Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Clin Transl Hepatol ; 11(1): 45-57, 2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36406315

ABSTRACT

Background and Aims: Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease caused by over-nutrition. Impaired autophagy is closely related to NAFLD progression. Recently, ubiquitin-specific peptidase-10 (USP10) was reported to ameliorate hepatic steatosis, but the underlying mechanism is still unclear. In view of the potential effects of USP10 on autophagy, we investigated whether USP10 alleviated steatosis through autophagy. Methods: HepG2 cells were treated with palmitic acid (PA) to model NAFLD in vitro. Lentivirus was used to regulate USP10 level in cells. Autophagic regulators were used to autophagic progression in cells. Western blotting, real-time fluorescence quantitative polymerase chain reaction, lipid drop staining and immunofluorescent staining were performed to determine the effect of USP10 on lipid autophagy. Student's t-test and Tukey's post hoc test were used to compare the means among groups. Results: PA induced cellular steatosis with dependance on autophagy. USP10 overexpression alleviated PA-induced steatosis, restored autophagic activity, promoted autophagic flux, including synthesis and degradation of autophagosomes, and lipid-targeted autophagy. In the presence of autophagy inhibitors, the protective effectiveness of USP10 on steatosis decreased. Furthermore, the specific inhibitor to C-jun N-terminal protein kinase-1 (JNK1), DB07268, abolished USP10-induced autophagy. However, during early stage inhibition of JNK1, compensatory expression of tuberous sclerosis complex-2 (TSC2) maintained autophagy. The degree of TSC2-to-JNK1 compensation was positively associated with USP10 level. Functionally, JNK1 and TSC2 were involved in the lipid-lowering effect of USP10. Conclusions: USP10 alleviated hepatocellular steatosis in autophagy-dependent manner. JNK1/TSC2 signaling pathways were required for USP10-induced autophagy.

2.
Acta Pharm Sin B ; 12(5): 2129-2149, 2022 May.
Article in English | MEDLINE | ID: mdl-35646540

ABSTRACT

Cardiometabolic disease (CMD), characterized with metabolic disorder triggered cardiovascular events, is a leading cause of death and disability. Metabolic disorders trigger chronic low-grade inflammation, and actually, a new concept of metaflammation has been proposed to define the state of metabolism connected with immunological adaptations. Amongst the continuously increased list of systemic metabolites in regulation of immune system, bile acids (BAs) represent a distinct class of metabolites implicated in the whole process of CMD development because of its multifaceted roles in shaping systemic immunometabolism. BAs can directly modulate the immune system by either boosting or inhibiting inflammatory responses via diverse mechanisms. Moreover, BAs are key determinants in maintaining the dynamic communication between the host and microbiota. Importantly, BAs via targeting Farnesoid X receptor (FXR) and diverse other nuclear receptors play key roles in regulating metabolic homeostasis of lipids, glucose, and amino acids. Moreover, BAs axis per se is susceptible to inflammatory and metabolic intervention, and thereby BAs axis may constitute a reciprocal regulatory loop in metaflammation. We thus propose that BAs axis represents a core coordinator in integrating systemic immunometabolism implicated in the process of CMD. We provide an updated summary and an intensive discussion about how BAs shape both the innate and adaptive immune system, and how BAs axis function as a core coordinator in integrating metabolic disorder to chronic inflammation in conditions of CMD.

3.
Toxicol Appl Pharmacol ; 374: 53-64, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31051157

ABSTRACT

Although numerous in vivo studies have suggested that alpha-ketoglutarate (AKG), i.e. the key intermediate in the Krebs cycle, may have an anabolic effect on bone tissue, the direct influence of AKG on osteoblasts and the underlying mechanism of its action have not been investigated so far. The aim of this study was to assess the impact of AKG (disodium salt dihydrate) on osteogenesis in vitro and identification of some signaling mechanisms involved in this activity. The human and mouse normal osteoblast cell lines hFOB 1.19 and MC3T3-E1 were used in this study. The results showed that AKG did not increase the proliferation of osteoblasts; however, it upregulated the expression of transcription factors RUNX2 and Osterix, the mRNA and protein levels of osteoblast differentiation markers (alkaline phosphatase, type I collagen, bone sialoprotein II, osteopontin, osteocalcin), and the mineralization levels in the hFOB 1.19 and MC3T3-E1 cell cultures. Moreover, AKG increased JNK, mTOR, S6K1, and S6 phosphorylation and decreased ERK1/2 phosphorylation in both osteoblast cell lines. The JNK inhibitor and rapamycin, but not the ERK inhibitor, abolished the AKG-promoted osteoblast differentiation. Using immunofluorescence staining, qRT-PCR, and Western blot analysis, we detected the presence of an AKG receptor GPR99 activated by alpha ketoglutaric acid in the tested osteoblast cell lines. However, AKG salt did not activate GPR99. Our findings suggest that AKG salt activates the JNK and mTOR/S6K1/S6 signaling pathways to promote differentiation of osteoblasts, independently of GPR99 activation. We can conclude that AKG salts might be promising candidates for bone anabolic drugs used for prevention or/and treatment of osteoporosis.


