Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 14: 1193006, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37305546

RESUMO

Background: A high-fat Western diet is a risk factor for obesity and steatosis. Reducing intestinal absorption of a high-fat diet (HFD) is a feasible strategy to control obesity. Sulfosuccinimidyl oleate (SSO) inhibits intestinal fatty acid transport. Therefore, the aim of this study was to investigate the effects of SSO on HFD-induced glucose and lipid metabolism in mice and its possible underlying mechanisms. Methods: Male C57/BL were fed a HFD (60% calories) for 12 weeks and were administered an oral dose of SSO (50 mg/kg/day). The expression of lipid absorption genes (CD36, MTTP, and DGAT1) and the serum levels of triglycerides (TGs), total cholesterol (TC), and free fatty acids (FFAs) were detected. Lipid distribution in the liver was detected by oil red and hematoxylin and eosin staining. In addition, serum levels of inflammatory factors, alanine aminotransferase (ALT), and aspartate aminotransferase (AST) were measured to detect side effects. Results: SSO was effective in the treatment of obesity and metabolic syndrome induced by HFD in mice. It attenuated the assembly of intestinal epithelial chylomicrons by inhibiting intestinal epithelial transport and absorption of fatty acids, thereby reducing the gene expression levels of MTTP and DGAT1, resulting in decreased plasma TG and FFA levels. At the same time, it inhibited the transport of fatty acids in the liver and improved the steatosis induced by a HFD. The results of oil red staining showed that SSO treatment can reduce lipid accumulation in the liver by 70%, with no drug-induced liver injury detected on the basis of interleukin-6, C-reactive protein, ALT, and AST levels. In addition, SSO treatment significantly improved insulin resistance, decreased fasting blood glucose levels, and improved glucose tolerance in HFD-fed mice. Conclusion: SSO is effective in the treatment of obesity and metabolic syndrome induced by a HFD in mice. SSO reduces intestinal fatty acid absorption by reducing the inhibition of intestinal CD36 expression, followed by decreased TG and FFA levels, which attenuates HFD-induced fatty liver.

2.
Cell Mol Neurobiol ; 41(8): 1759-1772, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32889668

RESUMO

Spontaneous intracerebral hemorrhage (ICH) is a clinical challenge with high disability and lacks an effective treatment. miR-29a strongly expressed in the brain has been implicated in various neurological disorders. In this study, we investigated the biological roles of miR-29a in axonal outgrowth and neurological outcomes after ICH and relevant molecular mechanism. The rat model of ICH was established by injection of autologous whole blood into the right basal ganglia. First, a significant decrease in miR-29a level was found in perihematomal brain tissues and cerebrospinal fluid (CSF) after ICH in vivo and hemin-treated neurons in vitro. Further study documented that lentivirus-mediated miR-29a overexpression could remarkably attenuate hemorrhagic brain injury, promoted regenerative outgrowth of injured axons and improved neurobehavioral and cognitive impairments after ICH in rats. In addition, we also identified that overexpression of miR-29a obviously alleviated neuronal damage and mitochondrial dysfunctions, and facilitated neurite outgrowth in cultured neurons exposed to hemin in vitro. Furthermore, luciferase reporter assay showed that miR-29a directly targeted the 3'-UTR region of phosphatase and tensin homolog (PTEN) mRNA and negatively regulated its expression. More importantly, pharmacological inhibition of PTEN has similar neuroprotective effects as miR-29a overexpression involving activation of the PI3K/Akt pathway after hemorrhagic stroke. Collectively, these results suggested that elevated miR-29a could contribute to axonal outgrowth and neurological recovery through targeting PTEN/PI3K/Akt pathway after ICH, thereby providing a potential therapeutic target for patients with ICH.


Assuntos
Hemorragia Cerebral/metabolismo , MicroRNAs/biossíntese , Crescimento Neuronal/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Axônios/metabolismo , Axônios/patologia , Células Cultivadas , Hemorragia Cerebral/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Transdução de Sinais/fisiologia
3.
Biochem Biophys Res Commun ; 527(1): 22-28, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32446371

RESUMO

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.


