Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 2448, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38291092

ABSTRACT

In China, there has been a persistent upward trend in the incidence and mortality rates of colorectal cancer (CRC), with CRC ranking second in incidence and fifth in mortality among all malignant tumors. Although circular RNAs (circRNAs) have been implicated in the progression of various cancers, their specific role in CRC progression remains largely unexplored. The objective of this study was to elucidate the role and underlying mechanisms of circXRN2 in CRC. Differential expression of circXRN2 was identified through whole transcriptome sequencing. The expression levels of circXRN2 and miR-149-5p were quantified in CRC tissues, corresponding adjacent normal tissues, and CRC cell lines using quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The stability of circXRN2 was confirmed through RNase R and actinomycin D experiments. The binding interaction between circXRN2 and miR-149-5p was validated through RNA pull-down, RNA immunoprecipitation, and dual-luciferase assays. The biological functions of circXRN2 were assessed through a battery of in vitro experiments, including the CCK-8 assay, EdU assay, scratch assay, Transwell assay, and flow cytometry assay. Additionally, in vivo experiments involving a tumor transplantation model and a liver-lung metastasis model were conducted. The influence of circXRN2 on the expression of epithelial-mesenchymal transition (EMT)-related genes was determined via Western blotting analysis. In CRC tissues and cells, there was an upregulation in the expression levels of both circXRN2 and ENC1, while miR-149-5p exhibited a downregulation in its expression. The overexpression of circXRN2 was found to enhance tumor proliferation and metastasis, as evidenced by results from both in vitro and in vivo experiments. Functionally, circXRN2 exerted its antitumor effect by suppressing cell proliferation, migration, and invasion while also promoting apoptosis. Mechanistically, the dysregulated expression of circXRN2 had an impact on the expression of proteins within the EMT signaling pathway. Our results demonstrated that circXRN2 promoted the proliferation and metastasis of CRC cells through the miR-149-5p/ENC1/EMT axis, suggesting that circXRN2 might serve as a potential therapeutic target and novel biomarker in the progression of CRC.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Lung Neoplasms , MicroRNAs , Humans , MicroRNAs/metabolism , Colorectal Neoplasms/pathology , Cell Line , Lung Neoplasms/genetics , Liver Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Trans-Activators/metabolism
2.
PLoS One ; 19(1): e0296508, 2024.
Article in English | MEDLINE | ID: mdl-38180977

ABSTRACT

OBJECTIVE: To evaluate the efficacy and safety of Ginkgolide Meglumine Injection (GMI) combined with Butylphthalide in the treatment of Acute Ischemic Stroke (AIS), and provide reference for rational clinical medication. METHODS: PubMed, Embase, Web of science, CNKI, Wanfang, VIP and other databases were searched for published studies on the treatment of AIS with GMI combined with Butylphthalide in both Chinese and English. The search period was from the establishment of the database to July 2023. The included studies that met the inclusion criteria were analyzed using RevMan 5.3 software for Meta-analysis. RESULTS: A total of 25 studies involving 2362 patients (experimental group = 1182, control group = 1180) were included. The results of meta-analysis showed that the overall effective rate of the experimental group was significantly higher than that of the control group [RR = 1.21, 95% CI (1.16, 1.26), P< 0.00001]. In addition, compared with the control group, the experimental group showed significant improvement in NIHSS score [SMD = -1.59, % CI (-2.00-1.18), P< 0.00001] and ADL score [SMD = 2.12, 95% CI (1.52, -2.72), P<0.00001], significant decrease in CRP [SMD = -2.24, 95% CI (-3.31, -1.18), P<0.0001] and TNF-α [SMD = -2.74, 95% CI (-4.45, -1.03), P< 0.005] levels, and improvement in plasma viscosity [SMD = -0.86, 95% CI (-1.07, -0.66), P< 0.00001]. However, the influence on homocysteine level remains inconclusive. Furthermore, there was no significant difference in the incidence of adverse reactions between the two groups [SMD = 0.95, 95% CI (0.71, 1.28), P> 0.05]. CONCLUSION: GMI combined with Butylphthalide shows good clinical application effects and good safety in the treatment of AIS. However, more large-sample, multicenter, randomized controlled are needed to confirm these findings.


