Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cell Mol Neurobiol ; 41(8): 1759-1772, 2021 Nov.
Article in English | MEDLINE | ID: mdl-32889668

ABSTRACT

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.


Subject(s)
Cerebral Hemorrhage/metabolism , MicroRNAs/biosynthesis , Neuronal Outgrowth/physiology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Axons/metabolism , Axons/pathology , Cells, Cultured , Cerebral Hemorrhage/pathology , Male , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Signal Transduction/physiology
2.
Biochem Biophys Res Commun ; 527(1): 22-28, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446371

ABSTRACT

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.


Subject(s)
Cerebral Hemorrhage/complications , Cerebral Hemorrhage/drug therapy , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Neurons/drug effects , Animals , Cells, Cultured , Cerebral Hemorrhage/physiopathology , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Ligands , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Neurons/pathology , Recovery of Function
3.
Mol Cell Biochem ; 442(1-2): 111-127, 2018 May.
Article in English | MEDLINE | ID: mdl-29238898

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell , Computational Biology , Esophageal Neoplasms , Gene Knockdown Techniques , MicroRNAs , Neoplasm Proteins , Phosphoinositide Phospholipase C , RNA, Neoplasm , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle Checkpoints/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphoinositide Phospholipase C/biosynthesis , Phosphoinositide Phospholipase C/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics
4.
Med Sci Monit ; 24: 1588-1596, 2018 Mar 18.
Article in English | MEDLINE | ID: mdl-29550832

ABSTRACT

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.


Subject(s)
Apoptosis , Cerebral Hemorrhage/drug therapy , Glycogen Synthase Kinase 3 beta/metabolism , Osteopontin/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/therapeutic use , Recovery of Function , Animals , Apoptosis/drug effects , Brain/pathology , Caspase 3/metabolism , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Down-Regulation/drug effects , Edema/drug therapy , Edema/pathology , Edema/physiopathology , Male , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Osteopontin/administration & dosage , Osteopontin/pharmacology , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recovery of Function/drug effects , Water , bcl-2-Associated X Protein/metabolism
5.
Cell Physiol Biochem ; 42(1): 137-144, 2017.
Article in English | MEDLINE | ID: mdl-28505619

ABSTRACT

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.


Subject(s)
Cerebral Hemorrhage/therapy , GAP-43 Protein/metabolism , Mesenchymal Stem Cell Transplantation , Signal Transduction , Animals , Axons/physiology , Bone Marrow Cells/cytology , Cells, Cultured , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/pathology , Chromones/pharmacology , Disease Models, Animal , Flavonoids/pharmacology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Regeneration , Signal Transduction/drug effects , Up-Regulation/drug effects
6.
Cell Physiol Biochem ; 42(5): 1888-1896, 2017.
Article in English | MEDLINE | ID: mdl-28772270

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Brain Injuries, Traumatic/pathology , Calcitriol/pharmacology , Receptors, Calcitriol/metabolism , Animals , Brain Injuries, Traumatic/metabolism , Chloroquine/toxicity , Disease Models, Animal , Injections, Intraperitoneal , Male , Membrane Glycoproteins/metabolism , Microtubule-Associated Proteins/metabolism , Neuroprotective Agents/pharmacology , Nuclear Pore Complex Proteins/metabolism , Rats , Receptors, Calcitriol/agonists
7.
Neurol Sci ; 35(5): 695-700, 2014 May.
Article in English | MEDLINE | ID: mdl-24277205

ABSTRACT

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.


Subject(s)
Brain Injuries/drug therapy , Ceftriaxone/pharmacology , Neuroprotective Agents/pharmacology , Animals , Autophagy/drug effects , Autophagy/physiology , Brain Edema/drug therapy , Brain Edema/etiology , Brain Edema/pathology , Brain Edema/physiopathology , Brain Injuries/complications , Brain Injuries/pathology , Brain Injuries/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Disease Models, Animal , Excitatory Amino Acid Transporter 2/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Male , Maze Learning/drug effects , Maze Learning/physiology , Microtubule-Associated Proteins/metabolism , Random Allocation , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Up-Regulation/drug effects
8.
Front Pharmacol ; 14: 1193006, 2023.
Article in English | MEDLINE | ID: mdl-37305546

ABSTRACT

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.

9.
Zhonghua Wai Ke Za Zhi ; 50(2): 166-70, 2012 Feb 01.
Article in Zh | MEDLINE | ID: mdl-22490359

ABSTRACT

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.