Assuntos
Hemorragia Cerebral/complicações , Hemorragia Cerebral/tratamento farmacológico , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Neurônios/efeitos dos fármacos , Animais , Células Cultivadas , Hemorragia Cerebral/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Modelos Animais de Doenças , Ligantes , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Recuperação de Função Fisiológica
4.
Cancer Res ; 80(11): 2175-2189, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32066565

RESUMO

Esophageal squamous cell carcinoma (ESCC) is one of the deadliest malignant diseases. Multiple studies with large clinic-based cohorts have revealed that variations of phospholipase C epsilon 1 (PLCE1) correlate with esophageal cancer susceptibility. However, the causative role of PLCE1 in ESCC has remained elusive. Here, we observed that hypomethylation-mediated upregulation of PLCE1 expression was implicated in esophageal carcinogenesis and poor prognosis in ESCC cohorts. PLCE1 inhibited cell autophagy and suppressed the protein expression of p53 and various p53-targeted genes in ESCC. Moreover, PLCE1 decreased the half-life of p53 and promoted p53 ubiquitination, whereas it increased the half-life of mouse double minute 2 homolog (MDM2) and inhibited its ubiquitination, leading to MDM2 stabilization. Mechanistically, the function of PLCE1 correlated with its direct binding to both p53 and MDM2, which promoted MDM2-dependent ubiquitination of p53 and subsequent degradation in vitro. Consequently, knockdown of PLCE1 combined with transfection of a recombinant adenoviral vector encoding wild-type p53 resulted in significantly increased levels of autophagy and apoptosis of esophageal cancer in vivo. Clinically, the upregulation of PLCE1 and mutant p53 protein predicted poor overall survival of patients with ESCC, and PLCE1 was positively correlated with p53 in ESCC cohorts. Collectively, this work identified an essential role for PLCE1- and MDM2-mediated ubiquitination and degradation of p53 in inhibiting ESCC autophagy and indicates that targeting the PLCE1-MDM2-p53 axis may provide a novel therapeutic approach for ESCC. SIGNIFICANCE: These findings identify hypomethylation-mediated activation of PLCE1 as a potential oncogene that blocks cellular autophagy of esophageal carcinoma by facilitating the MDM2-dependent ubiquitination of p53 and subsequent degradation. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/11/2175/F1.large.jpg.


Assuntos
Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , Fosfoinositídeo Fosfolipase C/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Autofagia/fisiologia , Carcinogênese , Linhagem Celular Tumoral , Metilação de DNA , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfoinositídeo Fosfolipase C/genética , Regiões Promotoras Genéticas , Estabilidade Proteica , Ubiquitinação , Regulação para Cima
5.
Folia Neuropathol ; 58(4): 317-323, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33480236

RESUMO

INTRODUCTION: This study investigated the protective effects of antcin C against cerebral haemorrhage injury. MATERIAL AND METHODS: Cerebral haemorrhage was treated with antcin C 100 mg/kg i.p. at 60 min after the induction of cerebral injury. Neurological scores and volumes of cerebral injury were assessed to determine the effects of antcin C, based on oxidative stress and serum mediators of inflammation by ELISA. qRT-PCR was used to estimate the mRNA expression of Toll-like receptor 4 (TLR-4) and interleukin-1 receptor-associated kinase 4 (IRAK4) proteins in the cerebral tissue of rats with cerebral haemorrhage. Western blot assay and histopathology were also performed. RESULTS: The findings suggest that treatment with antcin C reduced the neurological scores and volumes of cerebral injury in cerebral injured rats. Parameters of oxidative stress and cytokine levels were reduced in the serum of the antcin C-treated group compared with the negative control group. Treatment with antcin C ameliorated the expression of TLR-4, IRAK4, and zonula occludens-1 (ZO-1) proteins in the cerebral tissue of cerebral injured rats. CONCLUSIONS: The results revealed that treatment with antcin C protected against cerebral haemorrhage damage by controlling microglia inflammation through the TLR-4 pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Hemorragia Cerebral/patologia , Neurônios/efeitos dos fármacos , Extratos Vegetais/farmacologia , Polyporales , Receptor 4 Toll-Like/efeitos dos fármacos , Animais , Inflamação/patologia , Masculino , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo
6.
Med Sci Monit ; 24: 1588-1596, 2018 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-29550832