Subject(s)
Benzofurans , Ischemic Stroke , Humans , Benzofurans/adverse effects , Ginkgolides/adverse effects , Meglumine , Multicenter Studies as Topic
3.
Medicine (Baltimore) ; 102(41): e33199, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37832102

ABSTRACT

BACKGROUND: To evaluate the effectiveness and safety of Xingnaojing combined with naloxone in the treatment of carbon monoxide poisoning. METHODS: By retrieving the literatures published in the databases of PubMed, Cochrane Library, Web of Science, Embase, Wanfang Database, Weipu Database, and China National Knowledge Infrastructure from January 2010 to September 2021, the data of randomized controlled trials (RCTs) of Xingnaojing combined with naloxone in the treatment of carbon monoxide poisoning were extracted. The methodological quality of the included RCTs was evaluated by using the tools of bias risk evaluation of Cochrane Collaboration, and the data were statistically analyzed by using RevMan 5.3 software. RESULTS: A total of 20 literatures were included, involving in 771 cases treated by Xingnaojing combined with naloxone and 761 cases in the control group. The effective rate of the experimental group is higher than that of the control group [risk ratio (RR) = 1.20, 95% confidence interval (CI) (1.14, 1.26)]. The average awake time (STD mean difference = -2.08, 95% CI [-2.60, -1.56]), physical recovery time (STD mean difference = -2.94, 95% CI [-3.59, -2.28]), delayed encephalopathy (RR = 0.44, 95% CI [0.31, 0.62]), and adverse reactions (RR = 0.23, 95% CI [0.10, 0.54]) was lower than that of the control group. CONCLUSION: Xingnaojing combined with naloxone in the treatment of carbon monoxide poisoning is significantly superior to naloxone, but it still needs to be further verified by high-quality large samples of RCTs.


Subject(s)
Carbon Monoxide Poisoning , Humans , Carbon Monoxide Poisoning/drug therapy , Drugs, Chinese Herbal/therapeutic use , Naloxone/therapeutic use
4.
World J Clin Cases ; 11(5): 1049-1057, 2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36874432

ABSTRACT

BACKGROUND: Hepatic colon carcinoma invading the duodenum is not common in clinical practice. Surgical treatment of colonic hepatic cancer that invades the duodenum is difficult, and the surgical risk is high. AIM: To discuss the efficacy and safety of duodenum-jejunum Roux-en-Y anastomosis for the treatment of hepatic colon carcinoma invading the duodenum. METHODS: From 2016 to 2020, 11 patients from Panzhihua Central Hospital diagnosed with hepatic colon carcinoma were enrolled in this study. Clinical and therapeutic effects and prognostic indicators were retrospectively analyzed to determine the efficacy and safety of our surgical procedures. All patients underwent radical resection of right colon cancer combined with duodenum-jejunum Roux-en-Y anastomosis. RESULTS: The median tumor size was 65 mm (r50-90). Major complications (Clavien-DindoI-II) occurred in 3 patients (27.3%); the average length of hospital stay was 18.09 ± 4.21 d; and only 1 patient (9.1%) was readmitted during the 1st mo after the surgery. The 30-d mortality rate was 0%. After a median follow-up of 41 m (r7-58), the disease-free survival at 1, 2, and 3 years was 90.9%, 90.9% and 75.8%, respectively; the overall survival at 1, 2, and 3 years was 90.9%. CONCLUSION: In selected patients, radical resection of right colon cancer combined with duodenum-jejunum Roux-en-Y anastomosis is clinically effective, and the complications are manageable. The surgical procedure also has an acceptable morbidity rate and mid-term survival.

5.
World J Clin Cases ; 10(31): 11523-11528, 2022 Nov 06.
Article in English | MEDLINE | ID: mdl-36387825

ABSTRACT

BACKGROUND: It is very rare to suffer from colorectal adenocarcinoma and abdominal tuberculosis simultaneously. Even in a country such as China, where tuberculosis is still endemic, its diagnosis and treatment are challenging. This article describes in detail a case of rectal cancer complicated by abdominal tuberculosis and its pathological features. CASE SUMMARY: We outline the case of a 71-year-old female who was admitted with intermittent blood in the stool over the past year. The patient was diagnosed with low rectal cancer and received neoadjuvant therapy. The patient then returned to the hospital for surgery, but diffusely distributed nodules were found during laparoscopic exploration. The diagnosis of rectal cancer with extensive metastasis was considered during the operation. There was no opportunity for radical surgery. Thus, nodules were taken for pathological examination, and the abdomen was closed. The histopathological diagnosis was tuberculous granuloma, and the patient was treated with standardized anti-tuberculosis drugs in a specialized hospital. Later, the patient again came to our hospital and underwent abdominoperineal resection. She was discharged 10 d after the operation in good clinical condition. CONCLUSION: We aim to emphasize the importance of preoperative and postoperative pathological examination in diagnosis and treatment.