Subject(s)
Ketoglutaric Acids/pharmacology , Osteoblasts/drug effects , Osteogenesis/drug effects , 3T3 Cells , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation/drug effects , Humans , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sp7 Transcription Factor/genetics , Sp7 Transcription Factor/metabolism
4.
Ann Transl Med ; 7(1): 6, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30788353

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the protective effects of Momordica charantia polysaccharides (MCP) on depressive-like behaviors. METHODS: The chronic social defeat stress (CSDS) mice model was used to evaluate the effects of MCP and their underlying mechanisms. Social interaction test (SIT), sucrose preference test (SPT), and tail suspension test (TST) were performed for behavioral assessments. Expression levels of inflammation mediators and phosphatidylinositol 3-kinase (PI3K) activity were determined using commercial ELISA kits. The expression of key proteins in the c-Jun N-terminal protein kinase (JNK3)/PI3K/protein kinase B (AKT) pathway were measured using western blot and RT-PCR. RESULTS: The results showed that chronic administration of MCP (100, 200, 400 mg/kg/day) significantly prevented depressive-like behaviors in CSDS mice as assessed by SIT, TST and SPT. Elevated levels of proinflammatory cytokines [tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1ß)], and expression of JNK3, c-Jun, P-110ß proteins were observed in the hippocampus of CSDS mice. Moreover, the activity of PI3K and phosphorylation level of AKT were reduced in the hippocampus of CSDS mice. Interestingly, the administration of MCP reversed these changes. Furthermore, the protective effects of MCP on CSDS mice were partly inhibited by the PI3K inhibitor, LY294002. CONCLUSIONS: In conclusion, the protective effects of MCP against depressive-like behaviors in CSDS mice might be due to a reduction in neuroinflammation and the down-regulation of the JNK3/PI3K/AKT pathway in the hippocampus.

5.
Cell Biol Toxicol ; 34(4): 251-262, 2018 08.
Article in English | MEDLINE | ID: mdl-28540451

ABSTRACT

Kidney denervation prevents the development of tubulointerstitial fibrosis, but the neuropeptide calcitonin gene-related peptide (CGRP) in the denervated kidneys restores the fibrotic feature through the upregulation of profibrogenic growth factors. CGRP is involved in aggravation of inflammation by increasing the number of circulating cells and chemotactic factors. However, it is not clear how CGRP contributes to the upregulation of profibrogenic factors during fibrogenesis. In both human and pig kidney proximal tubular cell lines, administration of 1 nM CGRP significantly increased the levels of transforming growth factor-ß1 (TGF-ß1) production and connective tissue growth factor (CTGF) expression at 6 and 24 h after the administration. Exogenous CGRP also increased the TGF-ß1 and CTGF protein levels in the incubation media, indicating release of these proteins from the cells. Treatment with 100 nM CGRP receptor antagonist (CGRP8-37) for 24 h significantly inhibited the increase in intracellular levels and released levels of TGF-ß1 and CTGF in CGRP-treated cells. Genetic inhibition of CGRP receptor using siRNA transfection also suppressed the increase in TGF-ß1 production and release at 24 h after CGRP stimulation. Furthermore, treatment with a specific protein kinase C (PKC) inhibitor chelerythrine (1 thru 10 µM) markedly reduced the upregulation and release of TGF-ß1 and CTGF 6 h after CGRP administration. Finally, inhibition of c-Jun N-terminal protein kinase (JNK) phosphorylation using 1 µM SP600125 prevented the increase in TGF-ß1 and CTGF upregulation and release 6 h after CGRP administration. Consistent with the in vitro data, exogenous CGRP in denervated UUO kidneys upregulated and secreted TGF-ß1 and CTGF in dependence on PKC activation and JNK phosphorylation. In conclusion, these data suggest that exogenous CGRP induces the upregulation and secretion of profibrogenic TGF-ß1 and CTGF proteins through the CGRP receptor/PKC/JNK signaling pathway in kidney proximal tubular cells.


Subject(s)
Calcitonin Gene-Related Peptide/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , MAP Kinase Signaling System/drug effects , Protein Kinase C/metabolism , Up-Regulation/drug effects , Animals , Connective Tissue Growth Factor/metabolism , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibrosis , Humans , Kidney Tubules, Proximal/drug effects , LLC-PK1 Cells , Male , Mice, Inbred C57BL , Protein Kinase C/antagonists & inhibitors , Receptors, Calcitonin Gene-Related Peptide/metabolism , Swine , Transforming Growth Factor beta1/metabolism
6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-838304

ABSTRACT

Objective To investigate the protective effect of ginkgolide A and ginkgolide B (GKAB) mixture on neurons of rats with permanent middle cerebral artery occlusion (pMCAO) and related molecular mechanisms. Methods Sixty male Sprague-Dawley rats were randomly divided into sham group, pMCAO permanent focal cerebral ischemia group and GKAB-treated low-, medium- and high-dose groups. In addition to the sham group (only isolated without interruption of the arteries), the rats in the remaining four groups were induced pMCAO by blocking the right middle cerebral artery. Rats in the GKAB-treated low-, medium- and high-dose groups were injected with GKAB 12.5, 25, and 50 mg/kg through sublingual vein at 10 min after pMCAO, while the sham and pMCAO groups were injected with saline of the same volume as the medium-dose group. After 12 h of treatment, the neuronal apoptosis was determined by TUNEL method, the level of phosphorylated c-Jun N-terminal kinase (p-JNK) was determined by immunohistochemistry, the expressions of p-JNK, Bcl-2, Bax, cytochrome C (Cyt C), caspase-9, caspase-3, cleaved caspase-9, and cleaved caspase-3 in brain tissues were detected by Western blotting. Results Compared with the sham group, the apoptosis rate and p-JNK expression of neurons in the pMCAO group were significantly increased (P<0.01), and the expressions of apoptosis-related proteins Bax, cleaved caspase-9 and cleaved caspase-3 in brain tissues were significantly increased (P<0.01), while the expressions of Bcl-2, caspase-9 and caspase-3 in brain tissues were significantly decreased (P<0.01). Compared with the pMCAO group, the apoptosis rate and p-JNK expression of neurons in GKAB-treated low-, medium- and high-dose groups were significantly decreased (P<0.01), the expressions of Bax, cleaved caspase-9 and cleaved caspase-3 protein were significantly decreased (P<0.01), and the expressions of Bcl-2, caspase-9 and caspase-3 were significantly increased (P<0.01) in a dose-dependent manner. Compared with the sham group, the expression of Cyt C in cytoplasm in the pMCAO group was significantly increased, and the expression of mitochondrial Cyt C was significantly decreased (P<0.01). Compared with the pMCAO group, the expressions of Cyt C in cytoplasm in the GKAB-treated low-, medium- and high-dose groups were significantly decreased in a dose-dependent manner, and the expressions of mitochondrial Cyt C were significantly increased (P<0.05, P<0.01). Conclusion GKAB can inhibit neuronal apoptosis after pMCAO in rats, and its mechanism may be related to the inhibition of JNK phosphorylation and JNK signaling pathway and the block of mitochondrial apoptosis pathway.

7.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-513487

ABSTRACT

Objective To observe the effects of Yiqi Huoxue Tongluo Jiedu fang (YHTJF) on pneumocyte apoptosis after lung ischemia/reperfusion (I/R) injury (LIRI) in mice and to investigate whether c-Jun N-terminal protein kinase (JNK) is involved in the mechanism of apoptosis.Methods Seventy C57BL/6J male mice were randomly divided into seven groups:normal control group (C group),carboxyl methyl cellulose-Na+normal control group (CMC-Na+C group),CMC-Na+sham group (CMC-Na+S group),CMC-Na+I/R group (CMC-Na+I/R group) and CMC-Na+YHTJF-low,-middle,-high dose groups (CMC-Na+YL,CMC-Na+YM,CMC-Na+YH groups).C group did not undergo any processing;in CMC-Na+S group,only was chest opened without clipping the lung hilum;in the rest of the four groups,they all underwent opening of the chest and clipping the lung hilum for 30 minutes,then the clipping of artery was relieved and left lung reperfusion was carried out for 3 hours.After operation,the mice were sacrificed,the lung tissues were harvested.Under light and electron microscopes,the lung morphological and ultra-structural changes were observed,and the changes of index of quantitative evaluation for alveolar damage (IQA) were determined.The terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) was applied to evaluate the apoptosis index (AI) of the lung tissues.The protein and mRNA expressions of JNK and glucose regulating protein 78 (GRP78) in lung tissues were detected by Western Blot and reverse transcription-polymerase chain reaction (RT-PCR);the correlations between lung AI and the expressions of mRNA and protein of JNK and GRP78,IQA were analyzed.Results Compared with CMC-Na+S group,IQA,AI and mRNA and the protein expressions of JNK and GRP78 in CMC-Na+I/R group were obviously higher [IQA:(74.00 ± 7.31)% vs.(7.00 ± 1.23)%,AI:(64.40 ± 11.97)% vs.(5.60 ± 1.14)%,JNK mRNA (gray value):1.143 ± 0.284 vs.0.152 ± 0.128,GRP78 mRNA (gray value):0.897 ± 0.129 vs.0.284 ± 0.044,JNK protein (A value):0.428 ± 0.074 vs.0.073 ± 0.052,GRP78 protein (A value):1.075 ± 0.145 vs.0.589 ± 0.060].Compared with CMC-Na+I/R group,the IQA,AI,protein and mRNA expressions of JNK and GRP78 in CMC-Na+YL,CMC-Na+YM,CMC-Na+YH groups were all lower,and the degree of reduction in group CMC-Na+YM was the most remarkable,greater than that in CMC-Na+YL or CMC-Na+YH group [IQA:(26.20 ± 3.35)% vs.(34.00±5.34)%,(41.20±9.18)%,AI:(29.40±3.05)% vs.(48.20±3.83)%,(39.20±6.14)%,JNK mRNA (gray value):0.681 ± 0.130 vs.0.804 ± 0.153,0.938 ± 0.11,GRP78 mRNA (gray value):0.450 ± 0.105 vs.0.747 ± 0.231,0.566 ± 0.115,JNK protein (A value):0.188 ± 0.049 vs.0.261 ± 0.065,0.209 ± 0.063,all P < 0.01],compared with the CMC-Na+I/R group,the expression of GRP78 protein was obviously higher in CMC-Na+YH,CMC-Na+YL,CMC-Na+YM groups and the most remarkably high was in CMC-Na+YH group (A value:1.429 ±0.226 vs.1.130±0.169,1.128 ±0.177,all P < 0.01).The apoptosis of each group was mainly in the pulmonary vascular endothelial cells and alveolar epithelial cells,and brown particles were positive cells under light microscope.Under transmission electron microscope:nuclear pyknosis and margination under the nuclear membrane,cytoplasm condensed,lamellar bodies decreased and emptying increased,cell membrane microvilli decreased or disappeared,mitochondria swelling,inflammatory cells increased in alveolar septum and adhering onto the capillary walls could be seen in CMC-Na+I/R group.Compared with CMC-Na+I/R group,the lung tissue ultrastructural damage alleviated,ultrastructure of alveoli clearly seen,nuclear chromatin relatively uniform,cytoplasm increased,type Ⅱ alveolar epithelial cell surface microvilli relatively plenty,lamellar corpuscle number increased,mitochondria swelling ameliorated in CMC-Na+YH,CMC-Na+YL,CMC-Na+YM groups and the most remarkable one was CMC-Na+YM group.AI was significantly positive correlated with the mRNA and protein expressions of JNK,GRP78 and IQA (r =0.907,0.928,0.880,0.712,0.911,all P < 0.01).Conclusions YHTJF may effectively alleviate the cell apoptosis in mice LIRI,and its mechanism may be related to the inhibition of JNK pathway.

8.
Eur J Pharmacol ; 766: 114-21, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26435026

ABSTRACT

Chronic inflammation, imbalance between the extracellular matrix synthesis and degradation, and loss of vascular smooth muscle cells (SMCs) contribute to the development of abdominal aortic aneurysm (AAA). The purpose of this study was to investigate the effect of the therapy with periaortic incubation of c-Jun N-terminal protein kinase inhibitor SP600125 infused from an osmotic pump and subadventitial injection of lysyl oxidase (LOX) gene modified autologous smooth muscle progenitor cells (SPCs) on treatment of AAA in a rabbit model. Obvious dilation of the abdominal aorta in the control group was caused by periaortic incubation of calcium chloride and elastase. But the progression of aortic dilation was significantly decreased after the treatment with SP600125 and LOX gene modified SPCs compared to the treatment with phosphate-buffered saline. This therapy could inhibit matrix metalloproteinases expression, enhance elastin synthesis, improve preservation of elastic laminar integrity, benefit SPCs survival and restore SMCs population. It seemed that this method might provide a novel therapeutic strategy to treat AAA.


Subject(s)
Anthracenes/therapeutic use , Aortic Aneurysm, Abdominal/therapy , Protein Kinase Inhibitors/therapeutic use , Protein-Lysine 6-Oxidase/genetics , Stem Cell Transplantation , Animals , Anthracenes/pharmacology , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/physiopathology , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/physiopathology , Combined Modality Therapy , Humans , Leukocytes, Mononuclear , Male , Myocytes, Smooth Muscle/cytology , Protein Kinase Inhibitors/pharmacology , Protein-Lysine 6-Oxidase/metabolism , Rabbits , Stem Cells , Ultrasonography
9.
Oncol Lett ; 9(6): 2767-2770, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26137143

ABSTRACT

Receptor for activated C kinase 1 (RACK1) is up-regulated in hepatocellular carcinoma (HCC) and has been reported to augment c-Jun N-terminal protein kinase (JNK) activity in HCC SMMC-7721 cells. By contrast, activator protein-1, a downstream JNK transcription factor, has been revealed to mediate the overexpression of RACK1 in melanoma cells. Therefore, the association between RACK1 and JNK activity in HCC cells has yet to be completely elucidated. The present study analyzed the effects of RACK1 or JNK loss of function on the levels of RACK1 protein, JNK activity, cell proliferation and apoptosis induced by tumor necrosis factor-related apoptosis inducing ligand in HCC SMMC-7721 cells. It was found that JNK loss of function exhibited no effect on RACK1 expression, whereas a loss of RACK1 function led to reduced JNK activity in SMMC-7721 cells. RACK1 and JNK loss of function resulted in the impaired oncogenic growth of SMMC-7721 cells. The present data further support a pivotal role of RACK1 in mediating enhanced JNK activity in HCC cells and also indicate that a novel mechanism exists for RACK1 overexpression in HCC SMMC-7721 cells.

10.
Mol Immunol ; 58(2): 214-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24389043

ABSTRACT

INTRODUCTION: Mixed lineage kinase 3 (MLK3) is part of the intracellular regulatory system that connects extracellular cytokine or mitogen signals received through G-protein coupled receptors to changes in gene expression. MLK3 activation stimulates motility of epithelial cells and epithelial-derived tumor cells, but its role in mediating the migration of other cell types remains unknown. Since neutrophils play a crucial role in innate immunity and contribute to the pathogenesis of several diseases, we therefore examined whether MLK3 might regulate the motility of mouse neutrophils responding to a chemotactic stimulus, the model bacterial chemoattractant fMLP. METHODS: The expression of Mlk3 in mouse neutrophils was determined by immunocytochemistry and by RT-PCR. In vitro chemotaxis in a gradient of fMLP, fMLP-stimulated random motility, fMLP-stimulated F-actin formation were measured by direct microscopic observation using neutrophils pre-treated with a novel small molecule inhibitor of MLK3 (URMC099) or neutrophils obtained from Mlk3-/- mice. In vivo effects of MLK3 inhibition were measured by counting the fMLP-induced accumulation of neutrophils in the peritoneum following pre-treatment with URMC099 in wild-type C57Bl/6 or mutant Mlk3-/- mice. RESULTS: The expression of Mlk3 mRNA and protein was observed in neutrophils purified from wild-type C57Bl/6 mice but not in neutrophils from mutant Mlk3-/- mice. Chemotaxis by wild-type neutrophils induced by a gradient of fMLP was reduced by pre-treatment with URMC099. Neutrophils from C57Bl/6 mice pretreated with URMC099 and neutrophils from Mlk3-/- mice moved far less upon fMLP-stimulation and did not form F-actin as readily as untreated neutrophils from C57Bl/6 controls. In vivo recruitment of neutrophils into the peritoneum by fMLP was significantly reduced in wild-type mice treated with URMC099, as well as in untreated Mlk3-/- mice-thereby confirming the role of MLK3 in neutrophil migration. CONCLUSIONS: Mlk3 mRNA is expressed in murine neutrophils. Genetic or pharmacologic inhibition of MLK3 blocks fMLP-mediated motility of neutrophils both in vitro and in vivo, suggesting that MLK3 may be a therapeutic target in human diseases characterized by exuberant neutrophil migration.


Subject(s)
Chemotactic Factors/pharmacology , Immune System Diseases/chemically induced , Leukocyte Disorders/chemically induced , MAP Kinase Kinase Kinases/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Animals , Cell Migration Inhibition/drug effects , Cell Migration Inhibition/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Immune System Diseases/genetics , Leukocyte Disorders/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Activation/drug effects , Neutrophil Activation/genetics , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Mitogen-Activated Protein Kinase Kinase Kinase 11
11.
Brain Res ; 1544: 45-53, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24316243

ABSTRACT

N-hydroxy-N-(4-butyl-2-methylphenyl) formamidine (HET0016) is a specific 20-hydroxyeicosatetraenoic acid (20-HETE) inhibitor which was first synthesized in 2001. It has been demonstrated that HET0016 reduces cerebral infarction volume in rat middle cerebral artery occlusion (MCAO) models. However, little is known about the role of HET0016 in the blood-brain barrier (BBB) dysfunction after cerebral ischemia/reperfusion (I/R) injury. The present study was designed to examine the effect of HET0016 in a MCAO and reperfusion rat model to determine whether it protects against brain edema and BBB disruption. Rats were subjected to 90 min MCAO, followed by 4, 24, 48, and 72 h reperfusion. Brain edema was measured according to the wet and dry weight method. BBB permeability based on the extravasation of Evans blue and sodium fluorescein was detected. BBB ultrastructure alterations were presented through transmission electron microscope. Superoxide production in ischemic tissue was also measured by dihydroethidium fluorescent probe. Western blot was used to analyze the expression of Claudin-5, ZO-1, MMP-9, and JNK pathway. At 24h after reperfusion, HET0016 reduced brain edema and BBB leakage. Ultrastructural damage of BBB and the increase of superoxide production were attenuated by HET0016 treatment. Western blot showed that HET0016 suppressed the activation of MMP-9 and JNK pathway but restored the expression of Claudin-5 and ZO-1. In conclusion, these results suggest that HET0016 protects BBB dysfunction after I/R by regulating the expression of MMP-9 and tight junction proteins. Furthermore, inhibition of oxidative stress and JNK pathway may be involved in this protecting effect.


Subject(s)
Amidines/therapeutic use , Blood-Brain Barrier/drug effects , Brain Edema/drug therapy , Brain Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Reperfusion Injury/drug therapy , Animals , Blood-Brain Barrier/ultrastructure , Claudin-5/drug effects , Claudin-5/metabolism , MAP Kinase Signaling System/drug effects , Male , Matrix Metalloproteinase 9/drug effects , Rats , Rats, Sprague-Dawley , Zonula Occludens-1 Protein/drug effects , Zonula Occludens-1 Protein/metabolism
12.
Biochem Biophys Res Commun ; 441(1): 53-8, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24134840

ABSTRACT

Different groups including ours have shown that curcumin induces melanoma cell apoptosis, here we focused the role of mammalian Sterile 20-like kinase 1 (MST1) in it. We observed that curcumin activated MST1-dependent apoptosis in cultured melanoma cells. MST1 silencing by RNA interference (RNAi) suppressed curcumin-induced cell apoptosis, while MST1 over-expressing increased curcumin sensitivity. Meanwhile, curcumin induced reactive oxygen species (ROS) production in melanoma cells, and the ROS scavenger, N-acetyl-cysteine (NAC), almost blocked MST1 activation to suggest that ROS might be required for MST1 activation by curcumin. c-Jun N-terminal protein kinase (JNK) activation by curcumin was dependent on MST1, since MST1 inhibition by RNAi or NAC largely inhibited curcumin-induced JNK activation. Further, curcumin induced Foxo3 nuclear translocation and Bim-1 (Foxo3 target gene) expression in melanoma cells, such an effect by curcumin was inhibited by MST1 RNAi. In conclusion, we suggested that MST1 activation by curcumin mediates JNK activation, Foxo3a nuclear translocation and apoptosis in melanoma cells.


Subject(s)
Apoptosis/drug effects , Cell Nucleus/metabolism , Curcumin/pharmacology , Forkhead Transcription Factors/metabolism , Hepatocyte Growth Factor/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Melanoma/enzymology , Proto-Oncogene Proteins/metabolism , Acetylcysteine/pharmacology , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , Enzyme Activation/drug effects , Forkhead Box Protein O3 , Melanoma/pathology , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Protein Transport/drug effects , Reactive Oxygen Species/metabolism
13.
Environ Toxicol Pharmacol ; 36(2): 548-556, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23827194

ABSTRACT

Silver nanoparticles (AgNPs) have been widely used in commercial goods ranging from medical devices to home appliances. Their widespread application increase the risk related to their potential toxicity. Although several studies showed their acute hazardous effects on living animals, our understanding of chronic effects of AgNPs exposed by the environment we encounter in our everyday lives is still very limited. This is partly because of the lack of versatile animal model system for studying AgNPs effects on terrestrial animals including human. In this study, we used Drosophila model to study AgNPs toxicity in terrestrial animals, and found that long-term exposure of AgNPs, but not Ag ions, at low level (0.1 and 1µg/mL) significantly shortened the lifespan. By taking advantage of the power of Drosophila genetics, we also isolated a GAL4 enhancer trap line called M95, in which the expression of GAL4 is up-regulated in response to ingestion of AgNPs at concentrations as low as 0.1µg/mL. Interestingly M95 flies showed significantly increased tolerance to both AgNPs treatment and dry starvation probably due to up-regulation of JNK signaling. These findings suggest not only that M95 may be a very useful biomarker of AgNPs because of its high sensitivity and tolerance to AgNPs, but also that Drosophila may be a versatile terrestrial invertebrate model for studying the effects of AgNPs on human health.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila/drug effects , Metal Nanoparticles/toxicity , Saccharomyces cerevisiae Proteins/metabolism , Silver/toxicity , Toxicity Tests/methods , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Biomarkers/metabolism , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Drosophila/genetics , Drosophila/metabolism , Genes, Reporter , JNK Mitogen-Activated Protein Kinases/metabolism , Longevity/drug effects , MAP Kinase Signaling System/drug effects , Saccharomyces cerevisiae Proteins/genetics , Starvation/genetics , Starvation/metabolism , Transcription Factors/genetics , Transcription, Genetic/drug effects , Up-Regulation
14.
Biochim Biophys Acta ; 1833(12): 2856-2865, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23872419

