Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharmacol ; 885: 173419, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32750370

ABSTRACT

Rosmarinic acid (RA) is a polyphenolic compound with various pharmacological properties, including, anti-inflammatory, immunomodulatory, and neuroprotective, as well as having antioxidant and anticancer activities. This study evaluated the effects and mechanisms of RA in two racially different triple-negative breast cancer (TNBC) cell lines. Results obtained show that RA significantly caused cytotoxic and antiproliferative effects in both cell lines in a dose- and time-dependent manner. Remarkably, RA induced cell cycle arrest-related apoptosis and altered the expression of many apoptosis-involved genes differently. In MDA-MB-231 cells, RA arrested the cells in the G0/G1 phase. In contrast, the data suggest that RA causes S-phase arrest in MDA-MB-468 cells, leading to a 2-fold increase in the apoptotic effect compared to MDA-MB-231 cells. Further, in MDA-MB-231 cells, RA significantly upregulated the mRNA expression of three genes: harakiri (HRK), tumor necrosis factor receptor superfamily 25 (TNFRSF25), and BCL-2 interacting protein 3 (BNIP3). In contrast, in the MDA-MB-468 cell line, the compound induced a significant transcription activation in three genes, including TNF, growth arrest and DNA damage-inducible 45 alpha (GADD45A), and BNIP3. Furthermore, RA repressed the expression of TNF receptor superfamily 11B (TNFRSF11B) in MDA-MB-231 cells in comparison to the ligand TNF superfamily member 10 (TNFSF10) and baculoviral IAP repeat-containing 5 (BIRC5) in MDA-MB-468 cells. In conclusion, the data suggest that the polyphenol RA may have a potential role in TNBC therapies, particularly in MDA-MB-468 cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cinnamates/pharmacology , Depsides/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Apoptosis Regulatory Proteins/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Proteins/antagonists & inhibitors , Osteoprotegerin/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 25/antagonists & inhibitors , Rosmarinic Acid
2.
J Neurochem ; 138(6): 821-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27393374

ABSTRACT

Cocaine is a commonly abused central nervous system stimulant that enhances dopamine (DA) neurotransmission through its ability to block dopamine transporters (DATs). Recent evidence suggests there may be an interaction between DATs and D2/D3 autoreceptors that modulates cocaine's effects. The purpose of this study was to explore how D2/D3 autoreceptors modulate the ability of cocaine to inhibit DA uptake through DATs on pre-synaptic DA terminals. Using fast-scan cyclic voltammetry in brain slices containing the nucleus accumbens core from male and female C57BL/6J mice, we first sought to examine the effects of global autoreceptor blockade using the non-selective D2/D3 autoreceptor antagonist, raclopride. We found that the ability of cocaine to inhibit DA uptake was increased by raclopride and that this effect was consistent across sexes. Furthermore, using D2 (L-741,626) or D3 (SB-277011-A) autoreceptor selective antagonists, we discovered that blockade of D3, but not D2, autoreceptors was responsible for the increased cocaine potency. Alterations in cocaine potency were attributable to alterations in uptake inhibition, rather than cocaine effects on vesicular DA release, suggesting that these results may be a product of a functional D3/DAT interaction apart from the canonical inhibitory actions of D3 autoreceptors on DA release. In addition, application of D2 (sumanirole) and D3 (PD 128907) autoreceptor-specific agonists had inverse effects, whereby D2 autoreceptor activation decreased cocaine potency and D3 autoreceptor activation had no effect. Together, these data show that DA autoreceptors dynamically regulate cocaine potency at the DAT, which is important for understanding cocaine's rewarding and addictive properties. We propose a model whereby presynaptic dopamine autoreceptors dynamically modulate cocaine potency through two separate mechanisms. We demonstrate that D2 agonists decrease cocaine potency, whereas D3 antagonists increase cocaine potency, likely through an allosteric mechanism outside of their canonical actions on dopamine release. These findings give important and novel insight into the contribution of D2/D3 autoreceptors to dopamine transporter function.


Subject(s)
Autoreceptors/antagonists & inhibitors , Cocaine/pharmacology , Dopamine Antagonists/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacology , Receptors, Tumor Necrosis Factor, Member 25/antagonists & inhibitors , Animals , Dopamine Plasma Membrane Transport Proteins/drug effects , Drug Synergism , Female , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Nitriles/pharmacology , Piperidines/pharmacology , Raclopride/pharmacology , Receptors, Dopamine D2/drug effects , Sex Characteristics , Tetrahydroisoquinolines/pharmacology
3.
Mol Med Rep ; 11(2): 797-804, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25370568

ABSTRACT

Death receptor 3 (DR3) belongs to the tumor necrosis factor (TNF) receptor superfamily, primarily found in lymphoid tissues. Reports have determined that DR3 may also be distributed in numerous types of tumors. Therefore, it is thought that DR3 may have an important role in the process of tumorigenesis. The aim of the present study was to observe the effect of silencing DR3 expression on hepatocarcinoma cell growth, apoptosis and invasion in order to elucidate the role of DR3 in tumor development. The hepatocarcinoma cell lines (HepG2, Huh7, SMMC7721 and Bel­7402) and normal human liver cells (HL­7702) were transfected with three stealth RNA interference (RNAi) sequences that target the DR3 gene. Reverse transcription quantitative polymerase chain reaction was used to detect the expression levels of DR3 in hepatocarcinoma cell lines and normal liver HL­7702 cells. MTT assay and flow cytometry (FCM) were used to determine the rates of cell proliferation and apoptosis, respectively. Following silencing of the DR3 gene, western blot analysis was used to determine the protein expression of P53, Fas, Caspase8, nuclear factor kappa­light­chain­enhancer of activated B cells (NF­κB) and Caspase3. DR3 messenger RNA (mRNA) expression in hepatocarcinoma cell lines was significantly increased compared with that in the normal liver cell line. Three targeted DR3 gene small interfering RNAs significantly inhibited DR3 gene expression in Bel­7402 cells at the nucleic acid level. AF02670.1_stealth_883 and cocktail demonstrated the most efficient inhibition of DR3 gene expression at 48 and 72 h following transfection, with mRNA inhibition rates of 89.46 and 92.75%, and 90.53 and 94.25% (P<0.01), respectively. Cell viability was significantly reduced by AF02670.1_stealth_883 and RNAi cocktail at 24, 48 and 72 h following transfection. The inhibition rates of cell proliferation were 50.76 and 61.76% (P<0.05) at 72 h following transfection. FCM revealed that AF02670.1_stealth_883 and RNAi cocktail also induced apoptosis in Bel­7402 cells at 72 h following transfection. Reduction of NF­κB and P53 levels was observed (P<0.05) in Bel­7402 cells following DR3 silencing, whereas levels of Fas, Caspase3 and Caspase8 were markedly elevated (P<0.05). DR3 expression levels in hepatocellular carcinoma cells were significantly higher than those in normal cells. DR3 silencing effectively inhibited proliferation and invasion of hepatocellular carcinoma cells in vitro. However, silencing of the DR3 gene affect levels of apoptosis antigen­3 ligand in cells, therefore indicating that it may be involved with other pathways that regulate apoptosis in HCCs. In conclusion, the results of the present study indicated that DR3 may be a promising therapeutic target molecule for further study of hepatocellular carcinoma gene therapy.


Subject(s)
Receptors, Tumor Necrosis Factor, Member 25/metabolism , Apoptosis , Carcinoma, Hepatocellular/pathology , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line , Cell Movement , Cell Proliferation , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver Neoplasms/pathology , NF-kappa B/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Tumor Necrosis Factor, Member 25/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 25/genetics , Tumor Suppressor Protein p53/metabolism , fas Receptor/metabolism
4.
Int J Cancer ; 125(10): 2465-73, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19670328

ABSTRACT

Death receptors of the tumor necrosis factor (TNF) receptor super family have been implicated in constitutive activation of nuclear factor-kappa B (NF-kappaB) in pancreatic cancer (PaC) cells. In this study, we demonstrate that fisetin, a natural flavonoid, induces apoptosis and inhibits invasion of chemoresistant PaC AsPC-1 cells through suppression of DR3-mediated NF-kappaB activation. Fisetin treatment resulted in dose-dependent inhibition of PaC cell growth and cell proliferation with concomitant induction of apoptosis. A cDNA array analysis revealed that fisetin modulates expression of more than 20 genes at transcription level with maximum decrease observed in DR3 expression and a parallel increase observed in the expression levels of IkappaBalpha, an NF-kappaB inhibitor. Down-regulation of DR3 in PaC cells was found to down regulate activated pNF-kappaB/p65, pIkBalpha/beta kinases (pIKK's), MMP9 and XIAP that mostly impart chemoresistance in PaC. Immunoblotting and EMSA analysis showed a marked decrease in pNF-kappaB and NF-kappaB DNA binding activity, respectively, with modest decrease in NF-kappaB promoter activity and significant decrease in MMP9 promoter activity with fisetin treatment. Importantly, consistent with these findings, we further found that transient down-regulation of DR3 by RNA interference significantly augmented fisetin induced changes in cell proliferation, cell invasion and apoptosis paralleled with decrease in pNF-kappaB, pIKKalpha/beta, MMP9, XIAP and NF-kappaB DNA binding activity. Blocking of DR3 receptor with an extra cellular domain blocking antibody demonstrated similar effects. These data provide evidence that fisetin could provide a biological rationale for treatment of pancreatic cancer or as an adjuvant with conventional therapeutic regimens.


Subject(s)
Drug Resistance, Neoplasm , Flavonoids/pharmacology , NF-kappa B/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Electrophoretic Mobility Shift Assay , Flavonols , Gene Expression Profiling , Humans , I-kappa B Proteins/metabolism , Matrix Metalloproteinase 9/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , RNA, Small Interfering/pharmacology , Receptors, Tumor Necrosis Factor, Member 25/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction , X-Linked Inhibitor of Apoptosis Protein/metabolism
5.
Cancer Invest ; 27(2): 163-70, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19235588

ABSTRACT

Lupeol (Lup-20(29)-en-3H-ol), a novel dietary triterpene, was found in fruits, vegetables, and several medicinal plants. Here, we investigated its growth-inhibitory effect and associated mechanisms in hepatocellular carcinoma SMMC7721 cells. Lupeol treatment resulted in significant inhibition of cell viability in a dose-dependent manner and caused apoptotic death of this cell line with activation of caspase3 expression. Caspase8 inhibitor pretreatment was found to partially block the apoptosis induced by Lupeol. Moreover, Lupeol specifically caused a significant decrease in the expression of Death receptor 3 (DR3) mRNA and protein and a significant elevated expression of FADD mRNA whereas Fas mRNA and protein expression was not detectable. Further more, knockdown of DR3 by small interfering RNA inhibited the growth and induced apoptosis of hepatocellular carcinoma cell. These results suggested that Lupeol treatment induced growth inhibition and apoptosis in SMMC7721 cells, the mechanism is due to down-regulation of DR3 expression. We demonstrated that Lupeol appears to be a promising chemopreventive agent for treating hepatocellular carcinoma, and DR3 may be an important target for liver cancer therapy.


Subject(s)
Apoptosis/drug effects , Liver Neoplasms/drug therapy , Receptors, Tumor Necrosis Factor, Member 25/antagonists & inhibitors , Triterpenes/pharmacology , Caspase 3/analysis , Caspase 8/physiology , Cell Line, Tumor , Cell Survival/drug effects , Fas-Associated Death Domain Protein/genetics , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Pentacyclic Triterpenes , RNA, Messenger/analysis , Receptors, Tumor Necrosis Factor, Member 25/analysis , Receptors, Tumor Necrosis Factor, Member 25/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...