6.
PLoS One ; 17(10): e0275735, 2022.
Article in English | MEDLINE | ID: mdl-36215266

ABSTRACT

OBJECTIVE: To conduct a systematic review of the efficacy and safety of Shenyankangfu tablets in combination with losartan potassium in the treatment of chronic glomerulonephritis. METHOD: We searched PubMed, Embase, Cochrane Library, CNKI, WanFang Data, and WeiPu for comparative studies on Shenyankangfu tablets in combination with losartan potassium in the treatment of chronic glomerulonephritis. The search period runs from the establishment of the database until September 2021. Data extraction and quality evaluation were carried out on the documents that met the inclusion criteria, and a meta-analysis of the included literature was conducted using the RevMan5.3 software. RESULTS: A total of 17 randomized controlled trials that met the inclusion criteria were included, with a total sample size of 1680 patients (841 patients in the study group and 839 in the control group). The effective rate was significantly higher in the study group than in the control group [RR = 1.22, 95% CI (1.16, 1.27), P < 0.00001]. In addition, 24-hour urine protein levels [SMD = -1.11, 95% CI (-1.40, -0.83), P < 0.00001], urine NAG enzyme [SMD = -0.99, 95% CI (-1.27, -0.72), P < 0.00001], leukotactin-1 [SMD = -2.43, 95% CI (-3.50, -1.35), P < 0.00001], and the incidence of adverse reactions [RR = 0.43, 95% CI (0.28, 0.66), P < 0.00001] were lower in the study group when compared to the control group. CONCLUSION: It is safer to treat chronic glomerulonephritis with Shyenyankangfu tablets in combination with losartan potassium. At the same time, it alleviates disease-related symptoms, reduces the influence of cytokine levels, and has fewer adverse reactions, making it more conducive to disease recovery. However, additional multi-center, randomized, control trials with large sample sizes must be conducted to confirm the findings.


Subject(s)
Drugs, Chinese Herbal , Glomerulonephritis , Cytokines , Drug Combinations , Drugs, Chinese Herbal/adverse effects , Glomerulonephritis/drug therapy , Humans , Losartan/therapeutic use , Randomized Controlled Trials as Topic , Tablets
7.
Pharmacol Rep ; 70(4): 705-711, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29933208

ABSTRACT

BACKGROUND: This study aimed to study the osteo-preservative effects of captopril, an inhibitor on angiotensin-converting enzyme (ACE), on bone mass, micro-architecture and histomorphology as well as the modulation of captopril on skeletal renin-angiotensin system (RAS) and regulators for bone metabolism in mice with bilateral orchidectomy. METHODS: The orchidectomized (ORX) mice were orally administered with vehicle or captopril at low dose (10mg/kg) and high dose (50mg/kg) for six weeks. The distal femoral end, the proximal tibial head and the lumbar vertebra (LV) were stained by hematoxylin and eosin, Safranin O/Fast Green and masson-trichrome. Micro-computed tomography was performed to measure bone mineral density (BMD). RESULTS: Treatment with captopril increased trabecular bone area at distal metaphysis of femur, proximal metaphysis of tibia and LV-4, moreover, high dose of captopril significantly elevated trabecular BMD of LV-2 and LV-5. The mRNA expressions of renin receptor, angiotensinogen, carbonic anhydrase II, matrix metalloproteinase-9, and tumor necrosis factor-alpha were significantly decreased in tibia of ORX mice following treatment with captopril. The administration with captopril enhanced the ratio of OPG/RANKL mRNA expression, the mRNA expression of transforming growth factor-beta and the protein expression of bradykinin receptor-1. CONCLUSIONS: The inhibition on ACE by captopril exerts beneficial effects on trabecular bone of ORX mice. The therapeutic efficacy may be attributed to the regulation of captopril on local RAS and cytokines in bone.


Subject(s)
Bone Density/drug effects , Cancellous Bone/drug effects , Captopril/pharmacology , Femur/metabolism , Lumbar Vertebrae/metabolism , Tibia/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensinogen/biosynthesis , Animals , Carbonic Anhydrase II/biosynthesis , Dose-Response Relationship, Drug , Male , Matrix Metalloproteinase 9/biosynthesis , Mice , Orchiectomy , Osteoprotegerin/biosynthesis , Proton-Translocating ATPases/biosynthesis , RANK Ligand/biosynthesis , Receptor, Bradykinin B1/biosynthesis , Receptors, Cell Surface/biosynthesis , Renin-Angiotensin System/drug effects , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
8.
Eur J Pharmacol ; 819: 281-288, 2018 Jan 15.
Article in English | MEDLINE | ID: mdl-29242120

ABSTRACT

Salidroside, an active constituent from the root of Rhodiola rosea L., has multiple pharmacological effects, such as anti-cancer, anti-inflammatory and anti-oxidative properties, etc. However, its protective effect on bone tissue via regulating calcium homeostasis is yet to be determined. This study was performed to investigate if salidroside could protect against bone injuries induced by estrogen deficiency or hyperglycemia through modulating calcium homeostasis. Ovariectomized (OVX) mice and diabetic mice were treated with salidroside (20mg/kg) for 6 weeks. Safranin O staining and micro-CT were performed on the distal metaphysis of femur. The calcium content in serum, urine and femur was measured, and the mRNA and protein expressions of regulators in kidney were determined by PCR and immunoblotting, respectively. Treatment with salidroside increased bone calcium level and decreased urinary calcium excretion, consequently attenuating the deteriorations of trabecular bone in both OVX mice and diabetic mice. 25-Hydroxyvitamin D-24 hydroxylase expression was down-regulated and vitamin D receptor expression was up-regulated in kidney of both OVX mice and diabetic mice upon to salidroside treatment, which also inhibited the ovariectomy-induced decrease in expression of renal transcellular calcium transporters and the diabetes-induced enhancement in renal calcium-sensing receptor (CaSR) expression. Taken together, salidroside exerted osteoprotective effects by improving calcium homeostasis via regulating vitamin D metabolism and transcellular calcium transporters as well as modulating CaSR expression in kidney.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Femur/drug effects , Glucosides/pharmacology , Ovariectomy/adverse effects , Phenols/pharmacology , Animals , Blood Glucose/metabolism , Cytoprotection/drug effects , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/urine , Estrogens/blood , Fasting/blood , Femur/metabolism , Femur/pathology , Gene Expression Regulation/drug effects , Kidney/drug effects , Kidney/metabolism , Mice , Mice, Inbred C57BL , Vitamin D/metabolism
9.
Sci Rep ; 6: 36238, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27824091

ABSTRACT

A novel series of hybrids (7a-l, 8a-l) from ß-carboline and salicylic acid (SA) were designed and synthesized, and their in vitro biological activities were evaluated. Most of the hybrids displayed potent antiproliferative activity against five cancer cell lines in vitro, showing potencies superior to 5-FU and harmine. In particular, compound 8h selectively inhibited proliferation of liver cancer SMMC-7721 cells but not normal liver LO2 cells, and displayed greater inhibitory selectivity than intermediate 5h and SA. 8h also induced cancer cell apoptosis in an Annexin V-FITC/propidium iodide flow cytometry assay, and triggered the mitochondrial/caspase apoptosis by decreasing mitochondrial membrane potential which was associated with up-regulation of Bax, down-regulation of Bcl-2 and activation levels of the caspase cascade in a concentration-dependent manner. Our findings suggest that the ß-carboline/SA hybrids may hold greater promise as therapeutic agents for the intervention of human cancers.