RESUMO

BACKGROUND This study aimed to investigate the potential neuroprotective effect of recombinant osteopontin (r-OPN) on apoptotic changes via modulating phosphoinositide-3-kinase/Akt/glycogen synthase kinase 3 beta (PI3K/Akt/GSK-3ß) signaling in a rat model of intracerebral hemorrhage (ICH). MATERIAL AND METHODS We subjected 10-12-week-old Sprague-Dawley male rats (n=120) to injection of autologous blood into the right basal ganglia to induce ICH or sham surgery. ICH animals received vehicle administration, r-OPN (4 µL/pup), or r-OPN combined with phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin (86 ng/pup) at 30 min after injury. Neurological scores and rotarod latencies were evaluated on days 1-5 post-ICH. Brain water content was evaluated on days 1-3 post-ICH. The number of apoptotic cells changes were evaluated by terminal deoxynucleotidyl transferase-mediated 2-deoxyuridine 5-triphosphate-biotin nick-end labeling (TUNEL) and hematoxylin staining. Apoptosis-related proteins Bcl-2, Bax, and cleaved caspase-3 (CC3), and the phosphorylation levels of Akt and GSK-3b were assayed by Western blot. RESULTS Neurological deficits, rotarod latencies, and brain water content following ICH were reduced in the r-OPN group compared to the vehicle group. r-OPN also attenuated cell death in ICH. Furthermore, treatment with r-OPN significantly increased p-Akt expression and decreased p-GSK-3ß. These effects were associated with a decrease in the Bax/Bcl-2 ratio and the suppression of CC3 at 24 h after ICH. Importantly, all the beneficial effects of r-OPN in ICH were abrogated by the PI3K inhibitor wortmannin. CONCLUSIONS r-OPN may provide a wide range of neuroprotection by suppressing apoptosis through the PI3K/Akt/GSK-3ß signaling pathway after ICH.


Assuntos
Apoptose , Hemorragia Cerebral/tratamento farmacológico , Glicogênio Sintase Quinase 3 beta/metabolismo , Osteopontina/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Recombinantes/uso terapêutico , Recuperação de Função Fisiológica , Animais , Apoptose/efeitos dos fármacos , Encéfalo/patologia , Caspase 3/metabolismo , Hemorragia Cerebral/patologia , Hemorragia Cerebral/fisiopatologia , Regulação para Baixo/efeitos dos fármacos , Edema/tratamento farmacológico , Edema/patologia , Edema/fisiopatologia , Masculino , Camundongos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Osteopontina/administração & dosagem , Osteopontina/farmacologia , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Recuperação de Função Fisiológica/efeitos dos fármacos , Água , Proteína X Associada a bcl-2/metabolismo
7.
Mol Cell Biochem ; 442(1-2): 111-127, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29238898

RESUMO

Phospholipase C epsilon 1 (PLCE1) has been recognized as a novel susceptibility marker for esophageal squamous cell carcinoma (ESCC). The purpose of our study is to investigate its effect on the regulation of miRNA expression so as to translating the data into a novel strategy in control of ESCC. In this study, PLCE1 siRNA and vector-only plasmid were stably transfected into Eca109 and EC9706 cells and then subjected to miRNA array analysis, and quantitative real-time PCR was applied to validate miRNA array data. Then bioinformatic analyses, such as GO and pathway software, were conducted to obtain data on these differentially expressed miRNAs-targeted genes (DEGs) and clarify their function and pathway. The results showed that 36 miRNAs were found to be differentially expressed in PLCE1 siRNA-transfected cells compared with the control cells. In particular, 28 miRNAs were upregulated while 8 miRNAs were downregulated. Gene Ontology analysis showed that the function of the DEGs included cell cycle arrest, cell-matrix adhesion, apoptosis, etc. After this, the major pathways associated with the DEGs were regulation of actin cytoskeleton, TGF-beta signaling pathway, Notch signaling pathway and so on. Taken together, these results showed that the knockdown of PLCE1 may play a vital role in the control of ESCC. Further investigation will reveal and verify the function and pathway of the DEGs for the development of novel treatment strategy for the better control of ESCC.