ABSTRACT

Pancreatic cancer ranks fourth among cancer-related causes of death in North America. Minimal progress has been made in the diagnosis and treatment of patients with late-stage tumors. Moreover, pancreatic cancer aggressiveness is closely related to high levels of pro-survival mediators, which can ultimately lead to rapid disease progression, resistance and metastasis. The main goal of this study was to define the mechanisms by which calix[6]arene, but not other calixarenes, efficiently decreases the aggressiveness of a drug resistant human pancreas carcinoma cell line (Panc-1). Calix[6]arene was more potent in reducing Panc-1 cell viability than gemcitabine and 5-fluorouracil. In relation to the underlying mechanisms of cytotoxic effects, it led to cell cycle arrest in the G0/G1 phase through downregulation of PIM1, CDK2, CDK4 and retinoblastoma proteins. Importantly, calix[6]arene abolished signal transduction of Mer and AXL tyrosine kinase receptors, both of which are usually overexpressed in pancreatic cancer. Accordingly, inhibition of PI3K and mTOR was also observed, and these proteins are positively modulated by Mer and AXL. Despite decreasing the phosphorylation of AKT at Thr308, calix[6]arene caused an increase in phosphorylation at Ser473. These findings in conjunction with increased BiP and IRE1-α provide a molecular basis explaining the capacity of calix[6]arene to trigger endoplasmic reticulum stress and autophagic cell death. Our findings highlight calix[6]arene as a potential candidate for overcoming pancreatic cancer aggressiveness. Importantly, we provide evidence that calix[6]arene affects a broad array of key targets that are usually dysfunctional in pancreatic cancer, a highly desirable characteristic for chemotherapeutics.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Calixarenes/pharmacology , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Pancreatic Neoplasms/pathology , Phenols/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Calixarenes/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chloroquine/pharmacology , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/ultrastructure , Phenols/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
15.
Gene ; 527(1): 89-94, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23769970

ABSTRACT

Heat shock transcription factor 1 (HSF1), which has been identified as an endogenous cardioprotective factor, possesses potent anti-inflammatory effects. However, the underlying mechanisms have not been fully understood yet. In this study, we investigated the effects of HSF1-regulated RelA, a subunit of NFκB on cardiomyocyte death. Cultured cardiomyocytes were transfected with HSF1 plasmid before the treatment of TNFα. Cell death ratio was determined by cell staining. Additionally, the expression of RelA in the cytoplasm and cytonucleus as well as its subcellular location was detected, and the expression of heat shock proteins (HSP70 and HSP90) in the cardiomyocytes was also examined. Not only did TNFα remarkably enhanced cardiac cell death, but also elevated the expressions of intracellular RelA and elicited its translocation. Overexpression of HSF1 effectively attenuated cell death induced by TNFα. Although HSF1 didn't significantly inhibit the intracellular activation of RelA induced by TNFα at an early stage, HSF1 decreased the levels of RelA and the translocation of RelA in the cytoplasm and cell nucleus at late stage. Besides, the expression of HSP70 and HSP90 was significantly increased when HSF1 was overexpressed. These results suggested that HSF1 attenuated cardiomyocyte death via inhibiting activation of RelA as well as preventing its translocation from the cytoplasm to the cytonucleus, which was partially associated with HSP70 and HSP90 up-regulated by HSF1 overexpression.