Subject(s)
Antineoplastic Agents/chemical synthesis , Carbolines/chemistry , Neoplasms/metabolism , Salicylic Acid/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Design , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Harmine/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Neoplasms/drug therapy , bcl-2-Associated X Protein/metabolism
10.
Int J Neuropsychopharmacol ; 18(8)2015 Jan 24.
Article in English | MEDLINE | ID: mdl-25618406

ABSTRACT

BACKGROUND: Current antidepressants are clinically effective only after several weeks of administration. Tetramethylpyrazine (TMP) is an identified component of Ligusticum wallichii with neuroprotective effects. Here, we investigated the antidepressant effects of TMP in mice models of depression. METHODS: Antidepressant effects of TMP were first detected in the forced swim test (FST) and tail suspension test (TST), and further assessed in the chronic social defeat stress (CSDS) model. Changes in the brain-derived neurotrophic factor (BDNF) signaling pathway and in hippocampal neurogenesis after CSDS and TMP treatment were then investigated. A tryptophan hydroxylase inhibitor and BDNF signaling inhibitors were also used to determine the mechanisms of TMP. RESULTS: TMP exhibited potent antidepressant effects in the FST and TST without affecting locomotor activity. TMP also prevented the CSDS-induced symptoms. Moreover, TMP completely restored the CSDS-induced decrease of BDNF signaling pathway and hippocampal neurogenesis. Furthermore, a blockade of the BDNF signaling pathway prevented the antidepressant effects of TMP, while TMP produced no influence on the monoaminergic system. CONCLUSIONS: In conclusion, these data provide the first evidence that TMP has antidepressant effects, and this was mediated by promoting the BDNF signaling pathway.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder/drug therapy , Pyrazines/pharmacology , Animals , Depressive Disorder/physiopathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Neurogenesis/drug effects , Neurogenesis/physiology , Neuropsychological Tests , Signal Transduction/drug effects , Tryptophan Hydroxylase/antagonists & inhibitors , Tryptophan Hydroxylase/metabolism
11.
Clin Exp Pharmacol Physiol ; 41(12): 976-85, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224648

ABSTRACT

Our previous studies showed that protein phosphatase 1γ (PP1γ) exacerbates cardiomyocyte apoptosis through promotion of Ca(2+)/calmodulin-dependent protein kinase δ (CaMKIIδ) splicing. Here we determine the role of PP1γ in abdominal aorta constriction-induced hypertrophy and remodelling in rat hearts. Systolic blood pressure and echocardiographic measurements were used to evaluate the model of cardiac hypertrophy. Sirius red staining and invasive haemodynamic/cardiac index measurements were used to evaluate the effects of PP1γ or inhibitor 1 of PP1 transfection. Western blot, reverse transcription polymerase chain reaction and co-immunoprecipitation were applied to investigate the molecular mechanisms. Transfection of PP1γ increased the value of the heart mass index, left ventricular mass index and cardiac fibrosis, and simultaneously decreased the value of maximal left ventricular pressure increase and decline rate, ejection fraction, fractional shortening, and left ventricular end-diastolic pressure, as well as left ventricular systolic pressure. Transfection of inhibitor 1 of PP1, however, showed opposite effects on the aforementioned indexes. Overexpression of PP1γ potentiated CaMKIIδC production and decreased CaMKIIδB production in the hypertrophic heart. In contrast, inhibition of PP1γ re-balanced the CaMKIIδ splicing. Furthermore, CaMKII activity was found to be augmented or attenuated by PP1γ overexpression or inhibition, respectively. Further mechanistic studies showed that abdominal aorta constriction stress specifically increased the association of alternative splicing factor with PP1γ, but not with PP1ß. Overexpression of PP1γ, but not inhibitor 1 of PP1, further potentiated this association. These results suggest that PP1γ alters the cardiac hypertrophy and remodelling likely through promotion of the alternative splicing factor-mediated splicing of CaMKIIδ.


Subject(s)
Alternative Splicing/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Calmodulin/metabolism , Heart Failure/metabolism , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Animals , Apoptosis/physiology , Cardiomegaly/metabolism , Heart Failure/physiopathology , Heart Ventricles/metabolism , Male , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
12.
Can J Physiol Pharmacol ; 90(12): 1611-22, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23210440