Assuntos
Carcinoma de Células Escamosas , Biologia Computacional , Neoplasias Esofágicas , Técnicas de Silenciamento de Genes , MicroRNAs , Proteínas de Neoplasias , Fosfoinositídeo Fosfolipase C , RNA Neoplásico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Pontos de Checagem do Ciclo Celular/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Humanos , MicroRNAs/biossíntese , MicroRNAs/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Fosfoinositídeo Fosfolipase C/biossíntese , Fosfoinositídeo Fosfolipase C/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética
8.
Int J Mol Med ; 41(2): 659-668, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29207055

RESUMO

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.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Fibrose Pulmonar/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta1/genética , Células A549 , Actinas , Células da Medula Óssea/química , Células da Medula Óssea/metabolismo , Caderinas/genética , Meios de Cultivo Condicionados/química , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Isoquinolinas/farmacologia , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/metabolismo , Fibrose Pulmonar/metabolismo , Piridinas/farmacologia , Pirróis/farmacologia , Proteína Smad3/antagonistas & inibidores
9.
Int J Clin Exp Pathol ; 11(1): 427-437, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31938128

RESUMO

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.

10.
Cell Physiol Biochem ; 42(5): 1888-1896, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28772270

RESUMO

BACKGROUND/AIMS: Traumatic brain injury (TBI) is a major public health problem in the world and causes high rates of mortality and disability. Recent evidence suggests that vitamin D (VD) has neuroprotective actions and can promote function recovery after TBI. In vitro and in vivo studies have demonstrated that autophagy could be enhanced following supplementation with an active metabolite of VD (calcitriol). However, it is unclear whether autophagy participates in the protective effects of calcitriol after TBI. To test this hypothesis, we examined the protective effects of calcitriol on TBI-induced neurological impairment and further investigated whether calcitriol could modulate autophagy dysfunction-mediated cell death in the cortex region of rat brain. METHODS: Eighty-five male rats (250-280 g) were randomly assigned to sham (n=15), TBI model (TBI, n=35) and calcitriol treatment (calcitriol, n=35) groups. Rats were injected intraperitoneally with calcitriol (1 µg/kg) at 30 min, 24 h and 48 h post-TBI in the calcitriol group. The lysosomal inhibitor, chloroquine (CQ), was used to evaluate autophagic flux in the TBI and calcitriol groups. Neurological functions were evaluated via the modified neurological severity score test at 1-7 days after TBI or sham operation, and the terminal deoxynucleotidyl transferase-mediated FITC-dUTP nick-end labeling method was used to evaluate the ability of calcitriol to inhibit apoptosis. The expression of VDR, LC3 and p62 proteins was measured by western blot analysis at 1, 3 and 7 days post-injury Results: Calcitriol treatment attenuated mNSS at 2-7 days post-TBI (P < 0.05 versus TBI group). Calcitriol dramatically increased VDR protein expression compared with the untreated counterparts at 1, 3 and 7 days post-TBI (P < 0.05). The rate of apoptotic cells in calcitriol-treated rats was significantly reduced compared to that observed in the TBI group (P < 0.05). The LC3II/LC3I ratio was decreased in the cortex region at 1, 3 and 7 days post-TBI in rats treated with calcitriol (p < 0.05 versus TBI group), and the p62 expression was also attenuated (p < 0.05 versus TBI group). The LC3II/LC3I ratio in the calcitriol group was significantly increased when pretreated with CQ (P < 0.05). CONCLUSION: Calcitriol treatment activated VDR protein expression and attenuated neurological deficits in this rat TBI model. The protective effects might be associated with the restoration of autophagy flux and the decrease in apoptosis in the cortex region of rat brain.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Lesões Encefálicas Traumáticas/patologia , Calcitriol/farmacologia , Receptores de Calcitriol/metabolismo , Animais , Lesões Encefálicas Traumáticas/metabolismo , Cloroquina/toxicidade , Modelos Animais de Doenças , Injeções Intraperitoneais , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fármacos Neuroprotetores/farmacologia , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Ratos , Receptores de Calcitriol/agonistas
11.
Cell Physiol Biochem ; 42(1): 137-144, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28505619

RESUMO

BACKGROUND/AIMS: Intracerebral hemorrhage (ICH) occurs in hypertensive patients and results in high rates of mortality and disability. This study determined whether bone marrow mesenchymal stem cell (BMSC) transplantation affects axonal regeneration and examined the underlying mechanisms after the administration of PD98059 (p-ERK1/2 inhibitor) or/ and LY294002 (PI3K inhibitor). The hypothesis that was intended to be tested was that BMSC transplantation regulates the expression of growth-associated protein-43 (GAP-43) via the ERK1/2 and PI3K/Akt signaling pathways. METHODS: Seventy-five male rats (250-280 g) were subjected to intracerebral blood injection and then randomly received a vehicle, BMSCs, PD98059 or LY294002 treatment. Neurological deficits were evaluated prior to injury and at 1, 3 and 7 days post-injury. The expression of GAP-43, Akt, p-Akt, ERK1/2, and p-ERK1/2 proteins was measured by western blot analysis. RESULTS: BMSC transplantation attenuated neurological deficits 3-7 days post-ICH. The expression of GAP-43 was increased 3 days following BMSC transplantation. However, this increase was inhibited by either PD98059 or LY294002 treatment. Treatment with both PD98059 and LY294002 was more effective than was treatment with an individual compound. CONCLUSION: BMSC transplantation could attenuate neurological deficits and activate axonal regeneration in this rat ICH model. The protective effects might be associated with increased GAP-43 expression by activating both the ERK1/2 and PI3K/Akt signaling pathways.


Assuntos
Hemorragia Cerebral/terapia , Proteína GAP-43/metabolismo , Transplante de Células-Tronco Mesenquimais , Transdução de Sinais , Animais , Axônios/fisiologia , Células da Medula Óssea/citologia , Células Cultivadas , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/patologia , Cromonas/farmacologia , Modelos Animais de Doenças , Flavonoides/farmacologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Regeneração , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
12.
Mol Med Rep ; 15(4): 2374-2382, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28260000

RESUMO

Intracerebral hemorrhage (ICH) is a life-threatening type of stroke. Previous studies have reported that bone marrow mesenchymal stem cells (BMSCs) may exert beneficial effects on the treatment of ICH. However, it remains unknown whether the neuroprotection exerted by BMSCs on ICH is due to the differentiation of BMSCs, or the trophic factors secreted into their conditioned medium (CM). In addition, growth­associated protein­43 (GAP­43) is a protein associated with neurite extension, which may be considered a prospective therapeutic target in the treatment of ICH. The present study investigated whether administration of BMSC­CM could be considered as an alternative to the established treatment of direct BMSC transplantation; in addition, the underlying mechanisms were evaluated. Neurological function tests, brain water content, reverse transcription ­quantitative polymerase chain reaction and western blotting were used in present study. The current study indicated that the neuroprotective effects of BMSC implantation and BMSC-CM treatment are similar, and that both decrease the severity of post­ICH cerebral edema, as well as improving neurological functions. At the molecular level, treatment with BMSC­CM resulted in a marked elevation in the expression of GAP­43 and interleukin (IL)­10, in addition to a significant reduction in the expression levels of IL­1ß, tumor necrosis factor­α and IL­6. Following application of a phosphorylated­extracellular signal­regulated kinase (ERK1/2) inhibitor, PD98059, in a BMSC­CM rat model, the mRNA and protein expression levels of GAP­43 were significantly attenuated. Therefore, the findings of the present study demonstrated that treatment with BMSC­CM may be an alternative to direct BMSC transplantation in a rat model of ICH. The mechanism underlying BMSC­CM­mediated neuroprotection may be associated with anti-inflammatory effects, as well as activation of GAP­43 transcription and expression through ERK­1/2 phosphorylation. Therefore, the ERK-1/2-GAP-43 signaling pathway may be considered a potential novel application target of BMSC­CM for the treatment of neurological diseases.


Assuntos
Hemorragia Cerebral/terapia , Meios de Cultivo Condicionados/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Neuroproteção , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Edema Encefálico/patologia , Edema Encefálico/fisiopatologia , Edema Encefálico/terapia , Células Cultivadas , Hemorragia Cerebral/patologia , Hemorragia Cerebral/fisiopatologia , Interleucinas/análise , Masculino , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/análise
13.
Exp Ther Med ; 11(6): 2577-2582, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27284351

RESUMO

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.

14.
Mol Med Rep ; 13(6): 5248-54, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27122047

RESUMO

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.