Subject(s)
Autophagy , Brain Injuries/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/pathology , Animals , Anthracenes/pharmacology , Brain Injuries/pathology , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley
10.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 22(4): 230-3, 2010 Apr.
Article in Zh | MEDLINE | ID: mdl-20398468

ABSTRACT

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.


Subject(s)
Antipyrine/analogs & derivatives , Brain Injuries/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Antipyrine/pharmacology , Apoptosis/drug effects , Brain Injuries/drug therapy , Brain Injuries/pathology , Disease Models, Animal , Edaravone , Male , Neurons/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
11.
Folia Neuropathol ; 58(4): 317-323, 2020.
Article in English | MEDLINE | ID: mdl-33480236

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cerebral Hemorrhage/pathology , Neurons/drug effects , Plant Extracts/pharmacology , Polyporales , Toll-Like Receptor 4/drug effects , Animals , Inflammation/pathology , Male , Neurons/pathology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
12.
Cancer Res ; 80(11): 2175-2189, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32066565

ABSTRACT

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.


Subject(s)
Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Phosphoinositide Phospholipase C/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Autophagy/physiology , Carcinogenesis , Cell Line, Tumor , DNA Methylation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Gene Knockdown Techniques , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphoinositide Phospholipase C/genetics , Promoter Regions, Genetic , Protein Stability , Ubiquitination , Up-Regulation
13.
Int J Mol Med ; 41(2): 659-668, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207055

ABSTRACT

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.


Subject(s)
Culture Media, Conditioned/pharmacology , Pulmonary Fibrosis/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta1/genetics , A549 Cells , Actins , Bone Marrow Cells/chemistry , Bone Marrow Cells/metabolism , Cadherins/genetics , Culture Media, Conditioned/chemistry , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation/drug effects , Humans , Isoquinolines/pharmacology , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/metabolism , Pulmonary Fibrosis/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Smad3 Protein/antagonists & inhibitors
14.
Int J Clin Exp Pathol ; 11(1): 427-437, 2018.
Article in English | MEDLINE | ID: mdl-31938128

ABSTRACT

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.

15.
Mol Med Rep ; 15(4): 2374-2382, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28260000

ABSTRACT

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.


Subject(s)
Cerebral Hemorrhage/therapy , Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Neuroprotection , Animals , Brain/pathology , Brain/physiopathology , Brain Edema/pathology , Brain Edema/physiopathology , Brain Edema/therapy , Cells, Cultured , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Interleukins/analysis , Male , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/analysis
16.
Article in Zh | MEDLINE | ID: mdl-17034719

ABSTRACT

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.


Subject(s)
Acetylcysteine/pharmacology , Fibroblasts/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Silicon Dioxide/toxicity , Animals , Cells, Cultured , Fibroblasts/drug effects , Lung/cytology , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Wistar
17.
Exp Ther Med ; 11(6): 2577-2582, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27284351

ABSTRACT

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.

18.
Int J Mol Med ; 37(4): 921-30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26936125

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Injuries, Traumatic/drug therapy , Brain/drug effects , NF-kappa B/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Stilbenes/therapeutic use , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Autophagy/drug effects , Brain/immunology , Brain/pathology , Brain/physiopathology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Male , Memory/drug effects , Motor Activity/drug effects , NF-kappa B/analysis , NF-kappa B/immunology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Rats, Sprague-Dawley , Resveratrol , Toll-Like Receptor 4/analysis , Toll-Like Receptor 4/immunology
19.
Mol Med Rep ; 13(6): 5248-54, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27122047

ABSTRACT

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.


Subject(s)
Autophagy/drug effects , Brain Injuries, Traumatic/prevention & control , Neurons/metabolism , Neuroprotective Agents/pharmacology , Stilbenes/pharmacology , Synapses/metabolism , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Male , Neurons/pathology , Rats , Rats, Sprague-Dawley , Resveratrol , Synapses/pathology
20.
Oncol Rep ; 35(3): 1711-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26707131

ABSTRACT

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.


Subject(s)
Anthraquinones/administration & dosage , Apoptosis Regulatory Proteins/biosynthesis , Cyclin D/biosynthesis , Glioma/drug therapy , Lysine/analogs & derivatives , Membrane Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , bcl-2-Associated X Protein/biosynthesis , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/pathology , Humans , Lysine/administration & dosage , Mice , Reactive Oxygen Species , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL