Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Brain Res ; 1629: 351-60, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26474912

ABSTRACT

In the study, we investigated the effect of histamine microinjected into cerebellar fastigial nucleus (FN) on stress gastric mucosal damage (SGMD), and its mechanisms in rats. The model of SGMD was established by restraining and water (21±1°C)-immersion for 3h. The gastric mucosal damage index (GMDI) indicated the severity of gastric mucosal damage. Histamine or receptor antagonist was microinjected into the FN. The decussation of superior cerebellar peduncle (DSCP) and the lateral hypothalamic area (LHA) were destroyed, respectively. The pathological changes of gastric mucosa were evaluated using biological signal acquisition system, Laser-Doppler flowmeter, and western blotting. We found that the microinjection of histamine (0.05, 0.5, and 5µg) into FN significantly attenuated the SGMD, in a dose-dependent manner, whereas, the microinjection of histamine H2 receptor antagonist, ranitidine, and glutamic acid decarboxylase antagonist, 3-mercaptopropionic acid (3-MPA) exacerbated the SGMD. The protective effect of histamine on SGMD was abolished by electrical lesion of DSCP or chemical ablation of LHA. The microinjection of histamine decreased the discharge frequency of the greater splanchnic nerve, and the gastric mucosal blood flow was increased. In addition, the cellular proliferation was enhanced, but the cellular apoptosis was reduced in the gastric mucosa. Also the pro-apoptosis protein, Bax, and caspase-3 were down-regulated, and the anti-apoptosis protein, Bcl-2 was up-regulated following microinjection of histamine. In conclusion, the FN participated in the regulation of SGMD after histamine microinjected into FN, and cerebellar-hypothalamic circuits (include: DSCP, LHA) contribute to the process, which may provide a new therapeutic strategy for SGMD.


Subject(s)
Cerebellum/metabolism , Gastric Mucosa/metabolism , Histamine/administration & dosage , Neuroprotective Agents/administration & dosage , Stress, Psychological/metabolism , Stress, Psychological/prevention & control , Animals , Cerebellum/drug effects , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Male , Mice , Microinjections , Rats , Rats, Sprague-Dawley
2.
Acta Pharmacol Sin ; 35(2): 185-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24362327

ABSTRACT

AIM: To investigate the effects of glutamate microinjection into hypothalamic paraventricular nucleus (PVN) on ulcerative colitis (UC) in rats and to explore the relevant mechanisms. METHODS: 2,4,6-Trinitrobenzenesulfonic acid (100 mg/kg in 50% ethanol) was instilled into the colon of adult male SD rats to induce UC. A colonic damage score (CDS) was used to indicate the severity of the colonic mucosal damage. The pathological changes in the colonic mucosa were evaluated using immunohistochemistry, Western blotting, biochemical analyses or ELISA. Ten minutes before UC induction, drugs were microinjected into the relevant nuclei in rat brain to produce chemical stimulation or chemical lesion. RESULTS: Microinjection of glutamate (3, 6 and 12 µg) into the PVN dose-dependently decreased the CDS in UC rats. This protective effect was eliminated after kainic acid (0.3 µg) was microinjected into PVN or into the nucleus tractus solitarius (NTS) that caused chemical lesion of these nuclei. This protective effect was also prevented when the AVP-V1 receptor antagonist DPVDAV (200 ng) was microinjected into the NTS. The discharge frequency of the vagus was markedly decreased following microinjection of glutamate into the PVN. Microinjection of glutamate into the PVN in UC rats significantly increased the cell proliferation and anti-oxidant levels, and decreased the apoptosis and Bax and caspase 3 expression levels and reduced the pro-inflammatory factors in the colonic mucosa. CONCLUSION: The activation of hypothalamic PVN exerts protective effects against UC, which is mediated by the NTS and vagus. The effects may be achieved via anti-oxidative, anti-apoptotic, and anti-inflammatory factors.