ABSTRACT

Most cardiac diseases are associated with fibrosis. Calcineurin (CaN) is regulated by Ca(2+)/calmodulin (CaM). The CaN-NFAT (nuclear factor of activated T cell) pathway is involved in the process of cardiac diseases, such as cardiac hypertrophy, but its effect on myocardial fibrosis remains unclear. The present study investigates whether the CaN-NFAT pathway is involved in cardiac fibroblast (CF) proliferation induced by electrical field stimulation (EFS), which recently became a popular treatment for heart failure and cardiac tissue engineering. CF proliferation was evaluated by a cell survival assay (MTT) and cell counts. Myocardial fibrosis was assessed by collagen I and collagen III protein expression. Green fluorescent protein (GFP)-tagged NFAT was used to detect NFAT nuclear translocation. CF proliferation, myocardial fibrosis, CaN activity, and NFAT nuclear translocation were enhanced by EFS. More importantly, these effects were abolished by CaN inhibitors, dominant negative CaN (DN-CaN), and CaN gene silenced with siRNA. Furthermore, buffering intracellular Ca(2+) with BAPTA-AM and blocking Ca(2+) influx with nifedipine suppressed EFS-induced increase in intracellular Ca(2+) and CF proliferation. These results suggested that the CaN-NFAT pathway mediates CF proliferation, and that the CaN-NFAT pathway might be a possible therapeutic target for EFS-induced myocardial fibrosis and cardiac tissue engineering.


Subject(s)
Calcineurin/metabolism , Fibroblasts/physiology , Myocytes, Cardiac/physiology , NFATC Transcription Factors/metabolism , Animals , Calcium/metabolism , Cell Growth Processes/physiology , Cell Nucleus/metabolism , Cell Survival/physiology , Cells, Cultured , Collagen Type I/metabolism , Collagen Type III/metabolism , Electric Stimulation/methods , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Fibroblasts/cytology , Fibroblasts/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Transport , Rats , Rats, Sprague-Dawley , Signal Transduction
13.
Clin Exp Pharmacol Physiol ; 39(5): 429-37, 2012 May.
Article in English | MEDLINE | ID: mdl-22340635

ABSTRACT

Cardiac fibroblasts (CF) have direct and potent effects on myocardial remodelling by proliferating, differentiating and secreting extracellular matrix proteins. Prolonged activation of CF leads to cardiac fibrosis and reduces myocardial contractile function. In previous studies we showed that 2,3,4',5-tetrahydroxystilbene-2-O-ß-d-glucoside (TSG) exerts cardiac protection, but the mechanism involved remains unclear. The aim of the present study was to evaluate the effects of TSG on angiotensin (Ang) II-induced CF proliferation and to explore the underlying intracellular mechanisms. Angiotensin II (100 nmol/L)-induced proliferation of rat neonatal fibroblasts was significantly inhibited by TSG (3-100 µmol/L), as evidenced by investigations of cell numbers and 5-bromodeoxyuridine (BrdU) incorporation. In addition, 30 µmol/L TSG suppressed AngII-induced expression of nuclear antigen, matrix metalloproteinase (MMP)-2 and MMP-9. Moreover, TSG attenuated AngII-induced activation of mitogen-activated protein kinase kinase (MEK) and extracellular signal-regulated kinase (ERK) 1/2. Angiotensin II (100 nmol/L)-induced generation of reactive oxygen species (ROS) was reduced by 30 µmol/L TSG, as was H(2) O(2) -induced activation of ERK1/2. However, the MEK inhibitor 50 µmol/L PD98059 did not reduce ROS generation, although it did inhibit cell proliferation. There was a significant correlation between the inhibition of ERK1/2 activation and suppression of cell proliferation by TSG. However, there were no additive effects on either the inhibition of ERK1/2 or the suppression of cell proliferation following treatment of cells with both PD98059 and TSG. In conclusion, the results of the present study suggest that TSG inhibits ERK1/2 activation, likely via buffering of ROS, and consequently suppresses cell proliferation.