Assuntos
Autofagia/efeitos dos fármacos , Lesões Encefálicas Traumáticas/prevenção & controle , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Estilbenos/farmacologia , Sinapses/metabolismo , Animais , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Masculino , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Resveratrol , Sinapses/patologia
15.
Int J Mol Med ; 37(4): 921-30, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26936125

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Lesões Encefálicas Traumáticas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Fármacos Neuroprotetores/uso terapêutico , Estilbenos/uso terapêutico , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Autofagia/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Masculino , Memória/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , NF-kappa B/análise , NF-kappa B/imunologia , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/patologia , Ratos Sprague-Dawley , Resveratrol , Receptor 4 Toll-Like/análise , Receptor 4 Toll-Like/imunologia
16.
Oncol Rep ; 35(3): 1711-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26707131

RESUMO

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.


Assuntos
Antraquinonas/administração & dosagem , Proteínas Reguladoras de Apoptose/biossíntese , Ciclina D/biossíntese , Glioma/tratamento farmacológico , Lisina/análogos & derivados , Proteínas de Membrana/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Proteína X Associada a bcl-2/biossíntese , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Apoptose/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2 , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/patologia , Humanos , Lisina/administração & dosagem , Camundongos , Espécies Reativas de Oxigênio , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Mol Med Rep ; 12(2): 2323-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25872478

RESUMO

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.


Assuntos
Edema Encefálico/tratamento farmacológico , Lesões Encefálicas/tratamento farmacológico , Cloroquina/farmacologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Autofagia/efeitos dos fármacos , Biomarcadores/metabolismo , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Esquema de Medicação , Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/prevenção & controle , Injeções Intraperitoneais , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/biossíntese , Interleucina-1beta/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Índices de Gravidade do Trauma , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo
18.
Mol Med Rep ; 12(2): 2149-54, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25873133

RESUMO

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.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Fármacos Neuroprotetores/uso terapêutico , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Corantes de Rosanilina/uso terapêutico , Animais , Edema Encefálico/complicações , Edema Encefálico/tratamento farmacológico , Edema Encefálico/patologia , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Interleucina-1beta/análise , Proteína Quinase C/análise , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7/análise
19.
Neurol Sci ; 35(5): 695-700, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24277205

RESUMO

Traumatic brain injury (TBI) is a leading cause of mortality and disability in children and young adults worldwide. Neurologic impairment is caused by both immediate brain tissue disruption and post-injury cellular and molecular events that worsen the primary neurologic insult. The ß-lactam antibiotic ceftriaxone (CTX) has been reported to induce neuroprotection in animal models of diverse neurologic diseases via up-regulation of GLT-1. However, no studies have addressed the neuroprotective role of CTX in the setting of TBI, and whether the mechanism is involved in the modulation of neuronal autophagy remains totally unclear. The present study was designed to determine the hypothesis that administration of CTX could significantly enhance functional recovery in a rat model of TBI and whether CTX treatment could up-regulate GLT-1 expression and suppress post-TBI neuronal autophagy. The results demonstrated that daily treatment with CTX attenuated TBI-induced brain edema and cognitive function deficits in rats. GLT-1 is down-regulated following TBI and this phenomenon can be reversed by treatment of CTX. In addition, we also found that CTX significantly reduced autophagy marker protein, LC3 II, in hippocampus compared to the TBI group. These results suggest that CTX might provide a new therapeutic strategy for TBI and this protection might be associated with up-regulation of GLT-1 and suppression of neuronal autophagy.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Ceftriaxona/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Edema Encefálico/patologia , Edema Encefálico/fisiopatologia , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Transportador 2 de Aminoácido Excitatório/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Regulação para Cima/efeitos dos fármacos
20.
Mol Med Rep ; 9(1): 77-82, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24220542

RESUMO

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.


Assuntos
Astrócitos/metabolismo , Autofagia , Conexina 43/metabolismo , Hipocampo/metabolismo , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Carbenoxolona/farmacologia , Carbenoxolona/uso terapêutico , Antagonistas dos Receptores H2 da Histamina/farmacologia , Antagonistas dos Receptores H2 da Histamina/uso terapêutico , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação , Biossíntese de Proteínas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...