Subject(s)
Colitis, Ulcerative/drug therapy , Glutamic Acid/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Male , Microinjections/methods , Rats , Rats, Sprague-Dawley
3.
Int Immunopharmacol ; 16(2): 332-40, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23639256

ABSTRACT

Lupeol, a triterpene, was reported to possess beneficial effects as a therapeutic and preventive agent for a range of disorders. Many studies have confirmed that lupeol possesses strong activities such as antioxidative, antiinflammatory, antiarthritic, antimutagenic, and antimalarial, both in vitro and in vivo, and at its effective therapeutic doses exhibit no toxicity to normal cells and tissues. Lupeol was observed to inhibit the proliferation of gastric tumour cells in a dose-dependent manner, as assessed by MTT assay, and induce the proliferation of NK cells, as assessed by flow cytometry and Western blotting. The killing effect of NK cells on gastric tumour cells was assessed by LDH. Our experiment demonstrated that lupeol at appropriate concentrations could promote the proliferation of NK cells, inhibit the proliferation of gastric cancer cell lines BGC823, N87 and HGC27, and increase the killing effect of NK cells on gastric cancer cells. We speculated that lupeol might increase the expression of PFP, IFN-γ, and CD107a via the activation of PI3K/Akt and Wnt/ß-catenin signalling pathways. Lupeol could serve as a potential agent against gastric cancer; however, further in-depth in vivo studies are still required.


Subject(s)
Antineoplastic Agents/pharmacology , Killer Cells, Natural/drug effects , Pentacyclic Triterpenes/pharmacology , Stomach Neoplasms/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Cytokines/immunology , Humans , Interferon-gamma/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/physiology , Lysosomal-Associated Membrane Protein 1/immunology , Pore Forming Cytotoxic Proteins/immunology , Proto-Oncogene Proteins c-akt/immunology , Stomach Neoplasms/drug therapy , beta Catenin/immunology
4.
Int Immunopharmacol ; 15(1): 6-14, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23174508

ABSTRACT

OBJECTIVE: To explore the effect and mechanism of Phloretin on human γδ T cells killing colon cancer SW-1116 cells. METHODS: γδ T cells were amplified in vitro from human peripheral blood mononuclear cells through isopentenyl pyrophosphate method (IPP). After cocultured different concentrations of Phloretin with γδ T cells or SW-1116 cells for 48h respectively, MTT assay was used to test the growth curve of these two cells; Flow cytometry to test the expression of Granzyme B (GraB), perforin (PFP), CD107a and IFN-γ of γδ T cells; Lactate dehydrogenase (LDH) release assay to test the cytotoxic activity of the γδ T cells on SW-1116 cells; and Western blot to test the Wnt3a expression of the γδ T cells. RESULTS: After cultured with IPP for ten days, the percentage of γδ T cells increased from 3.31±3.00% to 78.40±10.30%. Compared to the control group, when the concentration of Phloretin increased from 2.35µg/ml to 18.75µg/ml, it could significantly proliferate the γδ T cell growth (P<0.05) and inhibit the growth of SW-1116 cells in dose-response, and the expression of GraB, PFP, CD107a and Wnt3a significantly increased (P<0.05). Significant positive relationships were observed among CD107a and PFP, GraB, cytotoxicity (P<0.05). The percentage of IFN-γ producing γδ T cells treated with Phloretin was significantly higher than control group. CONCLUSION: Phloretin can enhance the killing effect of γδ T cells on SW-1116 cells; the mechanism may be that Phloretin could proliferate the γδ T cell growth, increase the expression of PFP and GraB, activate the Wnt signaling pathway, and produce higher level of IFN-γ. Indeed CD107a expression probably correlates quite well with antitumor activity.


