunohistochemistry. RESULTS: The results showed that under light microscope degenerated and necrotic neurons were observed to be scattered in cortex at 6 h after operation in DBI group, but these changes were low in SP600125 intervene group. Compared with SP600125 intervene group, the expression of p-JNK in DBI group were enhanced obviously at 6, 12 and 24 h (F = 17.902, P < 0.05); the expression of p-P53 in DBI group were enhanced obviously at 12, 24, 48 and 72 h (F = 7.107, P < 0.05); the expression of DRAM in DBI group were enhanced obviously at 6, 12, 24, 48 and 72 h (F = 15.455, P < 0.05); the expression of Beclin-1 in DBI group were enhanced obviously at 6, 12, 24, 48 and 72 h (F = 11.517, P < 0.05). Compared with DBI group, the expression of p-JNK, p-P53, DRAM and Beclin-1 in DMSO group were similar at 1, 6, 12, 24, 48 and 72 h (F = 1.509, P > 0.05). CONCLUSIONS: The present results indicate that SP600125 can dramatically improve trauma brain injury from autophagy after DBI and the molecular mechanism is related to the modulation of JNK signal pathway following DBI, while it measures the neuron autophagy by means of intervening JNK signal pathway.


Asunto(s)
Autofagia , Lesiones Encefálicas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/patología , Animales , Antracenos/farmacología , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley
16.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 22(4): 230-3, 2010 Apr.
Artículo en Chino | MEDLINE | ID: mdl-20398468

RESUMEN

OBJECTIVE: To study the protective effect of edaravone on severe traumatic brain injury (TBI) and its potential mechanism. METHODS: Two hundred and seventy-three male Sprague-Dawley (SD) rats were divided randomly into four groups: control group (n=45), model group (n=88), low-dose edaravone treatment group (n=72), high-dose edaravone treatment group (n=68). TBI rat model was reproduced by weight-dropping injury. One, 6, 24, 48 and 72 hours after injury, changes in brain tissue were observed with light and electron microscopy. The expression of phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2) was determined by Western blotting. The rate of neuron apoptosis was observed with immunohistochemistry and terminal-deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) method. Learning and memory function assessments were performed with Morris water maze from 7th day to 10th day after injury. RESULTS: Compared with control group, a part of neurons in hippocampus displayed histopathologic changes denoting necrosis 6, 24, 48 and 72 hours after injury. The p-ERK1/2 expression level (pg/unit) increased 1, 6, 24, 48 hours after injury (2.05 + or - 0.40, 4.40 + or - 0.96, 6.70 + or - 0.87, 3.67 + or - 0.28 vs. 0.40 + or - 0.04, 0.41 + or - 0.05, 0.43 + or - 0.06, 0.40 + or - 0.03), and the number of apoptotic cells increased 6, 24, 48, 72 hours after injury (9.60 + or - 2.69, 12.68 + or - 2.99, 16.94 + or - 3.92, 25.82 + or - 4.61 vs. 2.42 + or - 0.38, 2.58 + or - 0.57, 2.74 + or - 0.56, 2.61 + or - 0.58); latent period to find the safety platform (s) was significantly prolonged (119.8 + or - 25.0, 105.6 + or - 24.5, 98.5 + or - 21.8, 92.0 + or - 19.5 vs. 49.5 + or - 7.5, 32.7 + or - 6.3, 25.8 + or - 6.5, 24.8 + or - 5.5, all P<0.05). After treatment with edaravone, the degree of morphological injury, p-ERK1/2 level and number of apoptotic neurons decreased, latent period to find the safety platform was significantly shortened (in low-dose edaravone treatment group, p-ERK1/2 expression level at 6, 24, 48 hours was 2.46 + or - 0.22, 4.00 + or - 0.84, 2.38 + or - 0.32, and in high-dose edaravone treatment group was 1.67 + or - 0.15, 1.86 + or - 0.38, 1.27 + or - 0.28; in low-dose edaravone treatment group, the apoptotic cells at 6, 24, 48, 72 hours was 5.20 + or - 1.23, 7.10 + or - 1.72, 9.54 + or - 1.36, 14.12 + or - 3.19, and in high-dose edaravone treatment group was 3.40 + or - 0.49 , 4.39 + or - 0.73, 5.02 + or - 1.12, 8.78 + or - 2.16; in low-dose edaravone treatment group, latent period to find the safety platform at 7-10 days was 94.8 + or - 22.8, 65.2 + or - 19.0, 62.0 + or - 16.7, 59.5 + or - 15.6, and in high-dose edaravone treatment group it was 81.5 + or - 20.7, 55.4 + or - 18.5, 40.0 + or - 12.3, 32.2 + or - 11.0, all P<0.05). High-dose edaravone showed a better effect (all P<0.05). CONCLUSION: Edaravone gives good therapeutic effect on severe TBI, and the molecular mechanism is related to attenuation of ERK1/2 pathway and neuronal apoptosis following severe brain trauma.


Asunto(s)
Antipirina/análogos & derivados , Lesiones Encefálicas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Animales , Antipirina/farmacología , Apoptosis/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Edaravona , Masculino , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
17.
Artículo en Chino | MEDLINE | ID: mdl-17034719

RESUMEN

OBJECTIVE: To explore the effect of N-acetyl L-cysteine (NAC) on expressions of matrix metalloproteinases-2, 9 (MMP-2, MMP-9) in lung fibroblasts of SiO(2) exposed rats. METHODS: Seventy-five Wistar rats were divided randomly into three groups. The control group was administered with normal Saline. The model group and the interventional group were administered with SiO(2), and the interventional group was administered with NAC before SiO(2) was administered. Lung fibroblasts were isolated on day 1, 3, 7, 14, 28 after exposure to SiO(2). The expressions of MMP-2 and MMP-9 were evaluated by Immunocytochemistry and RT-PCR. RESULTS: (1) The expressions of protein and mRNA of MMP-2 in the model group were higher than that in the control group on all days after exposure to SiO(2) (P < 0.01). The expression of protein of MMP-9 was higher than the control group on day 1, 3, 7, and mRNA was higher on day 1, 3 (P < 0.01). (2) The expression of protein of MMP-2 in the interventional group was lower than the model group on all days, higher than the control group on day 3, 7, 14, 28, and the expression of mRNA was higher than the control group, lower than the model group, on all days (P < 0.05 or P < 0.01). The expression of protein of MMP-9 in the interventional group was lower than the model group on day 1, 3, 7, but higher than the control group on day 3, 7, and mRNA was lower than the model group on days 1, 3, higher than the control group (P < 0.05). CONCLUSION: NAC inhibits the expressions of MMP-2, MMP-9 in lung fibroblasts.


Asunto(s)
Acetilcisteína/farmacología , Fibroblastos/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Dióxido de Silicio/toxicidad , Animales , Células Cultivadas , Fibroblastos/efectos de los fármacos , Pulmón/citología , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , ARN Mensajero/genética , Distribución Aleatoria , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...