Subject(s)
Angiotensin II/metabolism , Cell Proliferation/drug effects , Fibroblasts/drug effects , Glucosides/pharmacology , Growth Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Stilbenes/pharmacology , Angiotensin II/physiology , Animals , Animals, Newborn , Cells, Cultured , Fibroblasts/enzymology , MAP Kinase Signaling System/physiology , Myocytes, Cardiac/enzymology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/pharmacology
14.
Phytother Res ; 26(7): 1068-74, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22213189

ABSTRACT

The proliferation of vascular smooth muscle cells (VSMCs) induced by injury to the intima of arteries is an important etiologic factor in vascular proliferative disorders such as atherosclerosis and restenosis. 2,3,4',5-Tetrahydroxystilbene-2-O-ß-D-glucoside (TSG), an active component extracted from Polygonum multiflorum, has been found to have an antiatherosclerotic effect. The aim of this study was to investigate the effects of TSG on platelet derived growth factor (PDGF)-BB induced VSMCs proliferation and to explore the possible mechanisms of such effects. Pretreatment of VSMCs with TSG significantly inhibited PDGF-BB-induced cell proliferation in a concentration-dependent but not time-dependent manner. In addition, flow cytometry analysis of the DNA content revealed blocking of the PDGF-BB-inducible cell cycle progression by TSG. On the contrary, an inhibitory effect of TSG on VSMCs proliferation and expression of cell cycle regulators were markedly attenuated by addition of an nitric oxide (NO) synthase inhibitor, a soluble guanylate cyclase inhibitor and a cyclic GMP (cGMP)-dependent protein kinase (PKG) inhibitor: N(G)-nitro-L-arginine methyl ester (L-NAME), 1H-[1,2,4] oxadiazolo [4,3-α] quinoxalin-1-one (ODQ) and KT5823, respectively. It was also demonstrated that TSG enhanced NO and cGMP formation through up-regulating endothelial NO synthase expression in VSMCs. The findings indicate that TSG inhibited VSMCs proliferation induced by PDGF-BB may involve the NO/cGMP/PKG signal pathway.


Subject(s)
Cell Proliferation/drug effects , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Glucosides/pharmacology , Myocytes, Smooth Muscle/drug effects , Nitric Oxide/metabolism , Stilbenes/pharmacology , Animals , Becaplermin , Carbazoles , Cell Cycle/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Male , Muscle, Smooth, Vascular/cytology , NG-Nitroarginine Methyl Ester , Oxadiazoles , Polygonum/chemistry , Proto-Oncogene Proteins c-sis , Quinoxalines , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
15.
Clin Exp Pharmacol Physiol ; 38(5): 307-13, 2011 May.
Article in English | MEDLINE | ID: mdl-21348889

ABSTRACT

1. 2,3,4',5-Tetrahydroxystilbene-2-O-ß-d-glucoside (TSG) has been shown to have an anti-atherosclerotic effect. Vascular smooth muscle cell (VSMC) proliferation contributes to the pathobiology of atherosclerosis. The aim of the present study was to investigate the effects of TSG on platelet-derived growth factor (PDGF)-BB-induced VSMC proliferation and to explore the molecular mechanisms underlying the effects. 2. Cultured rat VSMC were pretreated with TSG (l-50 µmol/L) for 1 h, followed by exposure to PDGF-BB (10 ng/mL) for 24 h, after which cell proliferation and cell cycle stages were examined. The expression of protein cell cycle regulators, including retinoblastoma (Rb), cyclin D1/E, cyclin-dependent kinase (CDK) 2/4, CDK inhibitors p21 and p27 and proliferative cell nuclear antigen (PCNA), was examined. Activation of extracellular signal-regulated kinase (ERK) 1/2 was evaluated to elucidate the possible upstream mechanism by which TSG affects cell cycle regulators. 3. The results showed that TSG dose-dependently inhibited PDGF-BB-induced VSMC proliferation, possibly by blocking the progression of the cell cycle from the G(1) to S phase. In addition, TSG significantly inhibited PDGF-BB-induced phosphorylation of Rb and the expression of cyclin D1, CDK4, cyclin E, CDK2 and PCNA. In addition, TSG suppressed PDGF-BB-induced downregulation of p27 and upregulation of p21, as well as PDGF-BB-induced activation of ERK1/2. 4. Together, the findings of the present study provide the first evidence that TSG can inhibit PDGF-BB-stimulated VSMC proliferation via cell cycle arrest in association with modulation of the expression of cell cycle regulators, which may be mediated, at least in part, by suppression of ERK1/2 activation.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , Glucosides/pharmacology , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/pharmacology , Stilbenes/pharmacology , Animals , Becaplermin , Cardiovascular Agents/pharmacology , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Male , Models, Biological , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Platelet-Derived Growth Factor/antagonists & inhibitors , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...