Subject(s)
Colonic Neoplasms/immunology , Immunologic Factors/pharmacology , Phloretin/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocytes/drug effects , Adult , Cell Line, Tumor , Cell Survival/drug effects , Granzymes/immunology , Humans , Interferon-gamma/immunology , L-Lactate Dehydrogenase/immunology , Lysosomal-Associated Membrane Protein 1/immunology , Perforin/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Wnt3A Protein/immunology , Young Adult
5.
Acta Pharmacol Sin ; 34(2): 205-13, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23247592

ABSTRACT

AIM: To investigate the effects of microinjection of the GABA(A) receptor agonist muscimol into cerebellar fastigial nucleus (FN) on stress-induced gastric mucosal damage and the underlying mechanism in rats. METHODS: Stress-induced gastric mucosal damage was induced in adult male SD rats by restraining and immersing them in cold water for 3 h. GABA(A) receptor agonist or antagonist was microinjected into the lateral FN. The decussation of superior cerebellar peduncle (DSCP) was electrically destroyed and the lateral hypothalamic area (LHA) was chemically ablated by microinjection of kainic acid. The pathological changes in the gastric mucosa were evaluated using TUNEL staining, immunohistochemistry staining and Western blotting. RESULTS: Microinjection of muscimol (1.25, 2.5, and 5.0 µg) into FN significantly exacerbated the stress-induced gastric mucosal damage in a dose-dependent manner, whereas microinjection of GABA(A) receptor antagonist bicuculline attenuated the damage. The intensifying effect of muscimol on gastric mucosal damage was abolished by electrical lesion of DSCP or chemical ablation of LHA performed 3 d before microinjection of muscimol. Microinjection of muscimol markedly increased the discharge frequency of the greater splanchnic nerve, significantly increased the gastric acid volume and acidity, and further reduced the gastric mucosal blood flow. In the gastric mucosa, further reduced proliferation cells, enhanced apoptosis, and decreased anti-oxidant levels were observed following microinjection of muscimol. CONCLUSION: Cerebellar FN participates in the regulation of stress-induced gastric mucosal damage, and cerebello-hypothalamic circuits contribute to the process.


Subject(s)
Cerebellar Nuclei/drug effects , GABA-A Receptor Agonists/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Muscimol/pharmacology , Stress, Physiological , Animals , Apoptosis/drug effects , Bicuculline/administration & dosage , Bicuculline/pharmacology , Cerebellar Nuclei/pathology , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Antagonists/administration & dosage , GABA-A Receptor Antagonists/pharmacology , Gastric Mucosa/blood supply , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/physiology , Male , Microinjections , Muscimol/administration & dosage , Rats , Rats, Sprague-Dawley
6.
World J Gastroenterol ; 18(38): 5377-88, 2012 Oct 14.
Article in English | MEDLINE | ID: mdl-23082054