Subject(s)
Cell Death , DNA-Binding Proteins/physiology , Myocytes, Cardiac/physiology , Transcription Factor RelA/metabolism , Transcription Factors/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Nucleus/metabolism , Cells, Cultured , Gene Expression , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Primary Cell Culture , Protein Transport , Rats , Rats, Sprague-Dawley , Signal Transduction
16.
Cell Signal ; 25(10): 2060-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770287

ABSTRACT

Ras family small GTPases serve as binary molecular switches to regulate a broad array of cellular signaling cascades, playing essential roles in a vast range of normal physiological processes, with dysregulation of numerous Ras-superfamily G-protein-dependent regulatory cascades underlying the development of human disease. However, the physiological function for many "orphan" Ras-related GTPases remain poorly characterized, including members of the Rit subfamily GTPases. Rit is the founding member of a novel branch of the Ras subfamily, sharing close homology with the neuronally expressed Rin and Drosophila Ric GTPases. Here, we highlight recent studies using transgenic and knockout animal models which have begun to elucidate the physiological roles for the Rit subfamily, including emerging roles in the regulation of neuronal morphology and cellular survival signaling, and discuss new genetic data implicating Rit and Rin signaling in disorders such as cancer, Parkinson's disease, autism, and schizophrenia.


Subject(s)
Cell Differentiation/genetics , Cell Survival/genetics , Neurons/metabolism , ras Proteins/genetics , Animals , Drosophila/genetics , Humans , MAP Kinase Signaling System/genetics , Mice , Monomeric GTP-Binding Proteins , Signal Transduction , ras Proteins/metabolism
17.
Autoimmun Rev ; 12(11): 1070-5, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23685277

ABSTRACT

Most human autoimmune diseases have a relapsing-remitting or a chronic progressive course, while animal models are usually acute and monophasic. In our experimental animal model the disease can be either monophasic or remitting, depending on the autoantigen used for induction, and it appears to lie in the effector phenotype of the elicited T helper cell response. Since both, monophasic and relapsing courses of disease are induced by immunization as well as by adoptive transfer of peptide-specific, CD4(+) T cells, we were able to directly compare the transcriptomes of pathogenic T cell lines by gene array analysis and qPCR as well as protein expression. Upregulated genes were only determined in T cells inducing relapsing uveitis and belong to certain pathways of antigen presentation, activation, inflammation, migration and survival, comprising WNT, Hedgehog, MAP-kinase and JAK/STAT-pathways. These pathways are partially interacting with each other, and the central molecule upregulated in T cells causing relapsing disease was found to be IFN-γ. Here the course of the autoimmune diseases strictly depends on the characteristics of the autoreactive T cells, which are already determined at their early stage of antigen-specific activation. Our rat models of experimental autoimmune uveitis could help elucidating the immune mechanisms behind relapsing autoimmunity in order to develop better therapeutic strategies.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/pathology , Animals , Antigen Presentation/immunology , Autoantigens/adverse effects , Autoantigens/immunology , Autoimmune Diseases/immunology , Disease Models, Animal , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Rats, Inbred Lew , Recurrence , Uveitis/etiology , Uveitis/immunology , Uveitis/pathology
18.
J Biomed Res ; 24(2): 132-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-23554623

ABSTRACT

OBJECTIVE: To explore the role that ceramide plays in the activation of mitogen-activated protein kinases (MAPKs) during cerebral ischemia and reperfusion. METHODS: Rats were subjected to ischemia by the four-vessel occlusion (4-VO) method. The sphingomyelinase inhibitor TPCK was administered to the CA1 subregion of the rat hippocampus before inducing ischemia. Western blot was used to examine the activity of extracellular-signal regulated kinase (ERK) and c-Jun N-terminal protein kinase (JNK) using antibodies against ERK, JNK and diphosphorylated ERK and JNK. RESULTS: At 1h reperfusion post-ischemia, JNK reached its peak activity while ERK was undergoing a sharp inactivation (P < 0.05). The level of diphosphorylated JNK was significantly reduced but the sharp inactivation of ERK was visibly reversed (P < 0.05) by the sphingomyelinase inhibitor. CONCLUSION: The ceramide signaling pathway is up-regulated through sphingomyelin hydrolysis in brain ischemia, promoting JNK activation and suppressing ERK activation, culminating in the ischemic lesion.

19.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-556416

ABSTRACT

Aim To investigate the expression of phosphorylated JNK1/JNK2 and the protection of 11,12-EET in ischemic and reperfusion rat heart.Method The expression of JNK1/JNK2 was detected with western blot method and the changing of heart function during ischemia/reperfusion process was observed in different groups. Results The cardiac function (+dp/dt_(max)%,-dp/dt_(max)% and LVDP)of reperfusion periods(30 min) apparently decreased in ischemia/reperfusion (I/R) group contrasted with Sham group, short ischemia(SI)+I/R group and EET+I/R group,and the expression of phosphorylated JNK1/JNK2 increased in I/R group contrasted with nromal group,Sham group and EET+I/R group.Conclusion The myocardial protection of 11,12-EET ( 6.24?10~(-8) mol?L~(-1)) is able to inhibit the expression of phosphorylated JNK1/JNK2.

SELECTION OF CITATIONS
SEARCH DETAIL
...