ABSTRACT

AIM: To investigate the protective effect and mechanisms of ghrelin postconditioning against hypoxia/reoxygenation (H/R)-induced injury in human gastric epithelial cells. METHODS: The model of H/R injury was established in gastric epithelial cell line (GES-1) human gastric epithelial cells. Cells were divided into seven groups: normal control group (N); H/R postconditioning group; DMSO postconditioning group (DM); ghrelin postconditioning group (GH); D-Lys3-GHRP-6 + ghrelin postconditioning group (D + GH); capsazepine + ghrelin postconditioning group (C + GH); and LY294002 + ghrelin postconditioning group (L + GH). 3-(4,5-dimethylthazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay was used to detect GES-1 cell viability. Hoechst 33258 fluorochrome staining and flow cytometry were conducted to determine apoptosis of GES-1 cells. Spectrophotometry was performed to determine release of lactate dehydrogenate (LDH). Protein expression of Bcl-2, Bax, Akt, and glycogen synthase kinase (GSK)-3ß was determined by western blotting. Expression of vanilloid receptor subtype 1 (VR1), Akt and GSK-3ß was observed by immunocytochemistry. RESULTS: Compared with the H/R group, cell viability of the GH group was significantly increased in a dose-dependent manner (55.9% ± 10.0% vs 69.6% ± 9.6%, 71.9% ± 17.4%, and 76.3% ± 13.3%). Compared with the H/R group, the percentage of apoptotic cells in the GH group significantly decreased (12.38% ± 1.51% vs 6.88% ± 0.87%). Compared with the GH group, the percentage of apoptotic cells in the D + GH group, C + GH group and L + GH groups significantly increased (11.70% ± 0.88%, 11.93% ± 0.96%, 10.20% ± 1.05% vs 6.88% ± 0.87%). There were no significant differences in the percentage of apoptotic cells between the H/R and DM groups (12.38% ± 1.51% vs13.00% ± 1.13%). There was a significant decrease in LDH release following ghrelin postconditioning compared with the H/R group (561.58 ± 64.01 U/L vs 1062.45 ± 105.29 U/L). There was a significant increase in LDH release in the D + GH, C + GH and L + GH groups compared with the GH group (816.89 ± 94.87 U/L, 870.95 ± 64.06 U/L, 838.62 ± 118.45 U/L vs 561.58 ± 64.01 U/L). There were no significant differences in LDH release between the H/R and DM groups (1062.45 ± 105.29 U/L vs 1017.65 ± 68.90 U/L). Compared with the H/R group, expression of Bcl-2 and Akt increased in the GH group, whereas expression of Bax and GSK-3ß decreased. Compared with the GH group, expression of Bcl-2 decreased and Bax increased in the D + GH, C + GH and L + GH groups, and Akt decreased and GSK-3ß increased in the L + GH group. The H/R group also upregulated expression of VR1 and GSK-3ß and downregulated Akt. The number of VR1-positive and Akt-positive cells in the GH group significantly increased, whereas the number of GSK-3ß-positive cells significantly decreased. These effects of ghrelin were reversed by capsazepine and LY294002. CONCLUSION: Ghrelin postconditioning protected against H/R-induced injury in human gastric epithelial cells, which indicated that this protection might be associated with GHS-R, VR1 and the PI3K/Akt signaling pathway.


Subject(s)
Epithelial Cells/drug effects , Gastric Mucosa/blood supply , Ghrelin/therapeutic use , Ischemic Postconditioning/methods , Protective Agents/therapeutic use , Reperfusion Injury/prevention & control , Apoptosis/drug effects , Biomarkers/metabolism , Blotting, Western , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Flow Cytometry , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Ghrelin/pharmacology , Humans , Immunohistochemistry , Protective Agents/pharmacology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
7.
Neurosci Lett ; 525(1): 39-43, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22842393

ABSTRACT

We investigated the protective effects of chemical stimulation of cerebellar interpositus nucleus (IN) on gastric ischemia-reperfusion injury (GI-RI) and its possible regulatory mechanisms in rats. Gastric mucosal damage index (GMDI) indicated the severity of gastric mucosal injuries. Transferase dUTP nick end labeling (TUNEL) staining and proliferating cell nuclear antigen (PCNA) were performed to assess gastric mucosal cell apoptosis and proliferation. Microinjection of glutamate into IN markedly attenuated GI-RI. Either chemical lesion of IN or electrical ablation of the decussation of superior cerebellar peduncle (DSCP) obviously aggravated GI-RI. The protective effects of IN were reversed with the pretreatments of microinjection of 3-mercaptopropionic acid into IN or Bicuculline into lateral hypothalamic area (LHA), individually. The discharge frequency and intensity of greater splanchnic nerve (GSN) decreased and gastric mucosal blood flow increased after chemical stimulation of IN. The apoptosis of positive cells of gastric mucosa was decreased by chemical stimulation of IN, whereas proliferation increased. The gastric juice volume, acidity, and total acid output were all decreased after the chemical stimulation of IN. These results indicated that IN participates in regulation of GI-RI and is a specific area in central nervous system for exerting protective effects on GI-RI. DSCP, LHA and GSN may involve in this process. Apoptosis and proliferation may mediate this protective process in rats too.


Subject(s)
Cerebellum/drug effects , Glutamic Acid/pharmacology , Hypothalamic Area, Lateral/physiopathology , Reperfusion Injury/pathology , Stomach/blood supply , Animals , Apoptosis , Cell Proliferation , Cerebellum/physiopathology , Gastric Juice/chemistry , Gastric Juice/metabolism , Gastric Mucosa/pathology , Gastric Mucosa/physiopathology , Hydrogen-Ion Concentration , Male , Microinjections , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Stomach/pathology , Stomach/physiopathology , gamma-Aminobutyric Acid/physiology
8.
Neurosci Lett ; 509(2): 125-9, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-22240102

ABSTRACT

Cerebellum, primarily believed as a subcortical somatic motor center, is increasingly considered to be implicated in visceral activities. However, little is known about its regulation on gastrointestinal organs. In this research, we investigated the aggravated effect of microinjection of gamma-aminobutyric acid receptor subtype B (GABA(B)R) agonist, Baclofen into cerebellar fastigial nucleus (FN) on stress gastric mucosal damage (SGMD) and its possible regulatory mechanism. The gastric mucosal damage index was chosen to indicate the severity of gastric mucosal injure. Immunohistochemistry and transferase-mediated dUTP-biotin nick-endlabeling (TUNEL) methods were used to detect the variations of lateral hypothalamic area (LHA) and gastric mucosa. It had been demonstrated that FN participates in regulation of SGMD via its GABA(B)R and GABA neural pathway, which passes through the decussation of superior cerebellar peduncle and projects to the GABA receptors in LHA. Meanwhile, celiac sympathetic nerve involves in this process via mediating neural discharge, which results in the decrease of gastric mucosal blood flow. Additionally, apoptosis, proliferation and oxidation in gastric mucosa, and gastric acid contribute in the mechanism. It could be expected that these results might suggest insights to the cerebellar and hypothalamic function, and the treatment of gastrointestinal diseases.


Subject(s)
Baclofen/pharmacology , Cerebellar Nuclei/drug effects , Cerebellar Nuclei/physiology , Gastric Mucosa/innervation , Gastric Mucosa/pathology , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/physiology , Stress, Physiological/physiology , Animals , Apoptosis/drug effects , Baclofen/administration & dosage , Cell Proliferation/drug effects , GABA-B Receptor Agonists/administration & dosage , GABA-B Receptor Agonists/pharmacology , Ganglia, Sympathetic/physiology , Ganglia, Sympathetic/surgery , Ganglionectomy , Gastric Acid/metabolism , Gastric Mucosa/drug effects , Gastrointestinal Diseases/physiopathology , Microinjections , Neural Pathways/drug effects , Rats , Receptors, GABA/metabolism , gamma-Aminobutyric Acid/metabolism
9.
Anal Bioanal Chem ; 386(6): 1905-11, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17047947

ABSTRACT

Two novel N-(2-mercapto-1,3,4-thiadiazol-5-yl)-N'-(4-substituted-arylacetyl) urea compounds have been synthesized, characterized by NMR and MS, and used as self-assembly reagents to form self-assembled monolayers (SAMs) on Pt electrodes. The modified electrodes were characterized by electrochemical methods. The electrochemical behavior of p-benzenediol at the SAMs electrodes was investigated. It was found that the electrochemical response to p-benzenediol is controlled by diffusion and can be electrocatalyzed to obtain more symmetrical redox peaks and higher voltammetric current response at the SAMs electrodes, with a peak separation of 80 mV. For p-benzenediol the process at the SAMs electrodes is quasi-reversible with a rate constant of 0.6742 s-1. The SAMs electrodes have been used to determine p-benzenediol by differential pulse voltammetry. The peak current was linear for concentrations of p-benzenediol in the range 1x10(-7)-5x10(-4) mol L-1 and the detection limit was 4.0x10(-8) mol L-1. The SAMs electrodes were used to determine p-benzenediol in real photographic developer and in a synthetic waste water sample; the standard addition recovery was in the range 96.6-100.4%.


Subject(s)
Electrochemistry , Hydroquinones/chemistry , Platinum/chemistry , Thiadiazoles/chemistry , Urea/analogs & derivatives , Electrodes , Hydrogen-Ion Concentration , Molecular Structure , Reproducibility of Results , Solutions , Urea/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...